101
|
Weidenbusch M, Song S, Iwakura T, Shi C, Rodler S, Kobold S, Mulay SR, Honarpisheh MM, Anders H. IL-22 sustains epithelial integrity in progressive kidney remodeling and fibrosis. Physiol Rep 2018; 6:e13817. [PMID: 30156011 PMCID: PMC6113136 DOI: 10.14814/phy2.13817] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
IL-22, a member of the IL-10 cytokine family, accelerates tubule regeneration upon acute kidney injury, hence we speculated on a protective role also in chronic kidney disease. We quantified intrarenal IL-22 expression after unilateral ureteral (UUO) in wild-type mice and performed UUO in IL-22 knock-out animals. Obstruction phenotypic differences between IL22+/+ and IL22-/- mice were assessed by histology, immunohistochemistry, immunofluorescence as well as western blotting and reverse-transcriptase quantitative PCR ex vivo. Additionally, we performed in vitro experiments using both murine and human tubular cells to characterize IL-22 effects in epithelial healing. We found increasing IL-22 positivity in infiltrating immune cells over time upon UUO in wild-type mice. UUO in IL22-/- mice caused more tubular cell injury as defined by TUNEL positive cells and loss of tetragonolobus lectin staining. Instead, tubular dilation, loss of CD31+ perivascular capillaries, and interstitial fibrosis were independent of the Il22 genotype as assessed by standard histology, immunostaining, and mRNA expression profiling. In vitro experiments showed that recombinant human IL-22 significantly enhanced human tubular epithelial cell proliferation and wound closure upon mechanical injury, and electric cell-substrate impedance sensing studies revealed that recombinant IL-22 sustained tubular epithelial barrier function upon injury. In contrast, IL-22 had no such direct effects on human fibroblasts. Together, in progressive kidney remodeling upon UUO, infiltrating immune cells secrete IL-22, which augments tubular epithelial integrity and epithelial barrier function, but does not affect vascular rarefaction or fibrogenesis. We conclude that IL-22 could represent a molecular target to specifically modulate tubular atrophy.
Collapse
Affiliation(s)
- Marc Weidenbusch
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Shangqing Song
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
- Department of UrologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Takamasa Iwakura
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Chongxu Shi
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Severin Rodler
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Sebastian Kobold
- Center for Integrated Protein Science Munich (CIPSM) and Abteilung für Klinische PharmakologieMedizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Shrikant R. Mulay
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Mohsen M. Honarpisheh
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| | - Hans‐Joachim Anders
- Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenLudwig Maximilians University of MunichMunichGermany
| |
Collapse
|
102
|
Barthelemy A, Sencio V, Soulard D, Deruyter L, Faveeuw C, Le Goffic R, Trottein F. Interleukin-22 Immunotherapy during Severe Influenza Enhances Lung Tissue Integrity and Reduces Secondary Bacterial Systemic Invasion. Infect Immun 2018; 86:e00706-17. [PMID: 29661933 PMCID: PMC6013680 DOI: 10.1128/iai.00706-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/03/2018] [Indexed: 12/15/2022] Open
Abstract
Severe bacterial (pneumococcal) infections are commonly associated with influenza and are significant contributors to the excess morbidity and mortality of influenza. Disruption of lung tissue integrity during influenza participates in bacterial pulmonary colonization and dissemination out of the lungs. Interleukin-22 (IL-22) has gained considerable interest in anti-inflammatory and anti-infection immunotherapy over the last decade. In the current study, we investigated the effect of exogenous IL-22 delivery on the outcome of pneumococcal superinfection postinfluenza. Our data show that exogenous treatment of influenza virus-infected mice with recombinant IL-22 reduces bacterial dissemination out of the lungs but is without effect on pulmonary bacterial burden. Reduced systemic bacterial dissemination was linked to reinforced pulmonary barrier functions, as revealed by total protein measurement in the bronchoalveolar fluids, intratracheal fluorescein isothiocyanate-dextran tracking, and histological approaches. We describe an IL-22-specific gene signature in the lung tissue of influenza A virus (IAV)-infected (and naive) mice that might explain the observed effects. Indeed, exogenous IL-22 modulates the gene expression profile in a way that suggests reinforcement of tissue integrity. Our results open the way to alternative approaches for limiting postinfluenza bacterial superinfection, particularly, systemic bacterial invasion.
Collapse
Affiliation(s)
- Adeline Barthelemy
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Valentin Sencio
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Daphnée Soulard
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Lucie Deruyter
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Ronan Le Goffic
- Molecular Virology and Immunology, Institut National de la Recherche Agronomique, Université Paris-Saclay, Jouy-en-Josas, France
| | - François Trottein
- Universitaire de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
103
|
Crane MJ, Lee KM, FitzGerald ES, Jamieson AM. Surviving Deadly Lung Infections: Innate Host Tolerance Mechanisms in the Pulmonary System. Front Immunol 2018; 9:1421. [PMID: 29988424 PMCID: PMC6024012 DOI: 10.3389/fimmu.2018.01421] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
Much research on infectious diseases focuses on clearing the pathogen through the use of antimicrobial drugs, the immune response, or a combination of both. Rapid clearance of pathogens allows for a quick return to a healthy state and increased survival. Pathogen-targeted approaches to combating infection have inherent limitations, including their pathogen-specific nature, the potential for antimicrobial resistance, and poor vaccine efficacy, among others. Another way to survive an infection is to tolerate the alterations to homeostasis that occur during a disease state through a process called host tolerance or resilience, which is independent from pathogen burden. Alterations in homeostasis during infection are numerous and include tissue damage, increased inflammation, metabolic changes, temperature changes, and changes in respiration. Given its importance and sensitivity, the lung is a good system for understanding host tolerance to infectious disease. Pneumonia is the leading cause of death for children under five worldwide. One reason for this is because when the pulmonary system is altered dramatically it greatly impacts the overall health and survival of a patient. Targeting host pathways involved in maintenance of pulmonary host tolerance during infection could provide an alternative therapeutic avenue that may be broadly applicable across a variety of pathologies. In this review, we will summarize recent findings on tolerance to host lung infection. We will focus on the involvement of innate immune responses in tolerance and how an initial viral lung infection may alter tolerance mechanisms in leukocytic, epithelial, and endothelial compartments to a subsequent bacterial infection. By understanding tolerance mechanisms in the lung we can better address treatment options for deadly pulmonary infections.
Collapse
Affiliation(s)
| | | | | | - Amanda M. Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| |
Collapse
|
104
|
Elsegeiny W, Zheng M, Eddens T, Gallo RL, Dai G, Trevejo-Nunez G, Castillo P, Kracinovsky K, Cleveland H, Horne W, Franks J, Pociask D, Pilarski M, Alcorn JF, Chen K, Kolls JK. Murine models of Pneumocystis infection recapitulate human primary immune disorders. JCI Insight 2018; 3:91894. [PMID: 29925696 PMCID: PMC6124425 DOI: 10.1172/jci.insight.91894] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/17/2018] [Indexed: 12/19/2022] Open
Abstract
Despite the discovery of key pattern recognition receptors and CD4+ T cell subsets in laboratory mice, there is ongoing discussion of the value of murine models to reflect human disease. Pneumocystis is an AIDS-defining illness, in which risk of infection is inversely correlated with peripheral CD4+ T cell counts. Due to medical advances in the control of HIV, the current epidemiology of Pneumocystis infection is predominantly due to primary human immunodeficiencies and immunosuppressive therapies. To this end, we found that every human genetic immunodeficiency associated with Pneumocystis infection that has been tested in mice recapitulated susceptibility. For example, humans with a loss-of-function IL21R mutation are severely immunocompromised. We found that IL-21R, in addition to CD4+ T cell intrinsic STAT3 signaling, were required for generating protective antifungal class-switched antibody responses, as well as effector T cell-mediated protection. Furthermore, CD4+ T cell intrinsic IL-21R/STAT3 signaling was required for CD4+ T cell effector responses, including IL-22 production. Recombinant IL-22 administration to Il21r-/- mice induced the expression of a fungicidal peptide, cathelicidin antimicrobial peptide, which showed in vitro fungicidal activity. In conclusion, SPF laboratory mice faithfully replicate many aspects of human primary immunodeficiency and provide useful tools to understand the generation and nature of effector CD4+ T cell immunity.
Collapse
Affiliation(s)
- Waleed Elsegeiny
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mingquan Zheng
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Taylor Eddens
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Guixiang Dai
- Department of Medicine, Tulane School of Medicine, New Orleans, Louisiana, USA
| | - Giraldina Trevejo-Nunez
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Patricia Castillo
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kara Kracinovsky
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hillary Cleveland
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research and
| | - Jonathan Franks
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Derek Pociask
- Department of Medicine, Tulane School of Medicine, New Orleans, Louisiana, USA
| | - Mark Pilarski
- Richard King Mellon Foundation Institute for Pediatric Research and
| | - John F. Alcorn
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jay K. Kolls
- Richard King Mellon Foundation Institute for Pediatric Research and
- Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, Tulane School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
105
|
Jensen TJ, Shui JE, Finck CM. The effect of meconium exposure on the expression and differentiation of amniotic fluid mesenchymal stem cells. J Neonatal Perinatal Med 2018; 10:313-323. [PMID: 28854517 PMCID: PMC5676976 DOI: 10.3233/npm-16141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The goal of this study was to determine if exposure to meconium would alter the phenotype of amniotic fluid mesenchymal stem cells (AF-MSCs) and the ability of these cells to be differentiated into distal airway type cells. METHODS: Meconium was collected, lyophilized and resuspended in PBS at 3 different concentrations (high, medium, and low). AF-MSCs were cultured in the presence of this meconium suspension for 8 hours and then analyzed for changes in gene expression. Additionally, AF-MSCs exposed to meconium were differentiated for 14 days using modified small airway growth medium (mSAGM) and gene expression was determined. As a spontaneous differentiation control, meconium exposed AF-MSCs were cultured in amniotic fluid stem cell medium (AF medium). RESULTS: After 8 hours of exposure in culture, AF-MSCs had increased expression of distal airway genes aquaporin 5 (AQP5) and surfactant protein c (SPC) when cultured in AF medium containing meconium. These gene expression levels were similar to that of AF-MSCs that were differentiated in mSAGM for 14 days. Furthermore, there was an up regulation of pluripotency genes NANOG and OCT4 in response to low meconium concentration for 8 hours. Following 14 days of culture in mSAGM, there was an upregulation of TTF1, SPC and AQP5 expression in the control, as well as in the low and medium meconium exposed groups indicating that these cells were still able to be differentiated. High meconium concentration did, however, appear to influence the level of distal airway gene expression after 14 days in mSAGM. After 14 days in AF medium, there was significant downregulation in pluripotency and mesenchymal markers as well as distal airway gene expression in all groups. CONCLUSION: The phenotype of AF-MSCs is modulated by meconium exposure; however, the cells were still able to differentiate into distal airway gene and protein expression. This result supports the hypothesis that progenitor cells exist in the amniotic fluid and the presence of meconium may affect their initial phenotype. However, these cells were still able to be differentiated to a distal lung phenotype.
Collapse
Affiliation(s)
- T J Jensen
- Department of Pediatrics, UConn Health, Farmington, CT, USA
| | - J E Shui
- Department of Pediatrics, Connecticut Children's Medical Center, Hartford, CT, USA
| | - C M Finck
- Department of Surgery, Connecticut Children's Medical Center, Harford, CT, USA
| |
Collapse
|
106
|
Bloom K, Mohsen AW, Karunanidhi A, El Demellawy D, Reyes-Múgica M, Wang Y, Ghaloul-Gonzalez L, Otsubo C, Tobita K, Muzumdar R, Gong Z, Tas E, Basu S, Chen J, Bennett M, Hoppel C, Vockley J. Investigating the link of ACAD10 deficiency to type 2 diabetes mellitus. J Inherit Metab Dis 2018; 41:49-57. [PMID: 28120165 PMCID: PMC5524623 DOI: 10.1007/s10545-017-0013-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/27/2016] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
Abstract
The Native American Pima population has the highest incidence of insulin resistance (IR) and type 2 diabetes mellitus (T2DM) of any reported population, but the pathophysiologic mechanism is unknown. Genetic studies in Pima Indians have linked acyl-CoA dehydrogenase 10 (ACAD10) gene polymorphisms, among others, to this predisposition. The gene codes for a protein with a C-terminus region that is structurally similar to members of a family of flavoenzymes-the acyl-CoA dehydrogenases (ACADs)-that catalyze α,β-dehydrogenation reactions, including the first step in mitochondrial FAO (FAO), and intermediary reactions in amino acids catabolism. Dysregulation of FAO and an increase in plasma acylcarnitines are recognized as important in the pathophysiology of IR and T2DM. To investigate the deficiency of ACAD10 as a monogenic risk factor for T2DM in human, an Acad-deficient mouse was generated and characterized. The deficient mice exhibit an abnormal glucose tolerance test and elevated insulin levels. Blood acylcarnitine analysis shows an increase in long-chain species in the older mice. Nonspecific variable pattern of elevated short-terminal branch-chain acylcarnitines in a variety of tissues was also observed. Acad10 mice accumulate excess abdominal adipose tissue, develop an early inflammatory liver process, exhibit fasting rhabdomyolysis, and have abnormal skeletal muscle mitochondria. Our results identify Acad10 as a genetic determinant of T2DM in mice and provide a model to further investigate genetic determinants for insulin resistance in humans.
Collapse
MESH Headings
- Abdominal Fat/enzymology
- Abdominal Fat/physiopathology
- Acyl-CoA Dehydrogenase/genetics
- Adiposity
- Animals
- Blood Glucose/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Disease Models, Animal
- Genetic Predisposition to Disease
- Insulin/blood
- Insulin Resistance/genetics
- Lipid Metabolism, Inborn Errors/enzymology
- Lipid Metabolism, Inborn Errors/genetics
- Lipid Metabolism, Inborn Errors/pathology
- Lipid Metabolism, Inborn Errors/physiopathology
- Liver/enzymology
- Liver/pathology
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Muscle/enzymology
- Mitochondria, Muscle/pathology
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/pathology
- Non-alcoholic Fatty Liver Disease/enzymology
- Non-alcoholic Fatty Liver Disease/genetics
- Non-alcoholic Fatty Liver Disease/pathology
- Obesity, Abdominal/enzymology
- Obesity, Abdominal/genetics
- Obesity, Abdominal/physiopathology
- Phenotype
- Rhabdomyolysis/enzymology
- Rhabdomyolysis/genetics
- Rhabdomyolysis/pathology
Collapse
Affiliation(s)
- Kaitlyn Bloom
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Al-Walid Mohsen
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Anuradha Karunanidhi
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Dina El Demellawy
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Miguel Reyes-Múgica
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yudong Wang
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Lina Ghaloul-Gonzalez
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Chikara Otsubo
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Kimi Tobita
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Radhika Muzumdar
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Zhenwei Gong
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Emir Tas
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Shrabani Basu
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Jie Chen
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Michael Bennett
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Charles Hoppel
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jerry Vockley
- Department of Pediatrics, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
107
|
Seki N, Kan-O K, Matsumoto K, Fukuyama S, Hamano S, Tonai K, Ota K, Inoue H, Nakanishi Y. Interleukin-22 attenuates double-stranded RNA-induced upregulation of PD-L1 in airway epithelial cells via a STAT3-dependent mechanism. Biochem Biophys Res Commun 2017; 494:242-248. [PMID: 29032197 DOI: 10.1016/j.bbrc.2017.10.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 01/13/2023]
Abstract
Double-stranded RNA derived from viruses induces host immune responses. PD-L1, also known as B7-H1, is an immune-checkpoint molecule associated with the escape of viruses from host immune systems, which plays a role in the persistence of viral infection, resulting in exacerbations of underlying diseases such as asthma and chronic obstructive pulmonary disease. Interleukin (IL)-22 is produced from various immune cells and has protective properties on mucosal tissue. The binding of IL-22 to IL-22 receptor induces STAT3 activation. We investigated the effect of IL-22 on the expression in airway epithelial cells in vitro and in mouse lungs in vivo after the stimulation with an analog of viral double-stranded RNA, polyinosinic-polycytidylic acid (poly I:C). Stimulation with poly I:C upregulated PD-L1 expression on BEAS-2B cells. This upregulation of PD-L1 was attenuated by IL-22 administration. STAT3 phosphorylation was induced by IL-22 and poly I:C. Treatment of cells with STAT3 siRNA abolished the effect of IL-22 on the poly I:C-induced upregulation of PD-L1. This upregulation of PD-L1 was also attenuated by IL-11, a cytokine inducing STAT3 phosphorylation, in BEAS-2B cells. In mouse lung cells in vivo, IL-22 suppressed poly I:C-induced upregulation of PD-L1. These results suggest that IL-22 attenuates virus-induced upregulation of PD-L1 in airway epithelial cells via a STAT3-dependent mechanism.
Collapse
Affiliation(s)
- Nanae Seki
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Keiko Kan-O
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Satoru Fukuyama
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Saaka Hamano
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Ken Tonai
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Keiichi Ota
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-0075, Japan.
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
108
|
Shabgah AG, Navashenaq JG, Shabgah OG, Mohammadi H, Sahebkar A. Interleukin-22 in human inflammatory diseases and viral infections. Autoimmun Rev 2017; 16:1209-1218. [PMID: 29037907 DOI: 10.1016/j.autrev.2017.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/24/2022]
Abstract
Interleukin-22 (IL22) is one of the members of IL10 family. Elevated levels of this cytokine can be seen in diseases caused by T lymphocytes, such as Psoriasis, Rheumatoid arthritis, interstitial lung diseases. IL22 is produced by different cells in both innate and acquired immunities. Different types of T cells are able to produce IL22, but the major IL22-producing T-cell is the TCD4. TH22 cell is a new line of TCD4 cells, which differentiated from naive T cells in the presence of TNFα and IL6; 50% of peripheral blood IL22 is produced by these cells. IL22 has important functions in host defense at mucosal surfaces as well as in tissue repair. In this review, we assess the current understanding of this cytokine and focus on the possible roles of IL-22 in autoimmune diseases.
Collapse
Affiliation(s)
- Arezoo Gowhari Shabgah
- Immunology Research Center, Avicenna Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Blood Borne Infections Research Center, AcademicCenter for Education, Culture and Research (ACECR), Razavi Khorasan Branch,Mashhad, Iran
| | - Jamshid Gholizadeh Navashenaq
- Immunology Research Center, Avicenna Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Omid Gohari Shabgah
- Parasitology Department, Medical sciencesfaculty, Tarbiat Modares University, Tehran, Iran
| | - Hamed Mohammadi
- ImmunologyResearch Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Sahebkar
- BiotechnologyResearch Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
109
|
Carmo RF, Cavalcanti MSM, Moura P. Role of Interleukin-22 in chronic liver injury. Cytokine 2017; 98:107-114. [PMID: 27816383 DOI: 10.1016/j.cyto.2016.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/05/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is the result of an exacerbated wound-healing response associated with chronic liver injury. Advanced liver fibrosis results in cirrhosis, liver failure, and portal hypertension and frequently requires liver transplantation. The host immune response has an important role driving fibrosis deposition by activating hepatic stellate cells (HSCs). Interleukin-22 (IL-22) is a cytokine that plays a key role in promoting antimicrobial immunity and tissue repair at barrier surfaces. Data from literature suggest that IL-22 has a protective role in the liver by reducing fibrosis in some pathological conditions, however the results are contradictory. This review highlights current knowledge of IL-22' role in chronic liver injury, as well as its therapeutic potential for the treatment of chronic liver injury.
Collapse
Affiliation(s)
- Rodrigo F Carmo
- Colegiado de Farmácia, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, Brazil.
| | - Maria S M Cavalcanti
- Instituto de Ciências Biológicas, Universidade de Pernambuco (UPE), Recife, Brazil
| | - Patrícia Moura
- Instituto de Ciências Biológicas, Universidade de Pernambuco (UPE), Recife, Brazil
| |
Collapse
|
110
|
Chen CJ, Wu GH, Kuo RL, Shih SR. Role of the intestinal microbiota in the immunomodulation of influenza virus infection. Microbes Infect 2017; 19:570-579. [PMID: 28939355 DOI: 10.1016/j.micinf.2017.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/02/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023]
Abstract
Prevention and treatment measures against influenza virus infection remain limited, and alternative host protection strategies are badly needed. In this review, we discuss the regulatory role of intestinal microbiota in influenza infections, and present the latest evidence for strategies seeking to harness gut microbiota for the management of influenza infections.
Collapse
Affiliation(s)
- Chi-Jene Chen
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan; Research Center for Emerging Viruses, China Medical University Hospital, Taichung, Taiwan
| | - Guan-Hong Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Rei-Lin Kuo
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Shin-Ru Shih
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Clinical Virology Laboratory, Department of Laboratory Science, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
111
|
Gurczynski SJ, Moore BB. IL-17 in the lung: the good, the bad, and the ugly. Am J Physiol Lung Cell Mol Physiol 2017; 314:L6-L16. [PMID: 28860146 DOI: 10.1152/ajplung.00344.2017] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The IL-17 family of cytokines has emerged over the last two decades as a pleiotropic group of molecules that function in a wide variety of both beneficial and detrimental (pathological) processes, mainly in mucosal barrier tissue. The beneficial effects of IL-17 expression are especially important in the lung, where exposure to foreign agents is abundant. IL-17A plays an important role in protection from both extracellular bacteria and fungi, as well as viruses that infect cells of the mucosal tracts. IL-17 coregulated cytokines, such as IL-22, are involved in maintaining epithelial cell homeostasis and participate in epithelial cell repair/regeneration following inflammatory insults. Thus, the IL-17/IL-22 axis is important in both responding to, and recovering from, pathogens. However, aberrant expression or overexpression of IL-17 cytokines contributes to a number of pathological outcomes, including asthma, pneumonitis, and generation or exacerbation of pulmonary fibrosis. This review covers the good, bad, and ugly aspects of IL-17 in the lung.
Collapse
Affiliation(s)
- Stephen J Gurczynski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
112
|
Treerat P, Prince O, Cruz-Lagunas A, Muñoz-Torrico M, Salazar-Lezama MÁ, Selman M, Fallert-Junecko B, Reinhardt T, Alcorn JF, Kaushal D, Zuñiga J, Rangel-Moreno J, Kolls JK, Khader SA. Novel role for IL-22 in protection during chronic Mycobacterium tuberculosis HN878 infection. Mucosal Immunol 2017; 10:1069-1081. [PMID: 28247861 PMCID: PMC5477058 DOI: 10.1038/mi.2017.15] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/23/2017] [Indexed: 02/04/2023]
Abstract
Approximately 2 billion people are infected with Mycobacterium tuberculosis (Mtb), resulting in 1.4 million deaths every year. Among Mtb-infected individuals, clinical isolates belonging to the W-Beijing lineage are increasingly prevalent, associated with drug resistance, and cause severe disease immunopathology in animal models. Therefore, it is exceedingly important to identify the immune mechanisms that mediate protection against rapidly emerging Mtb strains, such as W-Beijing lineage. IL-22 is a member of the IL-10 family of cytokines with both protective and pathological functions at mucosal surfaces. Thus far, collective data show that IL-22 deficient mice are not more susceptible to aerosolized infection with less virulent Mtb strains. Thus, in this study we addressed the functional role for the IL-22 pathway in immunity to emerging Mtb isolates, using W-Beijing lineage member, Mtb HN878 as a prototype. We show that Mtb HN878 stimulates IL-22 production in TLR2 dependent manner and IL-22 mediates protective immunity during chronic stages of Mtb HN878 infection in mice. Interestingly, IL-22-dependent pathways in both epithelial cells and macrophages mediate protective mechanisms for Mtb HN878 control. Thus, our results project a new protective role for IL-22 in emerging Mtb infections.
Collapse
Affiliation(s)
- Puthayalai Treerat
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Oliver Prince
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Alfredo Cruz-Lagunas
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Marcela Muñoz-Torrico
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | | | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Beth Fallert-Junecko
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Todd Reinhardt
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F. Alcorn
- Division of Pulmonology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deepak Kaushal
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Centre, Covington, LA, USA
| | - Joaquin Zuñiga
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Centre, Rochester, NY, USA
| | - Jay K. Kolls
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
113
|
Andersson CK, Adams A, Nagakumar P, Bossley C, Gupta A, De Vries D, Adnan A, Bush A, Saglani S, Lloyd CM. Intraepithelial neutrophils in pediatric severe asthma are associated with better lung function. J Allergy Clin Immunol 2017; 139:1819-1829.e11. [PMID: 27746241 PMCID: PMC5457125 DOI: 10.1016/j.jaci.2016.09.022] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 08/04/2016] [Accepted: 09/06/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Neutrophils and IL-17A have been linked mechanistically in models of allergic airways disease and have been associated with asthma severity. However, their role in pediatric asthma is unknown. OBJECTIVES We sought to investigate the role of neutrophils and the IL-17A pathway in mediating pediatric severe therapy-resistant asthma (STRA). METHODS Children with STRA (n = 51; age, 12.6 years; range, 6-16.3 years) and controls without asthma (n = 15; age, 4.75 years; range, 1.6-16 years) underwent clinically indicated fiberoptic bronchoscopy, bronchoalveolar lavage (BAL), endobronchial brushings, and biopsy. Neutrophils, IL-17A, and IL-17RA-expressing cells and levels of IL-17A and IL-22 were quantified in BAL and biopsies and related to clinical features. Primary bronchial epithelial cells were stimulated with IL-17A and/or IL-22, with and without budesonide. RESULTS Children with STRA had increased intraepithelial neutrophils, which positively correlated with FEV1 %predicted (r = 0.43; P = .008). Neutrophilhigh patients also had better symptom control, despite lower dose maintenance inhaled steroids. Submucosal neutrophils were not increased in children with STRA. Submucosal and epithelial IL-17A-positive cells and BAL IL-17A and IL-22 levels were similar in children with STRA and controls. However, there were significantly more IL-17RA-positive cells in the submucosa and epithelium in children with STRA compared with controls (P = .001). Stimulation of primary bronchial epithelial cells with IL-17A enhanced mRNA expression of IL-17RA and increased release of IL-8, even in the presence of budesonide. CONCLUSIONS A proportion of children with STRA exhibit increased intraepithelial airway neutrophilia that correlated with better lung function. STRA was also characterized by increased airway IL-17RA expression. These data suggest a potential beneficial rather than adverse role for neutrophils in pediatric severe asthma pathophysiology.
Collapse
Affiliation(s)
- Cecilia K Andersson
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Alexandra Adams
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom; Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom
| | - Prasad Nagakumar
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom; Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom
| | - Cara Bossley
- Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom
| | - Atul Gupta
- Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom
| | - Daphne De Vries
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Afiqah Adnan
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Andrew Bush
- Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom
| | - Sejal Saglani
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom; Respiratory Paediatrics, the Royal Brompton and Harefield NHS Trust, Sydney Street, London, United Kingdom.
| | - Clare M Lloyd
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
114
|
Sasidharan S, Yajnik V, Khalili H, Garber J, Xavier R, Ananthakrishnan AN. Genetic risk factors for serious infections in inflammatory bowel diseases. Scand J Gastroenterol 2017; 52:570-576. [PMID: 28162010 PMCID: PMC5642969 DOI: 10.1080/00365521.2017.1286381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Immunosuppression, the cornerstone of management of Crohn's disease (CD) and ulcerative colitis (UC) (inflammatory bowel diseases; IBD) is associated with an increased risk of serious infections that is inadequately predicted by clinical risk factors. The role of genetics in determining susceptibility to infections is unknown. METHODS From a prospective-consented patient registry, we identified IBD patients with serious infections requiring hospitalization. Analysis was performed to identify IBD-related and non-IBD related immune response loci on the Immunochip that were associated with serious infections and a genetic risk score (GRS) representing the cumulative burden of the identified single nucleotide polymorphisms was calculated. Multivariable logistic regression used to identify effect of clinical and genetic factors. RESULTS The study included 1333 IBD patients (795 CD, 538 UC) with median disease duration of 13 years. A total of 133 patients (10%) had a serious infection requiring hospitalization. Patients with infections were more likely to have CD and had shorter disease duration. The most common infections were skin and soft-tissue, respiratory and urinary tract infections. Eight IBD risk loci and two other polymorphisms were significantly associations with serious infections. Each one point increase in the infection GRS was associated with a 50% increase in risk of infections (OR = 1.53, 95% CI = 1.37-1.70) (p = 1 × 10-14), confirmed on multivariable analysis. Genetic risk factors improved performance of a model predicting infections over clinical covariates alone (p < 0.001). CONCLUSIONS Genetic risk factors may predict susceptibility to infections in patients with IBD.
Collapse
Affiliation(s)
- Saranya Sasidharan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School
| | - Vijay Yajnik
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School
| | - Hamed Khalili
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School
| | - John Garber
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School
| | - Ramnik Xavier
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School
| | | |
Collapse
|
115
|
Detecting Perturbed Subpathways towards Mouse Lung Regeneration Following H1N1 Influenza Infection. COMPUTATION 2017. [DOI: 10.3390/computation5020020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
116
|
Tavares LP, Teixeira MM, Garcia CC. The inflammatory response triggered by Influenza virus: a two edged sword. Inflamm Res 2017; 66:283-302. [PMID: 27744631 DOI: 10.1007/s00011-016-0996-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV) is a relevant respiratory tract pathogen leading to a great number of deaths and hospitalizations worldwide. Secondary bacterial infections are a very common cause of IAV associated morbidity and mortality. The robust inflammatory response that follows infection is important for the control of virus proliferation but is also associated with lung damage, morbidity and death. The role of the different components of immune response underlying protection or disease during IAV infection is not completely elucidated. Overall, in the context of IAV infection, inflammation is a 'double edge sword' necessary to control infection but causing disease. Therefore, a growing number of studies suggest that immunomodulatory strategies may improve disease outcome without affecting the ability of the host to deal with infection. This review summarizes recent aspects of the inflammatory responses triggered by IAV that are preferentially involved in causing severe pulmonary disease and the anti-inflammatory strategies that have been suggested to treat influenza induced immunopathology.
Collapse
Affiliation(s)
- Luciana P Tavares
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristiana C Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, 21040360, Rio de Janeiro, Brazil.
| |
Collapse
|
117
|
IL-6 ameliorates acute lung injury in influenza virus infection. Sci Rep 2017; 7:43829. [PMID: 28262742 PMCID: PMC5338329 DOI: 10.1038/srep43829] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/01/2017] [Indexed: 12/20/2022] Open
Abstract
Interleukin 6 (IL-6) is involved in innate and adaptive immune responses to defend against pathogens. It also participates in the process of influenza infection by affecting viral clearance and immune cell responses. However, whether IL-6 impacts lung repair in influenza pathogenesis remains unclear. Here, we studied the role of IL-6 in acute influenza infection in mice. IL-6-deficient mice infected with influenza virus exhibited higher lethality, lost more body weight and had higher fibroblast accumulation and lower extracellular matrix (ECM) turnover in the lung than their wild-type counterparts. Deficiency in IL-6 enhanced proliferation, migration and survival of lung fibroblasts, as well as increased virus-induced apoptosis of lung epithelial cells. IL-6-deficient lung fibroblasts produced elevated levels of TGF-β, which may contribute to their survival. Furthermore, macrophage recruitment to the lung and phagocytic activities of macrophages during influenza infection were reduced in IL-6-deficient mice. Collectively, our results indicate that IL-6 is crucial for lung repair after influenza-induced lung injury through reducing fibroblast accumulation, promoting epithelial cell survival, increasing macrophage recruitment to the lung and enhancing phagocytosis of viruses by macrophages. This study suggests that IL-6 may be exploited for lung repair during influenza infection.
Collapse
|
118
|
Influenza A virus-induced release of interleukin-10 inhibits the anti-microbial activities of invariant natural killer T cells during invasive pneumococcal superinfection. Mucosal Immunol 2017; 10:460-469. [PMID: 27220813 DOI: 10.1038/mi.2016.49] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/07/2016] [Indexed: 02/04/2023]
Abstract
During influenza A virus (IAV) infection, changes in the lung's physical and immunological defenses predispose the host to bacterial superinfections. Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that have beneficial or harmful functions during infection. We investigated the iNKT cells' role in a model of invasive pneumococcal superinfection. The use of Jα18-/- mice indicated that iNKT cells limited susceptibility to influenza-pneumococcal infection and reduced the lethal synergism. This role did not depend on immune-based anti-bacterial mechanisms. At the time of bacterial exposure, iNKT cells from IAV-experienced mice failed to produce antipneumococcal interferon-γ and adoptive transfer of fresh iNKT cells before Streptococcus pneumoniae challenge did not restore anti-bacterial host defenses. Impaired iNKT cell activation in superinfected animals was related to the IAV-induced immunosuppressive cytokine interleukin-10 (IL-10), rather than to an intrinsic functional defect. IL-10 dampened the activation of iNKT cells in response to pneumococci by inhibiting the production of IL-12 by pulmonary monocyte-derived dendritic cells. Neutralization of IL-10 restored iNKT cell activation and tends to increase resistance to secondary bacterial infection. Overall, iNKT cells have a beneficial role (upstream of bacterial colonization) in controlling influenza-pneumococcal superinfection, although they represent novel targets of immunosuppression at the time of bacterial challenge.
Collapse
|
119
|
De Luna X, Hartshorn KL. Influenza Casts a Lung Shadow. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:697-699. [PMID: 28222276 DOI: 10.1016/j.ajpath.2017.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
This commentary highlights the article by Pociask et al that provides new insights into the lingering effects of influenza infection.
Collapse
Affiliation(s)
- Xavier De Luna
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, Massachusetts
| | - Kevan L Hartshorn
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
120
|
Pociask DA, Robinson KM, Chen K, McHugh KJ, Clay ME, Huang GT, Benos PV, Janssen-Heininger YMW, Kolls JK, Anathy V, Alcorn JF. Epigenetic and Transcriptomic Regulation of Lung Repair during Recovery from Influenza Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:851-863. [PMID: 28193481 DOI: 10.1016/j.ajpath.2016.12.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/07/2016] [Accepted: 12/21/2016] [Indexed: 11/28/2022]
Abstract
Seasonal and pandemic influenza is a cause of morbidity and mortality worldwide. Most people infected with influenza virus display mild-to-moderate disease phenotypes and recover within a few weeks. Influenza is known to cause persistent alveolitis in animal models; however, little is known about the molecular pathways involved in this phenotype. We challenged C57BL/6 mice with influenza A/PR/8/34 and examined lung pathologic processes and inflammation, as well as transcriptomic and epigenetic changes at 21 to 60 days after infection. Influenza induced persistent parenchymal lung inflammation, alveolar epithelial metaplasia, and epithelial endoplasmic reticulum stress that were evident after the clearance of virus and resolution of morbidity. Influenza infection induced robust changes in the lung transcriptome, including a significant impact on inflammatory and extracellular matrix protein expression. Despite the robust changes in lung gene expression, preceding influenza (21 days) did not exacerbate secondary Staphylococcus aureus infection. Finally, we examined the impact of influenza on miRNA expression in the lung and found an increase in miR-155. miR-155 knockout mice recovered from influenza infection faster than controls and had decreased lung inflammation and endoplasmic reticulum stress. These data illuminate the dynamic molecular changes in the lung in the weeks after influenza infection and characterize the repair process, identifying a novel role for miR-155.
Collapse
Affiliation(s)
- Derek A Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Richard K. Mellon Foundation Institute, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Keven M Robinson
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kong Chen
- Richard K. Mellon Foundation Institute, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Kevin J McHugh
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Michelle E Clay
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Grace T Huang
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; PhD Program in Computational Biology, Carnegie Mellon University and University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Jay K Kolls
- Richard K. Mellon Foundation Institute, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Vikas Anathy
- Department of Pathology, University of Vermont, Burlington, Vermont
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania.
| |
Collapse
|
121
|
Gray J, Oehrle K, Worthen G, Alenghat T, Whitsett J, Deshmukh H. Intestinal commensal bacteria mediate lung mucosal immunity and promote resistance of newborn mice to infection. Sci Transl Med 2017; 9:eaaf9412. [PMID: 28179507 PMCID: PMC5880204 DOI: 10.1126/scitranslmed.aaf9412] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022]
Abstract
Immature mucosal defenses contribute to increased susceptibility of newborn infants to pathogens. Sparse knowledge of age-dependent changes in mucosal immunity has hampered improvements in neonatal morbidity because of infections. We report that exposure of neonatal mice to commensal bacteria immediately after birth is required for a robust host defense against bacterial pneumonia, the leading cause of death in newborn infants. This crucial window was characterized by an abrupt influx of interleukin-22 (IL-22)-producing group 3 innate lymphoid cells (IL-22+ILC3) into the lungs of newborn mice. This influx was dependent on sensing of commensal bacteria by intestinal mucosal dendritic cells. Disruption of postnatal commensal colonization or selective depletion of dendritic cells interrupted the migratory program of lung IL-22+ILC3 and made the newborn mice more susceptible to pneumonia, which was reversed by transfer of commensal bacteria after birth. Thus, the resistance of newborn mice to pneumonia relied on commensal bacteria-directed ILC3 influx into the lungs, which mediated IL-22-dependent host resistance to pneumonia during this developmental window. These data establish that postnatal colonization by intestinal commensal bacteria is pivotal in the development of the lung defenses of newborns.
Collapse
Affiliation(s)
- Jerilyn Gray
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45219, USA
| | - Katherine Oehrle
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45219, USA
| | - George Worthen
- Children's Hospital of Philadelphia, 34th Street and Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Theresa Alenghat
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45219, USA
| | - Jeffrey Whitsett
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45219, USA
| | - Hitesh Deshmukh
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45219, USA.
| |
Collapse
|
122
|
Jouan Y, Si-Tahar M, Guillon A. Immunité de la muqueuse respiratoire : physiologie et implications en réanimation. MEDECINE INTENSIVE REANIMATION 2016. [DOI: 10.1007/s13546-016-1245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
123
|
Bayes HK, Ritchie ND, Ward C, Corris PA, Brodlie M, Evans TJ. IL-22 exacerbates weight loss in a murine model of chronic pulmonary Pseudomonas aeruginosa infection. J Cyst Fibros 2016; 15:759-768. [PMID: 27375092 PMCID: PMC5154339 DOI: 10.1016/j.jcf.2016.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND Interleukin (IL)-22 is a critical mediator of mucosal immunity and tissue regeneration, protecting against a number of respiratory pathogens. Whether IL-22 confers protection against chronic Pseudomonas aeruginosa (PA) infection in cystic fibrosis (CF) is unknown. METHODS Explanted CF lungs were examined for IL-22 production and immune-localization. A murine model of persistent pulmonary PA infection was used to examine production of IL-22 following infective challenge. The role of IL-22 was examined using IL-22 knockout (KO) animals. RESULTS IL-22 is produced within the adult CF lung and localizes to the airway epithelium. IL-22 is produced by murine pulmonary lymph node cells following lung infection. The absence of IL-22 resulted in no significant difference in acute mortality, bacterial burden, chronic infection rates, histological changes or neutrophilic inflammation in the chronic PA infection model. However, IL-22 KO animals lost less weight following infection. CONCLUSION IL-22 is produced in the CF lung and in response to PA infection yet is dispensable in protection against chronic pulmonary P. aeruginosa infection in a murine model. However, we identified a novel role for the cytokine in promoting infection-related weight-loss, a significant prognostic factor in the CF population.
Collapse
Affiliation(s)
- Hannah K Bayes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA, United Kingdom.
| | - Neil D Ritchie
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Christopher Ward
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Paul A Corris
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Malcolm Brodlie
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Paediatric Respiratory Medicine, Great North Children's Hospital, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Thomas J Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA, United Kingdom
| |
Collapse
|
124
|
Abstract
The cytokine interleukin-22 (IL-22), which is a member of the IL-10 family, is produced exclusively by immune cells and activates signal transducer and activator of transcription 3 (STAT3) in nonimmune cells, such as hepatocytes, keratinocytes, and colonic epithelial cells, to drive various processes central to tissue homeostasis and immunosurveillance. Dysregulation of IL-22 signaling causes inflammatory diseases. IL-22 binding protein (IL-22BP; encoded by IL22RA2) is a soluble IL-22 receptor, which antagonizes IL-22 activity and has genetic associations with autoimmune diseases. Humans have three IL-22BP isoforms, IL-22BPi1 to IL-22BPi3, which are generated by alternative splicing; mice only have an IL-22BPi2 homolog. We showed that, although IL-22BPi3 had less inhibitory activity than IL-22BPi2, IL-22BPi3 was more abundant in various human tissues under homeostatic conditions. IL-22BPi2 was more effective than IL-22BPi3 at blocking the contribution of IL-22 to cooperative gene induction with the inflammatory cytokine IL-17, which is often present with IL-22 in autoimmune settings. In addition, we found that IL-22BPi1 was not secreted and therefore failed to antagonize IL-22 signaling. Furthermore, IL-22BPi2 was the only isoform that was increased in abundance when myeloid cells were activated by Toll-like receptor 2 signaling or retinoic acid, a maturation factor for myeloid cells. These data suggest that the human IL-22BP isoforms have distinct spatial and temporal roles and coordinately fine-tune IL-22-dependent STAT3 responses in tissues as a type of rheostat.
Collapse
Affiliation(s)
- Chrissie Lim
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - MeeAe Hong
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
125
|
Hall OJ, Limjunyawong N, Vermillion MS, Robinson DP, Wohlgemuth N, Pekosz A, Mitzner W, Klein SL. Progesterone-Based Therapy Protects Against Influenza by Promoting Lung Repair and Recovery in Females. PLoS Pathog 2016; 12:e1005840. [PMID: 27631986 PMCID: PMC5025002 DOI: 10.1371/journal.ppat.1005840] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/02/2016] [Indexed: 01/07/2023] Open
Abstract
Over 100 million women use progesterone therapies worldwide. Despite having immunomodulatory and repair properties, their effects on the outcome of viral diseases outside of the reproductive tract have not been evaluated. Administration of exogenous progesterone (at concentrations that mimic the luteal phase) to progesterone-depleted adult female mice conferred protection from both lethal and sublethal influenza A virus (IAV) infection. Progesterone treatment altered the inflammatory environment of the lungs, but had no effects on viral load. Progesterone treatment promoted faster recovery by increasing TGF-β, IL-6, IL-22, numbers of regulatory Th17 cells expressing CD39, and cellular proliferation, reducing protein leakage into the airway, improving pulmonary function, and upregulating the epidermal growth factor amphiregulin (AREG) in the lungs. Administration of rAREG to progesterone-depleted females promoted pulmonary repair and improved the outcome of IAV infection. Progesterone-treatment of AREG-deficient females could not restore protection, indicating that progesterone-mediated induction of AREG caused repair in the lungs and accelerated recovery from IAV infection. Repair and production of AREG by damaged respiratory epithelial cell cultures in vitro was increased by progesterone. Our results illustrate that progesterone is a critical host factor mediating production of AREG by epithelial cells and pulmonary tissue repair following infection, which has important implications for women's health.
Collapse
Affiliation(s)
- Olivia J. Hall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nathachit Limjunyawong
- Department of Environmental Health Sciences, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Meghan S. Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Dionne P. Robinson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nicholas Wohlgemuth
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Environmental Health Sciences, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Wayne Mitzner
- Department of Environmental Health Sciences, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
126
|
Trevejo-Nunez G, Elsegeiny W, Conboy P, Chen K, Kolls JK. Critical Role of IL-22/IL22-RA1 Signaling in Pneumococcal Pneumonia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1877-83. [PMID: 27456484 PMCID: PMC4992592 DOI: 10.4049/jimmunol.1600528] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022]
Abstract
IL-22-IL-22R signaling plays a crucial role in regulating host defenses against extracellular pathogens, particularly in the intestine, through the induction of antimicrobial peptides and chemotactic genes. However, the role of IL-22-IL-22R is understudied in Streptococcus pneumoniae lung infection, a prevalent pathogen of pneumonia. This paper presents the findings of IL-22 signaling during a murine model of pneumococcal pneumonia and improvement of bacterial burden upon IL-22 administration. IL-22 was rapidly induced in the lung during pneumococcal infection in wild-type mice, and Il22(-/-) mice had higher pneumococcal burdens compared with controls. Additionally, mice with hepatic-specific deletion of Il22ra1 also had higher bacterial burdens in lungs compared with littermate controls after intrapulmonary pneumococcal infection, suggesting that IL-22 signaling in the liver is important to control pneumococcal pneumonia. Thus, we hypothesized that enhancement of IL-22 signaling would control pneumococcal burden in lung tissues in an experimental pneumonia model. Administration of rIL-22 systemically to infected wild-type mice decreased bacterial burden in lung and liver at 24 h postinfection. Our in vitro studies also showed that mice treated with IL-22 had increased C3 expression in the liver compared with the isotype control group. Furthermore, serum from mice treated with IL-22 had improved opsonic capacity by increasing C3 binding on S. pneumoniae Taken together, endogenous IL-22 and hepatic IL-22R signaling play critical roles in controlling pneumococcal lung burden, and systemic IL-22 decreases bacterial burden in the lungs and peripheral organs by potentiating C3 opsonization on bacterial surfaces, through the increase of hepatic C3 expression.
Collapse
Affiliation(s)
- Giraldina Trevejo-Nunez
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224
| | - Waleed Elsegeiny
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224
| | - Parker Conboy
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224
| | - Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224
| |
Collapse
|
127
|
Wang K, Langevin S, O’Hern CS, Shattuck MD, Ogle S, Forero A, Morrison J, Slayden R, Katze MG, Kirby M. Anomaly Detection in Host Signaling Pathways for the Early Prognosis of Acute Infection. PLoS One 2016; 11:e0160919. [PMID: 27532264 PMCID: PMC4988711 DOI: 10.1371/journal.pone.0160919] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 07/27/2016] [Indexed: 01/09/2023] Open
Abstract
Clinical diagnosis of acute infectious diseases during the early stages of infection is critical to administering the appropriate treatment to improve the disease outcome. We present a data driven analysis of the human cellular response to respiratory viruses including influenza, respiratory syncytia virus, and human rhinovirus, and compared this with the response to the bacterial endotoxin, Lipopolysaccharides (LPS). Using an anomaly detection framework we identified pathways that clearly distinguish between asymptomatic and symptomatic patients infected with the four different respiratory viruses and that accurately diagnosed patients exposed to a bacterial infection. Connectivity pathway analysis comparing the viral and bacterial diagnostic signatures identified host cellular pathways that were unique to patients exposed to LPS endotoxin indicating this type of analysis could be used to identify host biomarkers that can differentiate clinical etiologies of acute infection. We applied the Multivariate State Estimation Technique (MSET) on two human influenza (H1N1 and H3N2) gene expression data sets to define host networks perturbed in the asymptomatic phase of infection. Our analysis identified pathways in the respiratory virus diagnostic signature as prognostic biomarkers that triggered prior to clinical presentation of acute symptoms. These early warning pathways correctly predicted that almost half of the subjects would become symptomatic in less than forty hours post-infection and that three of the 18 subjects would become symptomatic after only 8 hours. These results provide a proof-of-concept for utility of anomaly detection algorithms to classify host pathway signatures that can identify presymptomatic signatures of acute diseases and differentiate between etiologies of infection. On a global scale, acute respiratory infections cause a significant proportion of human co-morbidities and account for 4.25 million deaths annually. The development of clinical diagnostic tools to distinguish between acute viral and bacterial respiratory infections is critical to improve patient care and limit the overuse of antibiotics in the medical community. The identification of prognostic respiratory virus biomarkers provides an early warning system that is capable of predicting which subjects will become symptomatic to expand our medical diagnostic capabilities and treatment options for acute infectious diseases. The host response to acute infection may be viewed as a deterministic signaling network responsible for maintaining the health of the host organism. We identify pathway signatures that reflect the very earliest perturbations in the host response to acute infection. These pathways provide a monitor the health state of the host using anomaly detection to quantify and predict health outcomes to pathogens.
Collapse
Affiliation(s)
- Kun Wang
- Department of Mathematics, Colorado State University, Fort Collins, CO, United States of America
- Department of Mechanical Engineering & Materials Science, Yale University, New Haven, CT, United States of America
| | - Stanley Langevin
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Corey S. O’Hern
- Department of Mechanical Engineering & Materials Science, Yale University, New Haven, CT, United States of America
- Department of Applied Physics, Department of Physics, and Graduate Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, United States of America
| | - Mark D. Shattuck
- Department of Mechanical Engineering & Materials Science, Yale University, New Haven, CT, United States of America
- Department of Physics and Benjamin Levich Institute, The City College of the City University of New York, New York, NY, United States of America
| | - Serenity Ogle
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Adriana Forero
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Juliet Morrison
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Richard Slayden
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States of America
| | - Michael G. Katze
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Michael Kirby
- Department of Mathematics, Colorado State University, Fort Collins, CO, United States of America
- Department of Computer Science, Colorado State University, Fort Collins, CO, United States of America
- * E-mail:
| |
Collapse
|
128
|
Jamieson AM. Host resilience to emerging coronaviruses. Future Virol 2016; 11:529-534. [PMID: 32201496 PMCID: PMC7079962 DOI: 10.2217/fvl-2016-0060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022]
Abstract
Recently, two coronaviruses, severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus, have emerged to cause unusually severe respiratory disease in humans. Currently, there is a lack of effective antiviral treatment options or vaccine available. Given the severity of these outbreaks, and the possibility of additional zoonotic coronaviruses emerging in the near future, the exploration of different treatment strategies is necessary. Disease resilience is the ability of a given host to tolerate an infection, and to return to a state of health. This review focuses on exploring various host resilience mechanisms that could be exploited for treatment of severe acute respiratory syndrome coronavirus, Middle East respiratory syndrome coronavirus and other respiratory viruses that cause acute lung injury and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Amanda M Jamieson
- Department of Microbiology and Immunology, Brown University, Providence, RI, USA
| |
Collapse
|
129
|
Hiraoka T, Hirota Y, Saito-Fujita T, Matsuo M, Egashira M, Matsumoto L, Haraguchi H, Dey SK, Furukawa KS, Fujii T, Osuga Y. STAT3 accelerates uterine epithelial regeneration in a mouse model of decellularized uterine matrix transplantation. JCI Insight 2016; 1:87591. [PMID: 27358915 PMCID: PMC4922514 DOI: 10.1172/jci.insight.87591] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022] Open
Abstract
Although a close connection between uterine regeneration and successful pregnancy in both humans and mice has been consistently observed, its molecular basis remains unclear. We here established a mouse model of decellularized uterine matrix (DUM) transplantation. Resected mouse uteri were processed with SDS to make DUMs without any intact cells. DUMs were transplanted into the mouse uteri with artificially induced defects, and all the uterine layers were recovered at the DUM transplantation sites within a month. In the regenerated uteri, normal hormone responsiveness in early pregnancy was observed, suggesting the regeneration of functional uteri. Uterine epithelial cells rapidly migrated and formed a normal uterine epithelial layer within a week, indicating a robust epithelial-regenerating capacity. Stromal and myometrial regeneration occurred following epithelial regeneration. In ovariectomized mice, uterine regeneration of the DUM transplantation was similarly observed, suggesting that ovarian hormones are not essential for this regeneration process. Importantly, the regenerating epithelium around the DUM demonstrated heightened STAT3 phosphorylation and cell proliferation, which was suppressed in uteri of Stat3 conditional knockout mice. These data suggest a key role of STAT3 in the initial step of the uterine regeneration process. The DUM transplantation model is a powerful tool for uterine regeneration research.
Collapse
Affiliation(s)
- Takehiro Hiraoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Precursory Research for Innovative Medical Care (PRIME), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Tomoko Saito-Fujita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsunori Matsuo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mahiro Egashira
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Leona Matsumoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Haraguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sudhansu K Dey
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katsuko S Furukawa
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
130
|
Clinical and immunological analysis of measles patients admitted to a Beijing hospital in 2014 during an outbreak in China. Epidemiol Infect 2016; 144:2613-20. [PMID: 27251754 DOI: 10.1017/s0950268816001114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
At the end of 2013, China reported a countrywide outbreak of measles. From January to May 2014, we investigated the clinical and immunological features of the cases of the outbreak admitted to our hospital. In this study, all 112 inpatients with clinically diagnosed measles were recruited from the 302 Military Hospital of China. The virus was isolated from throat swabs from these patients, and cytokine profiles were examined. By detecting the measles virus of 30 of the 112 patients, we found that this measles outbreak was of the H1 genotype, which is the major strain in China. The rates of complications, specifically pneumonia and liver injury, differed significantly in patients aged 18 years: pneumonia was more common in children, while liver injury was more common in adults. Pneumonia was a significant independent risk factor affecting measles duration. Compared to healthy subjects, measles patients had fewer CD4+IL-17+, CD4+IFN-γ +, and CD8+IFN-γ + cells in both the acute and recovery phases. In contrast, measles patients in the acute phase had more CD8+IL-22+ cells than those in recovery or healthy subjects. We recommend that future studies focus on the age-related distribution of pneumonia and liver injury as measles-related complications as well as the association between immunological markers and measles prognosis.
Collapse
|
131
|
Abstract
Asthma is a complex, heterogeneous disorder with increasing prevalence. It is now recognized that several asthma phenotypes exist, including type 2-high and type 2-low (or non-type 2) subsets. As current research strives to identify subgroups of asthmatics that share disease pathobiology to establish endotypes, efforts to clarify the underlying molecular mechanisms of disease are needed and essential. IL-22 is thought to be a mediator of asthma pathogenesis, but whether this cytokine has a pathologic or beneficial role in the lung during severe disease is still debated. Studies focused on the regulation of this cytokine by its receptors and other inflammatory mediators during allergic airway responses are necessary to clarify its role in disease. Here, we discuss the ambiguity surrounding the role of IL-22 in asthma and considerations for targeting IL-22 therapeutically.
Collapse
Affiliation(s)
- Michelle L Manni
- a Department of Pediatrics , Children's Hospital of Pittsburgh of UPMC , Pittsburgh , PA , USA
| | - John F Alcorn
- a Department of Pediatrics , Children's Hospital of Pittsburgh of UPMC , Pittsburgh , PA , USA
| |
Collapse
|
132
|
Gimeno Brias S, Stack G, Stacey MA, Redwood AJ, Humphreys IR. The Role of IL-22 in Viral Infections: Paradigms and Paradoxes. Front Immunol 2016; 7:211. [PMID: 27303405 PMCID: PMC4885595 DOI: 10.3389/fimmu.2016.00211] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022] Open
Abstract
Interleukin-22 (IL-22) is a member of the IL-10 family of cytokines. Hematopoietic cells express IL-22, and this cytokine signals through the heterodimeric IL-22 receptor expressed by non-hematopoietic cells. A growing body of evidence points toward a role for IL-22 in a diverse array of biological functions ranging from cellular proliferation, tissue protection and regeneration, and inflammation. In recent years, the role that IL-22 plays in antiviral immune responses has been examined in a number of infection models. Herein, we assess our current understanding of how IL-22 determines the outcome of viral infections and define common mechanisms that are evident from, sometimes paradoxical, findings derived from these studies. Finally, we discuss the potential therapeutic utility of IL-22 manipulation in the treatment and prevention of viral infections and associated pathologies.
Collapse
Affiliation(s)
- Silvia Gimeno Brias
- Institute of Infection and Immunity, Cardiff University, Cardiff, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| | - Gabrielle Stack
- Institute of Infection and Immunity, Cardiff University, Cardiff, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| | - Maria A Stacey
- Institute of Infection and Immunity, Cardiff University, Cardiff, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| | - Alec J Redwood
- The Institute for Immunology and Infectious Diseases, Murdoch University , Murdoch, WA , Australia
| | - Ian R Humphreys
- Institute of Infection and Immunity, Cardiff University, Cardiff, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
133
|
Perusina Lanfranca M, Lin Y, Fang J, Zou W, Frankel T. Biological and pathological activities of interleukin-22. J Mol Med (Berl) 2016; 94:523-34. [PMID: 26923718 PMCID: PMC4860114 DOI: 10.1007/s00109-016-1391-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-22, a member of the IL-10 family, is a cytokine secreted by several types of immune cells including IL-22(+)CD4(+) T cells (Th22) and IL-22 expressing innate leukocytes (ILC22). Recent studies have demonstrated that IL-22 is a key component in mucosal barrier defense, tissue repair, epithelial cell survival, and proliferation. Furthermore, accumulating evidence has defined both protective and pathogenic properties of IL-22 in a number of conditions including autoimmune disease, infection, and malignancy. In this review, we summarize the expression and signaling pathway and functional characteristics of the IL-22 and IL-22 receptor axis in physiological and pathological scenarios and discuss the potential to target IL-22 signaling to treat human diseases.
Collapse
Affiliation(s)
- Mirna Perusina Lanfranca
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yanwei Lin
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
- Graduate Programs in Immunology and Tumor Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Timothy Frankel
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
134
|
Dugger DT, Gerriets JE, Miller LA. Attenuated Airway Epithelial Cell Interleukin-22R1 Expression in the Infant Nonhuman Primate Lung. Am J Respir Cell Mol Biol 2016; 53:761-8. [PMID: 26309027 DOI: 10.1165/rcmb.2014-0452rc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Respiratory tract infections are a leading cause of morbidity and mortality in children under 5 years of age. Increased susceptibility to infection is associated with deficiencies in immunity during early childhood. Airway epithelium represents the first line of mucosal defense against inhaled pathogens. However, little is known about epithelial immune mechanisms in the maturing lung. IL-22 and its receptor IL-22R1 are important in host defense and repair of epithelial barriers. The objective of this study was to determine whether a quantitative difference in IL-22R1 exists between infant and adult airways using the rhesus macaque monkey as a model of childhood lung development. Immunofluorescence staining of tracheal tissue revealed minimal expression of IL-22R1 in epithelium at 1 month of age, with a progressive increase in fluorescence-positive basal cells through 1 year of age. Western blot analysis of tracheal lysates confirmed significant age-dependent differences in IL-22R1 protein content. Further, primary tracheobronchial epithelial cell cultures established from infant and adult monkeys showed differential IL-22R1 mRNA and protein expression in vitro. To begin to assess the regulation of age-dependent IL-22R1 expression in airway epithelium, the effect of histone deacetylase and DNA methyltransferase inhibitors was evaluated. IL-22R1 mRNA in adult cultures was not altered by 5-aza-2'-deoxycytidine or trichostatin A. IL-22R1 mRNA in infant cultures showed no change with 5-aza-2'-deoxycytidine but was significantly increased after trichostatin A treatment; however, IL-22R1 protein did not increase concurrently. These data suggest that IL-22R1 in airway epithelium is regulated, in part, by epigenetic mechanisms that are dependent on chronologic age.
Collapse
Affiliation(s)
- Daniel T Dugger
- 1 California National Primate Research Center, University of California, Davis, California; and
| | - Joan E Gerriets
- 1 California National Primate Research Center, University of California, Davis, California; and
| | - Lisa A Miller
- 1 California National Primate Research Center, University of California, Davis, California; and.,2 Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, California
| |
Collapse
|
135
|
Zissler UM, Esser-von Bieren J, Jakwerth CA, Chaker AM, Schmidt-Weber CB. Current and future biomarkers in allergic asthma. Allergy 2016; 71:475-94. [PMID: 26706728 DOI: 10.1111/all.12828] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
Diagnosis early in life, sensitization, asthma endotypes, monitoring of disease and treatment progression are key motivations for the exploration of biomarkers for allergic rhinitis and allergic asthma. The number of genes related to allergic rhinitis and allergic asthma increases steadily; however, prognostic genes have not yet entered clinical application. We hypothesize that the combination of multiple genes may generate biomarkers with prognostic potential. The current review attempts to group more than 161 different potential biomarkers involved in respiratory inflammation to pave the way for future classifiers. The potential biomarkers are categorized into either epithelial or infiltrate-derived or mixed origin, epithelial biomarkers. Furthermore, surface markers were grouped into cell-type-specific categories. The current literature provides multiple biomarkers for potential asthma endotypes that are related to T-cell phenotypes such as Th1, Th2, Th9, Th17, Th22 and Tregs and their lead cytokines. Eosinophilic and neutrophilic asthma endotypes are also classified by epithelium-derived CCL-26 and osteopontin, respectively. There are currently about 20 epithelium-derived biomarkers exclusively derived from epithelium, which are likely to innovate biomarker panels as they are easy to sample. This article systematically reviews and categorizes genes and collects current evidence that may promote these biomarkers to become part of allergic rhinitis or allergic asthma classifiers with high prognostic value.
Collapse
Affiliation(s)
- U. M. Zissler
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - J. Esser-von Bieren
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - C. A. Jakwerth
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - A. M. Chaker
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery; Medical School; Technical University of Munich; Munich Germany
| | - C. B. Schmidt-Weber
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| |
Collapse
|
136
|
Rathore JS, Wang Y. Protective role of Th17 cells in pulmonary infection. Vaccine 2016; 34:1504-1514. [PMID: 26878294 DOI: 10.1016/j.vaccine.2016.02.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 01/14/2023]
Abstract
Th17 cells are characterized as preferential producer of interleukins including IL-17A, IL-17F, IL-21 and IL-22. Corresponding receptors of these cytokines are expressed on number of cell types found in the mucosa, including epithelial cells and fibroblasts which constitute the prime targets of the Th17-associated cytokines. Binding of IL-17 family members to their corresponding receptors lead to modulation of antimicrobial functions of target cells including alveolar epithelial cells. Stimulated alveolar epithelial cells produce antimicrobial peptides and are involved in granulepoesis, neutrophil recruitment and tissue repair. Mucosal immunity mediated by Th17 cells is protective against numerous pulmonary pathogens including extracellular bacterial and fungal pathogens. This review focuses on the protective role of Th17 cells during pulmonary infection, highlighting subset differentiation, effector cytokines production, followed by study of the binding of these cytokines to their corresponding receptors, the subsequent signaling pathway they engender and their effector role in host defense.
Collapse
Affiliation(s)
- Jitendra Singh Rathore
- University of Pennsylvania, Perelman School of Medicine, Department of Microbiology, Philadelphia, PA, USA; Gautam Buddha University, School of Biotechnology, Greater Noida, Yamuna Expressway, Uttar Pradesh, India.
| | - Yan Wang
- University of Pennsylvania, Perelman School of Medicine, Department of Microbiology, Philadelphia, PA, USA
| |
Collapse
|
137
|
Parker D, Ahn D, Cohen T, Prince A. Innate Immune Signaling Activated by MDR Bacteria in the Airway. Physiol Rev 2016; 96:19-53. [PMID: 26582515 DOI: 10.1152/physrev.00009.2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Health care-associated bacterial pneumonias due to multiple-drug resistant (MDR) pathogens are an important public health problem and are major causes of morbidity and mortality worldwide. In addition to antimicrobial resistance, these organisms have adapted to the milieu of the human airway and have acquired resistance to the innate immune clearance mechanisms that normally prevent pneumonia. Given the limited efficacy of antibiotics, bacterial clearance from the airway requires an effective immune response. Understanding how specific airway pathogens initiate and regulate innate immune signaling, and whether this response is excessive, leading to host-induced pathology may guide future immunomodulatory therapy. We will focus on three of the most important causes of health care-associated pneumonia, Staphylococcus aureus, Pseudomonas aeruginosa, and Klebsiella pneumoniae, and review the mechanisms through which an inappropriate or damaging innate immune response is stimulated, as well as describe how airway pathogens cause persistent infection by evading immune activation.
Collapse
Affiliation(s)
- Dane Parker
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Danielle Ahn
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Taylor Cohen
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Alice Prince
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| |
Collapse
|
138
|
Huan C, Kim D, Ou P, Alfonso A, Stanek A. Mechanisms of interleukin-22's beneficial effects in acute pancreatitis. World J Gastrointest Pathophysiol 2016; 7:108-116. [PMID: 26909233 PMCID: PMC4753176 DOI: 10.4291/wjgp.v7.i1.108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/11/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
Acute pancreatitis (AP) is a disorder characterized by parenchymal injury of the pancreas controlled by immune cell-mediated inflammation. AP remains a significant challenge in the clinic due to a lack of specific and effective treatment. Knowledge of the complex mechanisms that regulate the inflammatory response in AP is needed for the development of new approaches to treatment, since immune cell-derived inflammatory cytokines have been recognized to play critical roles in the pathogenesis of the disease. Recent studies have shown that interleukin (IL)-22, a cytokine secreted by leukocytes, when applied in the severe animal models of AP, protects against the inflammation-mediated acinar injury. In contrast, in a mild AP model, endogenous IL-22 has been found to be a predominantly anti-inflammatory mediator that inhibits inflammatory cell infiltration via the induction of Reg3 proteins in acinar cells, but does not protect against acinar injury in the early stage of AP. However, constitutively over-expressed IL-22 can prevent the initial acinar injury caused by excessive autophagy through the induction of the anti-autophagic proteins Bcl-2 and Bcl-XL. Thus IL-22 plays different roles in AP depending on the severity of the AP model. This review focuses on these recently reported findings for the purpose of better understanding IL-22's regulatory roles in AP which could help to develop a novel therapeutic strategy.
Collapse
|
139
|
Sakaguchi R, Chikuma S, Shichita T, Morita R, Sekiya T, Ouyang W, Ueda T, Seki H, Morisaki H, Yoshimura A. Innate-like function of memory Th17 cells for enhancing endotoxin-induced acute lung inflammation through IL-22. Int Immunol 2015; 28:233-43. [PMID: 26647405 DOI: 10.1093/intimm/dxv070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/20/2015] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS)-induced acute lung injury (ALI) is known as a mouse model of acute respiratory distress syndrome; however, the function of T-cell-derived cytokines in ALI has not yet been established. We found that LPS challenge in one lung resulted in a rapid induction of innate-type pro-inflammatory cytokines such as IL-6 and TNF-α, followed by the expression of T-cell-type cytokines, including IL-17, IL-22 and IFN-γ. We discovered that IL-23 is important for ALI, since blockage of IL-23 by gene disruption or anti-IL-12/23p40 antibody treatment reduced neutrophil infiltration and inflammatory cytokine secretion into the bronchoalveolar lavage fluid (BALF). IL-23 was mostly produced from F4/80(+)CD11c(+) alveolar macrophages, and IL-23 expression was markedly reduced by the pre-treatment of mice with antibiotics, suggesting that the development of IL-23-producing macrophages required commensal bacteria. Unexpectedly, among T-cell-derived cytokines, IL-22 rather than IL-17 or IFN-γ played a major role in LPS-induced ALI. IL-22 protein levels were higher than IL-17 in the BALF after LPS instillation, and the major source of IL-22 was memory Th17 cells. Lung memory CD4(+) T cells had a potential to produce IL-22 at higher levels than IL-17 in response to IL-1β plus IL-23 without TCR stimulation. Our study revealed an innate-like function of the lung memory Th17 cells that produce IL-22 in response to IL-23 and are involved in exaggeration of ALI.
Collapse
Affiliation(s)
- Ryota Sakaguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan Department of Anesthesiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takashi Shichita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan PRESTO (Precursory Research for Embryonic Science and Technology), Chiyoda-ku, Tokyo 102-0075, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takashi Sekiya
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wenjun Ouyang
- Department of Immunology, Genentech, 1 DNA Way, San Francisco, CA 94080, USA
| | - Tomomi Ueda
- Department of Anesthesiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroyuki Seki
- Department of Anesthesiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroshi Morisaki
- Department of Anesthesiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
140
|
Avitabile S, Odorisio T, Madonna S, Eyerich S, Guerra L, Eyerich K, Zambruno G, Cavani A, Cianfarani F. Interleukin-22 Promotes Wound Repair in Diabetes by Improving Keratinocyte Pro-Healing Functions. J Invest Dermatol 2015; 135:2862-2870. [PMID: 26168231 DOI: 10.1038/jid.2015.278] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/08/2015] [Accepted: 06/18/2015] [Indexed: 11/09/2022]
Abstract
Impaired re-epithelialization, imbalanced expression of cytokines and growth factors, and vascular disease contribute to healing impairment in diabetes. IL-22, a pro-inflammatory cytokine mediating a cross-talk between immune system and epithelial cells, has been shown to have a role in repair processes. In this study we aimed to investigate IL-22 regenerative potential in the poor healing context of diabetic wounds. By using streptozotocin-induced diabetic mice, we demonstrated that IL-22 wound treatment significantly accelerated the healing process, by promoting re-epithelialization, granulation tissue formation, and vascularization. Improved re-epithelialization was associated with increased keratinocyte proliferation and signal transducer and activator of transcription 3 (STAT3) activation. We showed that endogenous IL-22 content was reduced at both mRNA and protein level during the inflammatory phase of diabetic wounds, with fewer IL-22-positive cells infiltrating the granulation tissue. We demonstrated that IL-22 treatment promoted proliferation and injury repair of hyperglycemic keratinocytes and induced activation of STAT3 and extracellular signal-regulated kinase transduction pathways in keratinocytes grown in hyperglycemic condition or isolated from diabetic patients. Finally, we demonstrated that IL-22 treatment was able to inhibit diabetic keratinocyte differentiation while promoting vascular endothelial growth factor release. Our data indicate a pro-healing role of IL-22 in diabetic wounds, suggesting a therapeutic potential for this cytokine in diabetic ulcer management.
Collapse
Affiliation(s)
- Simona Avitabile
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Teresa Odorisio
- Laboratory of Biochemistry, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefania Madonna
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefanie Eyerich
- ZAUM - Center of Allergy and Environment, Technische Universität and Helmholtz Center, Munich, Germany
| | - Liliana Guerra
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Kilian Eyerich
- Department of Dermatology and Allergy, Technische Universität, Munich, Germany
| | - Giovanna Zambruno
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Andrea Cavani
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| | - Francesca Cianfarani
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| |
Collapse
|
141
|
Hidvegi T, Stolz DB, Alcorn JF, Yousem SA, Wang J, Leme AS, Houghton AM, Hale P, Ewing M, Cai H, Garchar EA, Pastore N, Annunziata P, Kaminski N, Pilewski J, Shapiro SD, Pak SC, Silverman GA, Brunetti-Pierri N, Perlmutter DH. Enhancing Autophagy with Drugs or Lung-directed Gene Therapy Reverses the Pathological Effects of Respiratory Epithelial Cell Proteinopathy. J Biol Chem 2015; 290:29742-57. [PMID: 26494620 DOI: 10.1074/jbc.m115.691253] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 11/06/2022] Open
Abstract
Recent studies have shown that autophagy mitigates the pathological effects of proteinopathies in the liver, heart, and skeletal muscle but this has not been investigated for proteinopathies that affect the lung. This may be due at least in part to the lack of an animal model robust enough for spontaneous pathological effects from proteinopathies even though several rare proteinopathies, surfactant protein A and C deficiencies, cause severe pulmonary fibrosis. In this report we show that the PiZ mouse, transgenic for the common misfolded variant α1-antitrypsin Z, is a model of respiratory epithelial cell proteinopathy with spontaneous pulmonary fibrosis. Intracellular accumulation of misfolded α1-antitrypsin Z in respiratory epithelial cells of the PiZ model resulted in activation of autophagy, leukocyte infiltration, and spontaneous pulmonary fibrosis severe enough to elicit functional restrictive deficits. Treatment with autophagy enhancer drugs or lung-directed gene transfer of TFEB, a master transcriptional activator of the autophagolysosomal system, reversed these proteotoxic consequences. We conclude that this mouse is an excellent model of respiratory epithelial proteinopathy with spontaneous pulmonary fibrosis and that autophagy is an important endogenous proteostasis mechanism and an attractive target for therapy.
Collapse
Affiliation(s)
- Tunda Hidvegi
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | - John F Alcorn
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | | | | | | | - Pamela Hale
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Michael Ewing
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Houming Cai
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Evelyn Akpadock Garchar
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Nunzia Pastore
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | - Patrizia Annunziata
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | | | | | | | - Stephen C Pak
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Gary A Silverman
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138 Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy, 80131, and
| | - David H Perlmutter
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| |
Collapse
|
142
|
Abstract
The HIV epidemic has clearly demonstrated the critical role CD4(+) T cells play in preventing opportunistic infections in the lung. The types of CD4(+) effector T-cell populations in the lung have significantly expanded over the last 8-10 years with the discovery of helper T type 17 cells, and this review summarizes the field and discusses how these effector cells may be exploited to augment mucosal immunity in the lung.
Collapse
|
143
|
Liu Y, Kumar VS, Zhang W, Rehman J, Malik AB. Activation of type II cells into regenerative stem cell antigen-1(+) cells during alveolar repair. Am J Respir Cell Mol Biol 2015; 53:113-24. [PMID: 25474582 DOI: 10.1165/rcmb.2013-0497oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The alveolar epithelium is composed of two cell types: type I cells comprise 95% of the gas exchange surface area, whereas type II cells secrete surfactant, while retaining the ability to convert into type I cells to induce alveolar repair. Using lineage-tracing analyses in the mouse model of Pseudomonas aeruginosa-induced lung injury, we identified a population of stem cell antigen (Sca)-1-expressing type II cells with progenitor cell properties that mediate alveolar repair. These cells were shown to be distinct from previously reported Sca-1-expressing bronchioalveolar stem cells. Microarray and Wnt reporter studies showed that surfactant protein (Sp)-C(+)Sca-1(+) cells expressed Wnt signaling pathway genes, and inhibiting Wnt/β-catenin signaling prevented the regenerative function of Sp-C(+)Sca-1(+) cells in vitro. Thus, P. aeruginosa-mediated lung injury induces the generation of a Sca-1(+) subset of type II cells. The progenitor phenotype of the Sp-C(+)Sca-1(+) cells that mediates alveolar epithelial repair might involve Wnt signaling.
Collapse
Affiliation(s)
| | | | | | - Jalees Rehman
- Departments of 1 Pharmacology.,3 Medicine, University of Illinois College of Medicine, Chicago, Illinois
| | | |
Collapse
|
144
|
Guillon A, Jouan Y, Brea D, Gueugnon F, Dalloneau E, Baranek T, Henry C, Morello E, Renauld JC, Pichavant M, Gosset P, Courty Y, Diot P, Si-Tahar M. Neutrophil proteases alter the interleukin-22-receptor-dependent lung antimicrobial defence. Eur Respir J 2015; 46:771-82. [PMID: 26250498 DOI: 10.1183/09031936.00215114] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/20/2015] [Indexed: 01/01/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is punctuated by episodes of infection-driven acute exacerbations. Despite the life-threatening nature of these exacerbations, the underlying mechanisms remain unclear, although a high number of neutrophils in the lungs of COPD patients is known to correlate with poor prognosis. Interleukin (IL)-22 is a cytokine that plays a pivotal role in lung antimicrobial defence and tissue protection. We hypothesised that neutrophils secrete proteases that may have adverse effects in COPD, by altering the IL-22 receptor (IL-22R)-dependent signalling.Using in vitro and in vivo approaches as well as reverse transcriptase quantitative PCR, flow cytometry and/or Western blotting techniques, we first showed that pathogens such as the influenza virus promote IL-22R expression in human bronchial epithelial cells, whereas Pseudomonas aeruginosa, bacterial lipopolysaccharide or cigarette smoke do not. Most importantly, neutrophil proteases cleave IL-22R and impair IL-22-dependent immune signalling and expression of antimicrobial effectors such as β-defensin-2. This proteolysis resulted in the release of a soluble fragment of IL-22R, which was detectable both in cellular and animal models as well as in sputa from COPD patients with acute exacerbations.Hence, our study reveals an unsuspected regulation by the proteolytic action of neutrophil enzymes of IL-22-dependent lung host response. This process probably enhances pathogen replication, and ultimately COPD exacerbations.
Collapse
Affiliation(s)
- Antoine Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France Service de Réanimation Polyvalente, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Youenn Jouan
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France Service de Réanimation Polyvalente, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Deborah Brea
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Fabien Gueugnon
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Emilie Dalloneau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Thomas Baranek
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Clémence Henry
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Eric Morello
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Brussels, Belgium De Duve Institute, Universite Catholique de Louvain, Brussels, Belgium
| | - Muriel Pichavant
- Université Lille Nord de France, Lille, France Lille Centre for Infection and Immunity, Institut Pasteur de Lille, Lille, France Unité Mixte de Recherche 8204, Centre National de la Recherche Scientifique, Lille, France INSERM, U1019, Team 8, Lille, France
| | - Philippe Gosset
- Université Lille Nord de France, Lille, France Lille Centre for Infection and Immunity, Institut Pasteur de Lille, Lille, France Unité Mixte de Recherche 8204, Centre National de la Recherche Scientifique, Lille, France INSERM, U1019, Team 8, Lille, France
| | - Yves Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Patrice Diot
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, Tours, France Université François Rabelais de Tours, Tours, France
| |
Collapse
|
145
|
Sahaza JH, Suárez-Alvarez R, Estrada-Bárcenas DA, Pérez-Torres A, Taylor ML. Profile of cytokines in the lungs of BALB/c mice after intra-nasal infection with Histoplasma capsulatum mycelial propagules. Comp Immunol Microbiol Infect Dis 2015; 41:1-9. [PMID: 26264521 DOI: 10.1016/j.cimid.2015.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 12/27/2022]
Abstract
The host pulmonary response to the fungus Histoplasma capsulatum was evaluated, through the profile of cytokines detected by the MagPix magnetic beads platform in lung homogenates and by lung-granulomas formation, from mice intra-nasally infected with mycelial propagules (M-phase) of two virulent H. capsulatum strains, EH-46 and G-217B. Results highlight that mice lung inflammatory response depends on the H. capsulatum strain used, during the first step of the fungal infection. IL-1β and TNF-α increased their concentrations in mice infected with both strains. The highest levels of IL-6, IL-17, and IL-23 were found in EH-46-infected mice, whereas levels of IL-22 were variable at all post-infection times for both strains. Significant increases of IL-12, IFN-γ, IL-4, and IL-10 were associated to EH-46-infected mice. Histological lung findings from EH-46-infected mice revealed incipient and numerous well-developed granulomas, distributed in lung-lobes at the 14th and the 21st days after infection, according to cytokine profiles.
Collapse
Affiliation(s)
- Jorge Humberto Sahaza
- Laboratorio de Inmunología de Hongos, Unidad de Micología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México DF, Mexico; Unidad de Micología Médica y Experimental, Corporación para Investigaciones Biológicas, Medellín, Colombia
| | | | - Daniel Alfonso Estrada-Bárcenas
- Laboratorio de Inmunología de Hongos, Unidad de Micología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México DF, Mexico; Colección Nacional de Cultivos Microbianos, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, México DF, Mexico
| | - Armando Pérez-Torres
- Laboratorio de Filogenia del Sistema Inmune de Piel y Mucosas, Departamento de Biología Celular y Tisular, Facultad de Medicina, UNAM, México DF, Mexico
| | - Maria Lucia Taylor
- Laboratorio de Inmunología de Hongos, Unidad de Micología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México DF, Mexico.
| |
Collapse
|
146
|
The Human IL-22 Receptor Is Regulated through the Action of the Novel E3 Ligase Subunit FBXW12, Which Functions as an Epithelial Growth Suppressor. J Immunol Res 2015; 2015:912713. [PMID: 26171402 PMCID: PMC4480942 DOI: 10.1155/2015/912713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/22/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Interleukin- (IL-) 22 signaling is protective in animal models of pneumonia and bacteremia by Klebsiella pneumoniae and mediates tissue recovery from influenza and Staph aureus infection. We recently described processing of mouse lung epithelial IL-22 receptor (IL-22R) by ubiquitination on the intracellular C-terminal. To identify cellular factors that regulate human IL-22R, we screened receptor abundance while overexpressing constituents of the ubiquitin system and identify that IL-22R can be shuttled for degradation by multiple previously uncharacterized F-box protein E3 ligase subunits. We observe that in human cells IL-22R is destabilized by FBXW12. FBXW12 causes depletion of endogenous and plasmid-derived IL-22R in lung epithelia, binds the E3 ligase constituent Skp-1, and facilitates ubiquitination of IL-22R in vitro. FBXW12 knockdown with shRNA increases IL-22R abundance and STAT3 phosphorylation in response to IL-22 cytokine treatment. FBXW12 shRNA increases human epithelial cell growth and cell cycle progression with enhanced constitutive activity of map kinases JNK and ERK. These findings indicate that the heretofore-undescribed protein FBXW12 functions as an E3 ligase constituent to ubiquitinate and degrade IL-22R and that therapeutic FBXW12 inhibition may enhance IL-22 signaling and bolster mucosal host defense and infection containment.
Collapse
|
147
|
Blaauboer SM, Mansouri S, Tucker HR, Wang HL, Gabrielle VD, Jin L. The mucosal adjuvant cyclic di-GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo. eLife 2015; 4. [PMID: 25898005 PMCID: PMC4428110 DOI: 10.7554/elife.06670] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/21/2015] [Indexed: 12/22/2022] Open
Abstract
Effective mucosal adjuvants enhance the magnitude and quality of the vaccine response. Cyclic di-GMP (CDG) is a promising mucosal vaccine adjuvant. However, its in vivo mechanisms are unclear. Here, we showed, in mice, that CDG elicits stronger Ab and TH responses than the mammalian 2'3'-cyclic GMP-AMP (cGAMP), and generated better protection against Streptococcus pneumoniae infection than 2'3'-cGAMP adjuvanted vaccine. We identified two in vivo mechanisms of CDG. First, intranasally administered CDG greatly enhances Ag uptake, including pinocytosis and receptor-mediated endocytosis in vivo. The enhancement depends on MPYS (STING, MITA) expression in CD11C(+) cells. Second, we found that CDG selectively activated pinocytosis-efficient-DCs, leading to T(H) polarizing cytokines IL-12p70, IFNγ, IL-5, IL-13, IL-23, and IL-6 production in vivo. Notably, CDG induces IFNλ, but not IFNβ, in vivo. Our study revealed previously unrecognized in vivo functions of MPYS and advanced our understanding of CDG as a mucosal vaccine adjuvant.
Collapse
Affiliation(s)
- Steven M Blaauboer
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| | - Samira Mansouri
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| | - Heidi R Tucker
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| | - Hatti L Wang
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| | - Vincent D Gabrielle
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| | - Lei Jin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States
| |
Collapse
|
148
|
Herold S, Becker C, Ridge KM, Budinger GRS. Influenza virus-induced lung injury: pathogenesis and implications for treatment. Eur Respir J 2015; 45:1463-78. [PMID: 25792631 DOI: 10.1183/09031936.00186214] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/07/2015] [Indexed: 01/21/2023]
Abstract
The influenza viruses are some of the most important human pathogens, causing substantial seasonal and pandemic morbidity and mortality. In humans, infection of the lower respiratory tract of can result in flooding of the alveolar compartment, development of acute respiratory distress syndrome and death from respiratory failure. Influenza-mediated damage of the airway, alveolar epithelium and alveolar endothelium results from a combination of: 1) intrinsic viral pathogenicity, attributable to its tropism for host airway and alveolar epithelial cells; and 2) a robust host innate immune response, which, while contributing to viral clearance, can worsen the severity of lung injury. In this review, we summarise the molecular events at the virus-host interface during influenza virus infection, highlighting some of the important cellular responses. We discuss immune-mediated viral clearance, the mechanisms promoting or perpetuating lung injury, lung regeneration after influenza-induced injury, and recent advances in influenza prevention and therapy.
Collapse
Affiliation(s)
- Susanne Herold
- Dept of Internal Medicine II, Universities Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christin Becker
- Dept of Internal Medicine II, Universities Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Karen M Ridge
- Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - G R Scott Budinger
- Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
149
|
Busman-Sahay KO, Walrath T, Huber S, O'Connor W. Cytokine crowdsourcing: multicellular production of TH17-associated cytokines. J Leukoc Biol 2015; 97:499-510. [PMID: 25548251 PMCID: PMC5477895 DOI: 10.1189/jlb.3ru0814-386r] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/17/2014] [Accepted: 11/20/2014] [Indexed: 12/14/2022] Open
Abstract
In the 2 decades since its discovery, IL-17A has become appreciated for mounting robust, protective responses against bacterial and fungal pathogens. When improperly regulated, however, IL-17A can play a profoundly pathogenic role in perpetuating inflammation and has been linked to a wide variety of debilitating diseases. IL-17A is often present in a composite milieu that includes cytokines produced by TH17 cells (i.e., IL-17F, IL-21, IL-22, and IL-26) or associated with other T cell lineages (e.g., IFN-γ). These combinatorial effects add mechanistic complexity and more importantly, contribute differentially to disease outcome. Whereas TH17 cells are among the best-understood cell types that secrete IL-17A, they are frequently neither the earliest nor dominant producers. Indeed, non-TH17 cell sources of IL-17A can dramatically alter the course and severity of inflammatory episodes. The dissection of the temporal regulation of TH17-associated cytokines and the resulting net signaling outcomes will be critical toward understanding the increasingly intricate role of IL-17A and TH17-associated cytokines in disease, informing our therapeutic decisions. Herein, we discuss important non-TH17 cell sources of IL-17A and other TH17-associated cytokines relevant to inflammatory events in mucosal tissues.
Collapse
Affiliation(s)
- Kathleen O Busman-Sahay
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Travis Walrath
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - William O'Connor
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
150
|
Abstract
Interleukin-22 (IL-22) is a recently described IL-10 family cytokine that is produced by T helper (Th) 17 cells, γδ T cells, NKT cells, and newly described innate lymphoid cells (ILCs). Knowledge of IL-22 biology has evolved rapidly since its discovery in 2000, and a role for IL-22 has been identified in numerous tissues, including the intestines, lung, liver, kidney, thymus, pancreas, and skin. IL-22 primarily targets nonhematopoietic epithelial and stromal cells, where it can promote proliferation and play a role in tissue regeneration. In addition, IL-22 regulates host defense at barrier surfaces. However, IL-22 has also been linked to several conditions involving inflammatory tissue pathology. In this review, we assess the current understanding of this cytokine, including its physiologic and pathologic effects on epithelial cell function.
Collapse
|