1
|
Shadid A, Rich HE, DeVaughn H, Domozhirov A, Doursout MF, Weng-Mills T, Eckel-Mahan KL, Karmouty-Quintana H, Restrepo MI, Shivshankar P. Persistent microbial infections and idiopathic pulmonary fibrosis - an insight into non-typeable Haemophilus influenza pathogenesis. Front Cell Infect Microbiol 2024; 14:1479801. [PMID: 39760094 PMCID: PMC11695292 DOI: 10.3389/fcimb.2024.1479801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Interstitial lung disease (ILD) is characterized by chronic inflammation and scarring of the lungs, of which idiopathic pulmonary fibrosis (IPF) is the most devastating pathologic form. Idiopathic pulmonary fibrosis pathogenesis leads to loss of lung function and eventual death in 50% of patients, making it the leading cause of ILD-associated mortality worldwide. Persistent and subclinical microbial infections are implicated in the acute exacerbation of chronic lung diseases. However, while epidemiological studies have highlighted pollutants, gastric aspirate, and microbial infections as major causes for the progression and exacerbation of IPF, the role of persistent microbial infections in the pathogenesis of IPF remains unclear. In this review, we have focused on the role of persistent microbial infections, including viral, bacterial, and fungal infections, and their mechanisms of action in the pathogenesis of IPF. In particular, the mechanisms and pathogenesis of the Gram-negative bacteria Non-typeable Haemophilus influenzae (NTHi) in ILDs are discussed, along with growing evidence of its role in IPF, given its unique ability to establish persistent intracellular infections by leveraging its non-capsulated nature to evade host defenses. While antibiotic treatments are presumably beneficial to target the extracellular, interstitial, and systemic burden of pathogens, their effects are significantly reduced in combating pathogens that reside in the intracellular compartments. The review also includes recent clinical trials, which center on combinatorial treatments involving antimicrobials and immunosuppressants, along with antifibrotic drugs that help mitigate disease progression in IPF patients. Finally, future directions focus on mRNA-based therapeutics, given their demonstrated effectiveness across a wide range of clinical applications and feasibility in targeting intracellular pathogens.
Collapse
Affiliation(s)
- Anthony Shadid
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Haydn E. Rich
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Hunter DeVaughn
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Aleksey Domozhirov
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Marie- Françoise Doursout
- Department of Anesthesiology, Critical Care and Pain Medicine, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Tingting Weng-Mills
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Kristin L. Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Marcos I. Restrepo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, South Texas Veterans Health Care System and the University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pooja Shivshankar
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| |
Collapse
|
2
|
Ye X, Zhang M, Gu H, Liu M, Zhao Y, Shi Y, Wu S, Jiang C, Ye X, Zhu H, Li Q, Huang X, Cao M. Animal models of acute exacerbation of pulmonary fibrosis. Respir Res 2023; 24:296. [PMID: 38007420 PMCID: PMC10675932 DOI: 10.1186/s12931-023-02595-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive scarring interstitial lung disease with an unknown cause. Some patients may experience acute exacerbations (AE), which result in severe lung damage visible on imaging or through examination of tissue samples, often leading to high mortality rates. However, the etiology and pathogenesis of AE-IPF remain unclear. AE-IPF patients exhibit diffuse lung damage, apoptosis of type II alveolar epithelial cells, and an excessive inflammatory response. Establishing a reliable animal model of AE is critical for investigating the pathogenesis. Recent studies have reported a variety of animal models for AE-IPF, each with its own advantages and disadvantages. These models are usually established in mice with bleomycin-induced pulmonary fibrosis, using viruses, bacteria, small peptides, or specific drugs. In this review, we present an overview of different AE models, hoping to provide a useful resource for exploring the mechanisms and targeted therapies for AE-IPF.
Collapse
Affiliation(s)
- Xu Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Mingrui Zhang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Gu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Mengying Liu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yichao Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanchen Shi
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shufei Wu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Jiang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huihui Zhu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Li
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinmei Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| | - Mengshu Cao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| |
Collapse
|
3
|
A Phase I Randomized, Controlled, Clinical Trial of Valganciclovir in Idiopathic Pulmonary Fibrosis. Ann Am Thorac Soc 2021; 18:1291-1297. [PMID: 33740394 DOI: 10.1513/annalsats.202102-108oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rationale: Human herpesviruses Epstein-Barr virus and cytomegalovirus are frequently detectable in the lungs of patients with idiopathic pulmonary fibrosis (IPF) and could contribute to disease pathogenesis. Objectives: With the goal of inhibiting herpesvirus replication, we tested the safety and tolerability of adding valganciclovir to standard IPF therapy (pirfenidone). Methods: We performed a single-center, Phase I, double-blind, randomized, placebo-controlled trial comparing valganciclovir 900 mg daily with placebo in patients with IPF with serologic evidence of prior Epstein-Barr virus and/or cytomegalovirus infection who were tolerating full-dose pirfenidone (2,403 mg/d). Subjects were randomized to valganciclovir or placebo 2:1 for 12 weeks of active treatment with off-treatment follow-up for up to 12 months. The primary safety endpoint was the number of subjects discontinuing the study drug before completing 12 weeks of treatment. Results: Thirty-one subjects with IPF were randomized to valganciclovir (n = 20) or placebo (n = 11). All subjects completed assigned therapy except one subject in the valganciclovir group, who discontinued the study drug after developing a rash. The total number of adverse events was similar between study groups. In a prespecified analysis of secondary physiologic endpoints, we observed a trend toward improved forced vital capacity from randomization to Week 12 in valganciclovir-treated subjects (-10 ml; interquartile range [IQR], -65 to 70 ml) versus placebo-treated subjects (40 ml; IQR, -130 to 60 ml), which persisted through 12 months of follow-up. Conclusions: Valganciclovir is safe and well tolerated as an add-on therapy to pirfenidone in patients with IPF. Clinical trial registered with ClinicalTrials.gov (NCT02871401).
Collapse
|
4
|
Duckworth A, Longhurst HJ, Paxton JK, Scotton CJ. The Role of Herpes Viruses in Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:704222. [PMID: 34368196 PMCID: PMC8339799 DOI: 10.3389/fmed.2021.704222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/24/2021] [Indexed: 12/29/2022] Open
Abstract
Pulmonary fibrosis (PF) is a serious lung disease which can result from known genetic or environmental exposures but is more commonly idiopathic (IPF). In familial PF (FPF), the majority of identified causal genes play key roles in the maintenance of telomeres, the protective end structures of chromosomes. Recent evidence suggests that short telomeres may also be implicated causally in a significant proportion of idiopathic cases. The possible involvement of herpes viruses in PF disease incidence and progression has been examined for many years, with some studies showing strong, statistically significant associations and others reporting no involvement. Evidence is thus polarized and remains inconclusive. Here we review the reported involvement of herpes viruses in PF in both animals and humans and present a summary of the evidence to date. We also present several possible mechanisms of action of the different herpes viruses in PF pathogenesis, including potential contributions to telomere attrition and cellular senescence. Evidence for antiviral treatment in PF is very limited but suggests a potential benefit. Further work is required to definitely answer the question of whether herpes viruses impact PF disease onset and progression and to enable the possible use of targeted antiviral treatments to improve clinical outcomes.
Collapse
Affiliation(s)
- Anna Duckworth
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Hilary J. Longhurst
- Department of Medicine, University of Auckland, Auckland, New Zealand
- Dyskeratosis Congenita (DC) Action, London, United Kingdom
| | - Jane K. Paxton
- Dyskeratosis Congenita (DC) Action, London, United Kingdom
| | - Chris J. Scotton
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
5
|
Crisan-Dabija R, Pavel CA, Popa IV, Tarus A, Burlacu A. "A Chain Only as Strong as Its Weakest Link": An Up-to-Date Literature Review on the Bidirectional Interaction of Pulmonary Fibrosis and COVID-19. J Proteome Res 2020; 19:4327-4338. [PMID: 32883081 PMCID: PMC7640958 DOI: 10.1021/acs.jproteome.0c00387] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic rapidly became a worldwide healthcare emergency affecting millions of people, with poor outcomes for patients with chronic conditions and enormous pressure on healthcare systems. Pulmonary fibrosis (PF) has been cited as a risk factor for a more severe evolution of COVID-19, primarily because its acute exacerbations are already associated with high mortality. We reviewed the available literature on biochemical, pathophysiological, and pharmacological mechanisms of PF and COVID-19 in an attempt to foresee the particular risk of infection and possible evolution of PF patients if infected with SARS-COV-2. We also analyzed the possible role of medication and risk factors (such as smoking) in the disease's evolution and clinical course. We found out that there is a complexity of interactions between coexisting idiopathic pulmonary fibrosis/interstitial lung disease (ILD) and COVID-19 disease. Also, patients recovering from severe COVID-19 disease are at serious risk of developing PF. Smokers seem to have, in theory, a chance for a better outcome if they develop a severe form of COVID-19 but statistically are at much higher risk of dying if they become critically ill.
Collapse
Affiliation(s)
- Radu Crisan-Dabija
- Clinic
of Pulmonary Diseases Iasi, Iasi, Romania
- ‘Grigore
T. Popa’ University of Medicine, Iasi, Romania
| | | | - Iolanda Valentina Popa
- ‘Grigore
T. Popa’ University of Medicine, Iasi, Romania
- Institute
of Gastroenterology and Hepatology, Iasi, Romania
| | - Andrei Tarus
- ‘Grigore
T. Popa’ University of Medicine, Iasi, Romania
- Department
of Cardiovascular Surgery, Cardiovascular
Diseases Institute, Iasi, Romania
| | - Alexandru Burlacu
- ‘Grigore
T. Popa’ University of Medicine, Iasi, Romania
- Department
of Interventional Cardiology, Cardiovascular
Diseases Institute, Iasi Romania
| |
Collapse
|
6
|
Turianová L, Lachová V, Svetlíkova D, Kostrábová A, Betáková T. Comparison of cytokine profiles induced by nonlethal and lethal doses of influenza A virus in mice. Exp Ther Med 2019; 18:4397-4405. [PMID: 31777543 PMCID: PMC6862669 DOI: 10.3892/etm.2019.8096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
Influenza viruses are among the most common human pathogens and are responsible for causing extensive seasonal morbidity and mortality. To investigate the immunological factors associated with severe influenza infection, the immune responses in mice infected with nonlethal (LD0) doses of A/PR/8/34 (H1N1) influenza virus were compared with those of mice infected with a lethal dose (LD100) of the virus. The virus titer and activation of retinoic acid-inducible gene (RIG)-I-like receptor signaling pathways were similar in the mice infected with LD0 and LD100 at 2 days post-infection; however, mice infected with LD100 exhibited a greater abundance of cytokines and a more diverse cytokine profile. Infection with LD100 induced the expression of the following factors: Interleukins (ILs), IL-4, IL-7, IL-10, IL-11, IL-12p40, IL-13 and IL-15; inflammatory chemokines, C-C motif chemokine ligand (CCL)2, CCL3/4, CCL12, CCL17, CCL19; and lung injury-associated cytokines, leptin, leukaemia inhibitory factor, macrophage colony stimulating factor, pentraxin (PTX)2 and PTX3, WNT1-inducible-signaling pathway protein 1, matrix metallopeptidase (MMP)-2, MMP-3, proprotein convertase subtilisin/kexin type 9, and T cell immunoglobulin and mucin domain. Switching in macrophage polarization from M1 to M2 was evidenced by the increase in M2 markers, including arginase-1 (Arg1) and early growth response protein 2 (Egr2), in the lungs of mice infected with LD100. Since IL-12 and interferon-γ are the major T helper (Th)1 cytokines, increased expression of interferon regulatory factor 4, IL-4, IL-10 and IL-13 promoted the differentiation of naïve CD4+ T cells into Th2 cells. In conclusion, the present study identified key cytokines involved in the pathogenicity of influenza infection, and demonstrated that lethal influenza virus infection induces a mixed Th1/Th2 response.
Collapse
Affiliation(s)
- Lucia Turianová
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Veronika Lachová
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Darina Svetlíkova
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Anna Kostrábová
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| | - Tatiana Betáková
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic.,Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| |
Collapse
|
7
|
Innate immunity and Toll-like receptor signaling in the pathogenesis of scleroderma: advances and opportunities for therapy. Curr Opin Rheumatol 2019; 30:600-605. [PMID: 30234721 DOI: 10.1097/bor.0000000000000542] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is an autoimmune connective tissue disease in which inflammation and cytokine dysregulation leads to skin fibrosis. Toll-like receptors (TLRs) are conserved pattern recognition receptors, recognizing pathogens danger-associated molecular patterns (DAMPs) that elicit a cascade of proinflammatory signaling. Recently, TLRs have been found to be critically important in SSc pathogenesis, with increased levels of the TLRs and their ligands present in the disease. Animal models have also been pivotal in delineating the role of these innate immune receptors in SSc. This current review examines the role of TLRs and the most recent evidence of the role of DAMPs and how these may be exploited therapeutically. RECENT FINDINGS Increasingly, studies have demonstrated the key roles of TLR4 and other intracellular TLRs in mediating fibrosis in SSc patients and animal models. TLR4 activation appears a key point and novel DAMPs, expressed upon tissue damage, appear critical in mediating the profibrotic effect through a downstream enhancement of transforming growth factor β. Deletion of Tenascin-C or a splice variant of fibronectin ameliorates animal models of skin fibrosis. Intracellular, nucleic acid sensing, TLR8 is critical in activating macrophages to secrete profibrotic molecules. The mechanism involves histone modification through epigenetic modifying enzymes. SUMMARY TLRs are key therapeutic targets in SSc.
Collapse
|
8
|
Invernizzi R, Molyneaux PL. The contribution of infection and the respiratory microbiome in acute exacerbations of idiopathic pulmonary fibrosis. Eur Respir Rev 2019; 28:28/152/190045. [DOI: 10.1183/16000617.0045-2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/16/2019] [Indexed: 01/25/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) arises in genetically susceptible individuals as a result of an aberrant wound-healing response following repetitive alveolar injury. The clinical course of the disease remains both variable and unpredictable with periods of more rapid decline, termed acute exacerbation of IPF (AE-IPF), often punctuating the disease trajectory. Exacerbations carry a significant morbidity and mortality, and their exact pathogenesis remains unclear. Given the emerging evidence that disruption and alteration in the lung microbiome plays a role in the pathogenesis and progression of IPF, this review discusses the current knowledge of the contribution of infection and the respiratory microbiome to AE-IPF.
Collapse
|
9
|
Martínez-Colón GJ, Warheit-Niemi H, Gurczynski SJ, Taylor QM, Wilke CA, Podsiad AB, Crespo J, Bhan U, Moore BB. Influenza-induced immune suppression to methicillin-resistant Staphylococcus aureus is mediated by TLR9. PLoS Pathog 2019; 15:e1007560. [PMID: 30682165 PMCID: PMC6364947 DOI: 10.1371/journal.ppat.1007560] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/06/2019] [Accepted: 01/03/2019] [Indexed: 12/15/2022] Open
Abstract
Bacterial lung infections, particularly with methicillin-resistant Staphylococcus aureus (MRSA), increase mortality following influenza infection, but the mechanisms remain unclear. Here we show that expression of TLR9, a microbial DNA sensor, is increased in murine lung macrophages, dendritic cells, CD8+ T cells and epithelial cells post-influenza infection. TLR9-/- mice did not show differences in handling influenza nor MRSA infection alone. However, TLR9-/- mice have improved survival and bacterial clearance in the lung post-influenza and MRSA dual infection, with no difference in viral load during dual infection. We demonstrate that TLR9 is upregulated on macrophages even when they are not themselves infected, suggesting that TLR9 upregulation is related to soluble mediators. We rule out a role for elevations in interferon-γ (IFNγ) in mediating the beneficial MRSA clearance in TLR9-/- mice. While macrophages from WT and TLR9-/- mice show similar phagocytosis and bacterial killing to MRSA alone, following influenza infection, there is a marked upregulation of scavenger receptor A and MRSA phagocytosis as well as inducible nitric oxide synthase (Inos) and improved bacterial killing that is specific to TLR9-deficient cells. Bone marrow transplant chimera experiments and in vitro experiments using TLR9 antagonists suggest TLR9 expression on non-hematopoietic cells, rather than the macrophages themselves, is important for regulating myeloid cell function. Interestingly, improved bacterial clearance post-dual infection was restricted to MRSA, as there was no difference in the clearance of Streptococcus pneumoniae. Taken together these data show a surprising inhibitory role for TLR9 signaling in mediating clearance of MRSA that manifests following influenza infection. Influenza-associated secondary bacterial infections, particularly with methicillin-resistant Staphylococcus aureus (MRSA), are a major cause of morbidity and mortality, and better therapeutic strategies are needed. Stimulation of TLR2 has shown promise for improving health in influenza-bacteria dual-infected animals. However, nothing is known about the role of other TLRs, including TLR9, in influenza-bacteria dual infection pathology. This is the first study of TLR9 regulation of influenza-bacterial superinfection and it highlights an unexpected pathologic role for TLR9 in regulating clearance of MRSA post-H1N1. It also highlights the important observation that TLR9 signaling has very different outcomes in the setting of influenza infection than in naïve mice and shows important distinctions in the mechanisms for susceptibility to MRSA vs. S. pneumoniae post-influenza. Our results also suggest that TLR9 expression on non-hematopoietic cells regulates macrophage function in vivo.
Collapse
Affiliation(s)
| | - Helen Warheit-Niemi
- Microbiology and Immunology Graduate Program, University of Michigan, Ann Arbor, MI United States of America
| | - Stephen J. Gurczynski
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Quincy M. Taylor
- Literature, Sciences and the Arts, Microbiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Carol A. Wilke
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Amy B. Podsiad
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Joel Crespo
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - Urvashi Bhan
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Bethany B. Moore
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
10
|
Bortz E, Wu TT, Patel P, Whitelegge JP, Sun R. Proteomics of Bronchoalveolar Lavage Fluid Reveals a Lung Oxidative Stress Response in Murine Herpesvirus-68 Infection. Viruses 2018; 10:v10120670. [PMID: 30486363 PMCID: PMC6316452 DOI: 10.3390/v10120670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Murine herpesvirus-68 (MHV-68) productively infects mouse lungs, exhibiting a complex pathology characteristic of both acute viral infections and chronic respiratory diseases. We sought to discover proteins differentially expressed in bronchoalveolar lavage (BAL) from mice infected with MHV-68. Mice were infected intranasally with MHV-68. After nine days, as the lytic phase of infection resolved, differential BAL proteins were identified by two-dimensional (2D) electrophoresis and mass spectrometry. Of 23 unique proteins, acute phase proteins, vitamin A transport, and oxidative stress response factors Pdx6 and EC-SOD (Sod3) were enriched. Correspondingly, iNOS2 was induced in lung tissue by seven days post-infection. Oxidative stress was partly a direct result of MHV-68 infection, as reactive oxygen species (ROS) were induced in cultured murine NIH3T3 fibroblasts and human lung A549 cells infected with MHV-68. Finally, mice infected with a recombinant MHV-68 co-expressing inflammatory cytokine murine interleukin 6 (IL6) showed exacerbated oxidative stress and soluble type I collagen characteristic of tissue recovery. Thus, oxidative stress appears to be a salient feature of MHV-68 pathogenesis, in part caused by lytic replication of the virus and IL6. Proteins and small molecules in lung oxidative stress networks therefore may provide new therapeutic targets to ameliorate respiratory virus infections.
Collapse
Affiliation(s)
- Eric Bortz
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA.
| | - Ting-Ting Wu
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Parthive Patel
- Center for Molecular Biology and German Cancer Research Center (DKFZ), University of Heidelberg (ZMBH), 69120 Heidelberg, Germany.
| | - Julian P Whitelegge
- The Pasarow Mass Spectrometry Laboratory & the Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Ren Sun
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Natural Secretory Immunoglobulins Promote Enteric Viral Infections. J Virol 2018; 92:JVI.00826-18. [PMID: 30232191 DOI: 10.1128/jvi.00826-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/05/2018] [Indexed: 12/27/2022] Open
Abstract
Noroviruses are enteric pathogens causing significant morbidity, mortality, and economic losses worldwide. Secretory immunoglobulins (sIg) are a first line of mucosal defense against enteric pathogens. They are secreted into the intestinal lumen via the polymeric immunoglobulin receptor (pIgR), where they bind to antigens. However, whether natural sIg protect against norovirus infection remains unknown. To determine if natural sIg alter murine norovirus (MNV) pathogenesis, we infected pIgR knockout (KO) mice, which lack sIg in mucosal secretions. Acute MNV infection was significantly reduced in pIgR KO mice compared to controls, despite increased MNV target cells in the Peyer's patch. Natural sIg did not alter MNV binding to the follicle-associated epithelium (FAE) or crossing of the FAE into the lymphoid follicle. Instead, naive pIgR KO mice had enhanced levels of the antiviral inflammatory molecules interferon gamma (IFN-γ) and inducible nitric oxide synthase (iNOS) in the ileum compared to controls. Strikingly, depletion of the intestinal microbiota in pIgR KO and control mice resulted in comparable IFN-γ and iNOS levels, as well as MNV infectious titers. IFN-γ treatment of wild-type (WT) mice and neutralization of IFN-γ in pIgR KO mice modulated MNV titers, implicating the antiviral cytokine in the phenotype. Reduced gastrointestinal infection in pIgR KO mice was also observed with another enteric virus, reovirus. Collectively, our findings suggest that natural sIg are not protective during enteric virus infection, but rather, that sIg promote enteric viral infection through alterations in microbial immune responses.IMPORTANCE Enteric virus, such as norovirus, infections cause significant morbidity and mortality worldwide. However, direct antiviral infection prevention strategies are limited. Blocking host entry and initiation of infection provides an established avenue for intervention. Here, we investigated the role of the polymeric immunoglobulin receptor (pIgR)-secretory immunoglobulin (sIg) cycle during enteric virus infections. The innate immune functions of sIg (agglutination, immune exclusion, neutralization, and expulsion) were not required during control of acute murine norovirus (MNV) infection. Instead, lack of pIgR resulted in increased IFN-γ levels, which contributed to reduced MNV titers. Another enteric virus, reovirus, also showed decreased infection in pIgR KO mice. Collectively, our data point to a model in which sIg-mediated microbial sensing promotes norovirus and reovirus infection. These data provide the first evidence of the proviral role of natural sIg during enteric virus infections and provide another example of how intestinal bacterial communities indirectly influence MNV pathogenesis.
Collapse
|
12
|
Kim KK, Dotson MR, Agarwal M, Yang J, Bradley PB, Subbotina N, Osterholzer JJ, Sisson TH. Efferocytosis of apoptotic alveolar epithelial cells is sufficient to initiate lung fibrosis. Cell Death Dis 2018; 9:1056. [PMID: 30333529 PMCID: PMC6193049 DOI: 10.1038/s41419-018-1074-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 09/14/2018] [Accepted: 09/21/2018] [Indexed: 12/30/2022]
Abstract
Type II alveolar epithelial cell (AEC) apoptosis is a prominent feature of fibrotic lung diseases and animal models of pulmonary fibrosis. While there is growing recognition of the importance of AEC injury and apoptosis as a causal factor in fibrosis, the underlying mechanisms that link these processes remain unknown. We have previously shown that targeting the type II alveolar epithelium for injury by repetitively administering diphtheria toxin to transgenic mice expressing the diphtheria toxin receptor off of the surfactant protein C promoter (SPC-DTR) develop lung fibrosis, confirming that AEC injury is sufficient to cause fibrosis. In the present study, we find that SPC-DTR mice develop increased activation of caspase 3/7 after initiation of diphtheria toxin treatment consistent with apoptosis within AECs. We also find evidence of efferocytosis, the uptake of apoptotic cells, by alveolar macrophages in this model. To determine the importance of efferocytosis in lung fibrosis, we treated cultured alveolar macrophages with apoptotic type II AECs and found that the uptake induced pro-fibrotic gene expression. We also found that the repetitive intrapulmonary administration of apoptotic type II AEC or MLE-12 cells induces lung fibrosis. Finally, mice lacking a key efferocytosis receptor, CD36, developed attenuated fibrosis in response to apoptotic MLE-12 cells. Collectively, these studies support a novel mechanism linking AEC apoptosis with macrophage pro-fibrotic activation via efferocytosis and reveal previously unrecognized therapeutic targets.
Collapse
Affiliation(s)
- Kevin K Kim
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Megan R Dotson
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Manisha Agarwal
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jibing Yang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Patrick B Bradley
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Natalia Subbotina
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John J Osterholzer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Thomas H Sisson
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
13
|
Cong X, Hubmayr RD, Li C, Zhao X. Plasma membrane wounding and repair in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2017; 312:L371-L391. [PMID: 28062486 PMCID: PMC5374305 DOI: 10.1152/ajplung.00486.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Various pathophysiological conditions such as surfactant dysfunction, mechanical ventilation, inflammation, pathogen products, environmental exposures, and gastric acid aspiration stress lung cells, and the compromise of plasma membranes occurs as a result. The mechanisms necessary for cells to repair plasma membrane defects have been extensively investigated in the last two decades, and some of these key repair mechanisms are also shown to occur following lung cell injury. Because it was theorized that lung wounding and repair are involved in the pathogenesis of acute respiratory distress syndrome (ARDS) and idiopathic pulmonary fibrosis (IPF), in this review, we summarized the experimental evidence of lung cell injury in these two devastating syndromes and discuss relevant genetic, physical, and biological injury mechanisms, as well as mechanisms used by lung cells for cell survival and membrane repair. Finally, we discuss relevant signaling pathways that may be activated by chronic or repeated lung cell injury as an extension of our cell injury and repair focus in this review. We hope that a holistic view of injurious stimuli relevant for ARDS and IPF could lead to updated experimental models. In addition, parallel discussion of membrane repair mechanisms in lung cells and injury-activated signaling pathways would encourage research to bridge gaps in current knowledge. Indeed, deep understanding of lung cell wounding and repair, and discovery of relevant repair moieties for lung cells, should inspire the development of new therapies that are likely preventive and broadly effective for targeting injurious pulmonary diseases.
Collapse
Affiliation(s)
- Xiaofei Cong
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Rolf D Hubmayr
- Emerius, Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota; and
| | - Changgong Li
- Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia;
| |
Collapse
|
14
|
Sattler C, Moritz F, Chen S, Steer B, Kutschke D, Irmler M, Beckers J, Eickelberg O, Schmitt-Kopplin P, Adler H, Stoeger T. Nanoparticle exposure reactivates latent herpesvirus and restores a signature of acute infection. Part Fibre Toxicol 2017; 14:2. [PMID: 28069010 PMCID: PMC5223553 DOI: 10.1186/s12989-016-0181-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/15/2016] [Indexed: 02/04/2023] Open
Abstract
Background Inhalation of environmental (nano) particles (NP) as well as persistent herpesvirus-infection are potentially associated with chronic lung disease and as both are omnipresent in human society a coincidence of these two factors is highly likely. We hypothesized that NP-exposure of persistently herpesvirus-infected cells as a second hit might disrupt immune control of viral latency, provoke reactivation of latent virus and eventually lead to an inflammatory response and tissue damage. Results To test this hypothesis, we applied different NP to cells or mice latently infected with murine gammaherpesvirus 68 (MHV-68) which provides a small animal model for the study of gammaherpesvirus-pathogenesis in vitro and in vivo. In vitro, NP-exposure induced expression of the typically lytic viral gene ORF50 and production of lytic virus. In vivo, lytic viral proteins in the lung increased after intratracheal instillation with NP and elevated expression of the viral gene ORF50 could be detected in cells from bronchoalveolar lavage. Gene expression and metabolome analysis of whole lung tissue revealed patterns with striking similarities to acute infection. Likewise, NP-exposure of human cells latently infected with Epstein-Barr-Virus also induced virus production. Conclusions Our results indicate that NP-exposure of persistently herpesvirus-infected cells – murine or human – restores molecular signatures found in acute virus infection, boosts production of lytic viral proteins, and induces an inflammatory response in the lung – a combination which might finally result in tissue damage and pathological alterations. Electronic supplementary material The online version of this article (doi:10.1186/s12989-016-0181-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine Sattler
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Franco Moritz
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Research Unit BioGeoChemistry, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Shanze Chen
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Beatrix Steer
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Marchioninistrasse 25, D-81377, Munich, Germany.,University Hospital Grosshadern, Ludwig-Maximilians-University, D-81377, Munich, Germany.,Comprehensive Pneumology Center, Member of the German Center of Lung Research (DZL), D-81377, Munich, Germany
| | - David Kutschke
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Martin Irmler
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Johannes Beckers
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.,Technische Universität München, Chair of Experimental Genetics, D-85354, Freising, Germany
| | - Oliver Eickelberg
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Research Unit BioGeoChemistry, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Heiko Adler
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Marchioninistrasse 25, D-81377, Munich, Germany. .,University Hospital Grosshadern, Ludwig-Maximilians-University, D-81377, Munich, Germany. .,Comprehensive Pneumology Center, Member of the German Center of Lung Research (DZL), D-81377, Munich, Germany.
| | - Tobias Stoeger
- Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.
| |
Collapse
|
15
|
Viruses in Idiopathic Pulmonary Fibrosis. Etiology and Exacerbation. Ann Am Thorac Soc 2016; 12 Suppl 2:S186-92. [PMID: 26595738 DOI: 10.1513/annalsats.201502-088aw] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Viral infections are important contributors to exacerbation of asthma and chronic obstructive pulmonary disease; however, the role of viruses in the pathogenesis of idiopathic pulmonary fibrosis (IPF) is less clear. This likely reflects that fact that IPF acute exacerbations are defined clinically as "noninfectious," and little attention has been paid to the outcomes of patients with IPF with diagnosed infections. However, accumulating evidence suggests that infections (both bacterial and viral) may influence disease outcomes either as exacerbating agents or initiators of disease. Support for a viral role in disease initiation comes from studies demonstrating the presence of herpesviral DNA and epithelial cell stress in the lungs of asymptomatic relatives at risk for developing familial IPF. In addition, the number of studies that can associate viral (especially herpesviral) signatures in the lung with the development of IPF is steadily growing, and activated leukocyte signatures in patients with IPF provide further support for infectious processes driving IPF progression. Animal modeling has been used to better understand how a gamma herpesvirus infection can modulate the pathogenesis of lung fibrosis and has demonstrated that preceding infections appear to reprogram lung epithelial cells during latency to produce profibrotic factors, making the lung more susceptible to subsequent fibrotic insult, whereas exacerbations of existing fibrosis, or infections in susceptible hosts, involve active viral replication and are influenced by antiviral therapy. In addition, there is new evidence that bacterial burden in the lungs of patients with IPF may predict a poor prognosis.
Collapse
|
16
|
Gurczynski SJ, Procario MC, O'Dwyer DN, Wilke CA, Moore BB. Loss of CCR2 signaling alters leukocyte recruitment and exacerbates γ-herpesvirus-induced pneumonitis and fibrosis following bone marrow transplantation. Am J Physiol Lung Cell Mol Physiol 2016; 311:L611-27. [PMID: 27448666 DOI: 10.1152/ajplung.00193.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/19/2016] [Indexed: 11/22/2022] Open
Abstract
CCR2-expressing leukocytes are required for the progression of fibrosis in models of induced lung injury as well as models of bone marrow transplant (BMT)-related idiopathic pneumonia syndrome. Infection with murid γ-herpesvirus-68 (γHV-68) results in severe pneumonitis and pulmonary fibrosis following syngeneic BMT; however, the roles that various proinflammatory leukocyte populations play in this process remain unclear. Deletion of CCR2 in both non-BMT and BMT mice increased early lytic viral replication and resulted in a reduction in the numbers of lung-infiltrating GR1+,F4/80+ and CXCR1+ cells, while maintaining robust neutrophil infiltration. Similarly, in γHV-68-infected CCR2(-/-) BMT mice, recruitment of monocytes and lymphocytes were reduced whereas neutrophil recruitment was increased compared with wild-type (WT) BMT mice. Interestingly, levels of profibrotic IL-17 were increased in infected CCR2 BMT mice compared with WT BMT. Furthermore, an increase in lung-associated collagen was detected even though there was an overall decrease in the number of profibrotic CCR2+ fibrocytes detected in the lungs of CCR2(-/-) BMT mice. These data indicate that, contrary to most models of fibrosis, deletion of CCR2 offers no protection from γ-herpesvirus-induced pneumonitis and fibrosis, and, indeed, CCR2+ cells play a suppressive role during the development of pulmonary fibrosis following γ-herpesvirus infection post-BMT by limiting IL-7 and collagen production.
Collapse
Affiliation(s)
- Stephen J Gurczynski
- Department of Internal Medicine, Pulmonary and Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan;
| | - Megan C Procario
- Graduate Program in Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan; and
| | - David N O'Dwyer
- Department of Internal Medicine, Pulmonary and Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan
| | - Carol A Wilke
- Department of Internal Medicine, Pulmonary and Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan
| | - Bethany B Moore
- Department of Internal Medicine, Pulmonary and Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
17
|
O’Flaherty BM, Matar CG, Wakeman BS, Garcia A, Wilke CA, Courtney CL, Moore BB, Speck SH. CD8+ T Cell Response to Gammaherpesvirus Infection Mediates Inflammation and Fibrosis in Interferon Gamma Receptor-Deficient Mice. PLoS One 2015; 10:e0135719. [PMID: 26317335 PMCID: PMC4552722 DOI: 10.1371/journal.pone.0135719] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/24/2015] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), one of the most severe interstitial lung diseases, is a progressive fibrotic disorder of unknown etiology. However, there is growing appreciation for the role of viral infection in disease induction and/or progression. A small animal model of multi-organ fibrosis, which involves murine gammaherpesvirus (MHV68) infection of interferon gamma receptor deficient (IFNγR-/-) mice, has been utilized to model the association of gammaherpesvirus infections and lung fibrosis. Notably, several MHV68 mutants which fail to induce fibrosis have been identified. Our current study aimed to better define the role of the unique MHV68 gene, M1, in development of pulmonary fibrosis. We have previously shown that the M1 gene encodes a secreted protein which possesses superantigen-like function to drive the expansion and activation of Vβ4+ CD8+ T cells. Here we show that M1-dependent fibrosis is correlated with heightened levels of inflammation in the lung. We observe an M1-dependent cellular infiltrate of innate immune cells with most striking differences at 28 days-post infection. Furthermore, in the absence of M1 protein expression we observed reduced CD8+ T cells and MHV68 epitope specific CD8+ T cells to the lungs-despite equivalent levels of viral replication between M1 null and wild type MHV68. Notably, backcrossing the IFNγR-/- onto the Balb/c background, which has previously been shown to exhibit weak MHV68-driven Vβ4+ CD8+ T cell expansion, eliminated MHV68-induced fibrosis-further implicating the activated Vβ4+ CD8+ T cell population in the induction of fibrosis. We further addressed the role that CD8+ T cells play in the induction of fibrosis by depleting CD8+ T cells, which protected the mice from fibrotic disease. Taken together these findings are consistent with the hypothesized role of Vβ4+ CD8+ T cells as mediators of fibrotic disease in IFNγR-/- mice.
Collapse
Affiliation(s)
- Brigid M. O’Flaherty
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Caline G. Matar
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Brian S. Wakeman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - AnaPatricia Garcia
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
| | - Carol A. Wilke
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Cynthia L. Courtney
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
| | - Bethany B. Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Samuel H. Speck
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
18
|
Smoktunowicz N, Alexander RE, Franklin L, Williams AE, Holman B, Mercer PF, Jarai G, Scotton CJ, Chambers RC. The anti-fibrotic effect of inhibition of TGFβ-ALK5 signalling in experimental pulmonary fibrosis in mice is attenuated in the presence of concurrent γ-herpesvirus infection. Dis Model Mech 2015; 8:1129-39. [PMID: 26138704 PMCID: PMC4582104 DOI: 10.1242/dmm.019984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/26/2015] [Indexed: 02/06/2023] Open
Abstract
TGFβ-ALK5 pro-fibrotic signalling and herpesvirus infections have been implicated in the pathogenesis and exacerbation of pulmonary fibrosis. In this study we addressed the role of TGFβ-ALK5 signalling during the progression of fibrosis in a two-hit mouse model of murine γ-herpesvirus 68 (MHV-68) infection on the background of pre-existing bleomycin-induced pulmonary fibrosis. Assessment of total lung collagen levels in combination with ex vivo micro-computed tomography (µCT) analysis of whole lungs demonstrated that MHV-68 infection did not enhance lung collagen deposition in this two-hit model but led to a persistent and exacerbated inflammatory response. Moreover, µCT reconstruction and analysis of the two-hit model revealed distinguishing features of diffuse ground-glass opacities and consolidation superimposed on pre-existing fibrosis that were reminiscent of those observed in acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF). Virally-infected murine fibrotic lungs further displayed evidence of extensive inflammatory cell infiltration and increased levels of CCL2, TNFα, IL-1β and IL-10. Blockade of TGFβ-ALK5 signalling attenuated lung collagen accumulation in bleomycin-alone injured mice, but this anti-fibrotic effect was reduced in the presence of concomitant viral infection. In contrast, inhibition of TGFβ-ALK5 signalling in virally-infected fibrotic lungs was associated with reduced inflammatory cell aggregates and increased levels of the antiviral cytokine IFNγ. These data reveal newly identified intricacies for the TGFβ-ALK5 signalling axis in experimental lung fibrosis, with different outcomes in response to ALK5 inhibition depending on the presence of viral infection. These findings raise important considerations for the targeting of TGFβ signalling responses in the context of pulmonary fibrosis.
Collapse
Affiliation(s)
- Natalia Smoktunowicz
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Robert E Alexander
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Linda Franklin
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Andrew E Williams
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Beverley Holman
- Institute of Nuclear Medicine, University College London, NW1 2BU, UK
| | - Paul F Mercer
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Gabor Jarai
- Novartis Institutes of Biomedical Research, Horsham, RH12 5AB, UK
| | - Chris J Scotton
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| | - Rachel C Chambers
- Centre for Inflammation & Tissue Repair, University College London, London, WC1E 6JF, UK
| |
Collapse
|
19
|
Moore BB. Following the path of CCL2 from prostaglandins to periostin in lung fibrosis. Am J Respir Cell Mol Biol 2014; 50:848-52. [PMID: 24605795 DOI: 10.1165/rcmb.2014-0075ps] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Without question, the greatest and most humbling honor of my scientific career was to learn that I was nominated for the American Thoracic Society Recognition Award for Scientific Accomplishments. On the occasion of this award, as I look back on the progress made in the last 15 years, I am pleased by the scientific insights; however, I am also saddened that we still have no internationally recognized efficacious therapy. This perspective will highlight the areas my laboratory has addressed regarding the pathogenesis of idiopathic pulmonary fibrosis in hopes of identifying new therapeutic targets.
Collapse
Affiliation(s)
- Bethany B Moore
- Departments of Internal Medicine and Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
20
|
Ashley SL, Jegal Y, Moore TA, van Dyk LF, Laouar Y, Moore BB. γ-Herpes virus-68, but not Pseudomonas aeruginosa or influenza A (H1N1), exacerbates established murine lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2014; 307:L219-30. [PMID: 24879051 DOI: 10.1152/ajplung.00300.2013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Patients with idiopathic pulmonary fibrosis (IPF) often do worse following infection, but the cause of the decline is not fully understood. We previously demonstrated that infection with a murine gamma herpes virus (γHV-68) could exacerbate established lung fibrosis following administration of fluorescein isothiocyanate (McMillan et al. Am J Respir Crit Care Med 177: 771-780, 2008). In the present study, we anesthetized mice and injected saline or bleomycin intratracheally on day 0. On day 14, mice were anesthetized again and infected with either a Gram-negative bacteria (Pseudomonas aeruginosa), or with H1N1 or γHV-68 viruses. Measurements were then made on days 15, 21, or 35. We demonstrate that infection with P. aeruginosa does not exacerbate extracellular matrix deposition post-bleomycin. Furthermore, fibrotic mice are effectively able to clear P. aeruginosa infection. In contrast, bleomycin-treated mice develop worse lung fibrosis when infected with γHV-68, but not when infected with H1N1. The differential ability of γHV-68 to cause increased collagen deposition could not be explained by differences in inflammatory cell recruitment or whole lung chemokine and cytokine responses. Alveolar epithelial cells from γHV-68-infected mice displayed increased expression of TGFβ receptor 1, increased SMAD3 phosphorylation, and evidence of apoptosis measured by cleaved poly-ADP ribose polymerase (PARP). The ability of γHV-68 to augment fibrosis required the ability of the virus to reactivate from latency. This property appears unique to γHV-68, as the β-herpes virus, cytomegalovirus, did not have the same effect.
Collapse
Affiliation(s)
- Shanna L Ashley
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Yangjin Jegal
- Department of Internal Medicine, Ulsan University Hospital, College of Medicine, University of Ulsan, Ulsan, Korea
| | - Thomas A Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan;
| | - Linda F van Dyk
- Departments of Microbiology and Immunology, University of Colorado, Denver, Colorado; and
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Bethany B Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
21
|
Abstract
Progressive lung fibrosis in humans, typified by idiopathic pulmonary fibrosis (IPF), is a serious cause of morbidity and mortality in people. Similar diseases have been described in dogs, cats, and horses. The cause and pathogenesis of such diseases in all species is poorly understood. There is growing evidence in human medicine that IPF is a manifestation of abnormal wound repair in response to epithelial injury. Because viruses can contribute to epithelial injury, there is increasing interest in a possible role of viruses, particularly gammaherpesviruses, in the pathogenesis of pulmonary fibrosis. This review provides background information on progressive fibrosing lung disease in human and veterinary medicine and summarizes the evidence for an association between gammaherpesvirus infection and pulmonary fibrosis, especially Epstein-Barr virus in human pulmonary fibrosis, and equine herpesvirus 5 in equine multinodular pulmonary fibrosis. Data derived from experimental lung infection in mice with the gammaherpesvirus murine herpesvirus are presented, emphasizing the host and viral factors that may contribute to lung fibrosis. The experimental data are considered in the context of the pathogenesis of naturally occurring pulmonary fibrosis in humans and horses.
Collapse
Affiliation(s)
- K. J. Williams
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
22
|
Experimental induction of pulmonary fibrosis in horses with the gammaherpesvirus equine herpesvirus 5. PLoS One 2013; 8:e77754. [PMID: 24147074 PMCID: PMC3795644 DOI: 10.1371/journal.pone.0077754] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 09/09/2013] [Indexed: 11/19/2022] Open
Abstract
Gammaherpesviruses (γHV) are implicated in the pathogenesis of pulmonary fibrosis in humans and murine models of lung fibrosis, however there is little direct experimental evidence that such viruses induce lung fibrosis in the natural host. The equine γHV EHV 5 is associated with equine multinodular pulmonary fibrosis (EMPF), a progressive fibrosing lung disease in its natural host, the horse. Experimental reproduction of EMPF has not been attempted to date. We hypothesized that inoculation of EHV 5 isolated from cases of EMPF into the lungs of clinically normal horses would induce lung fibrosis similar to EMPF. Neutralizing antibody titers were measured in the horses before and after inoculation with EHV 5. PCR and virus isolation was used to detect EHV 5 in antemortem blood and BAL samples, and in tissues collected postmortem. Nodular pulmonary fibrosis and induction of myofibroblasts occurred in EHV 5 inoculated horses. Mean lung collagen in EHV 5 inoculated horses (80 µg/mg) was significantly increased compared to control horses (26 µg/mg) (p < 0.5), as was interstitial collagen (32.6% ± 1.2% vs 23% ± 1.4%) (mean ± SEM; p < 0.001). Virus was difficult to detect in infected horses throughout the experiment, although EHV 5 antigen was detected in the lung by immunohistochemistry. We conclude that the γHV EHV 5 can induce lung fibrosis in the horse, and hypothesize that induction of fibrosis occurs while the virus is latent within the lung. This is the first example of a γHV inducing lung fibrosis in the natural host.
Collapse
|
23
|
Farina A, Cirone M, York M, Lenna S, Padilla C, Mclaughlin S, Faggioni A, Lafyatis R, Trojanowska M, Farina GA. Epstein-Barr virus infection induces aberrant TLR activation pathway and fibroblast-myofibroblast conversion in scleroderma. J Invest Dermatol 2013; 134:954-964. [PMID: 24129067 PMCID: PMC3961515 DOI: 10.1038/jid.2013.423] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 12/28/2022]
Abstract
Scleroderma (SSc) is a complex and heterogeneous connective tissue disease mainly characterized by autoimmunity, vascular damage, and fibrosis that mostly involve the skin and lungs. Epstein–Barr virus (EBV) is a lymphotropic γ-herpesvirus that has co-evolved with human species, infecting >95% of the adult population worldwide, and has been a leading candidate in triggering several autoimmune diseases. Here we show that EBV establishes infection in the majority of fibroblasts and endothelial cells in the skin of SSc patients, characterized by the expression of the EBV noncoding small RNAs (EBERs) and the increased expression of immediate-early lytic and latency mRNAs and proteins. We report that EBV is able to persistently infect human SSc fibroblasts in vitro, inducing an aberrant innate immune response in infected cells. EBV–Toll-like receptor (TLR) aberrant activation induces the expression of selected IFN-regulatory factors (IRFs), IFN-stimulated genes (ISGs), transforming growth factor-β1 (TGFβ1), and several markers of fibroblast activation, such as smooth muscle actin and Endothelin-1, and all of these genes play a key role in determining the profibrotic phenotype in SSc fibroblasts. These findings imply that EBV infection occurring in mesenchymal, endothelial, and immune cells of SSc patients may underlie the main pathological features of SSc including autoimmunity, vasculopathy, and fibrosis, and provide a unified disease mechanism represented by EBV reactivation.
Collapse
Affiliation(s)
- Antonella Farina
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA; Institute Pasteur-Fondazione Cenci Bolognetti, Department of Experimental Medicine, University of Rome Sapienza, Rome, Italy
| | - Mara Cirone
- Institute Pasteur-Fondazione Cenci Bolognetti, Department of Experimental Medicine, University of Rome Sapienza, Rome, Italy
| | - Michael York
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Stefania Lenna
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Cristina Padilla
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sarah Mclaughlin
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Alberto Faggioni
- Institute Pasteur-Fondazione Cenci Bolognetti, Department of Experimental Medicine, University of Rome Sapienza, Rome, Italy
| | - Robert Lafyatis
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Maria Trojanowska
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Giuseppina A Farina
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Adamali HI, Maher TM. Current and novel drug therapies for idiopathic pulmonary fibrosis. Drug Des Devel Ther 2012; 6:261-72. [PMID: 23055696 PMCID: PMC3463380 DOI: 10.2147/dddt.s29928] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Indexed: 02/04/2023] Open
Abstract
Over the past decade, there has been a cohesive effort from patients, physicians, clinical and basic scientists, and the pharmaceutical industry to find definitive treatments for idiopathic pulmonary fibrosis (IPF). As understanding of disease behavior and pathogenesis has improved, the aims of those treating IPF have shifted from reversing the disease to slowing or preventing progression of this chronic fibrotic illness. It is to be hoped that by slowing disease progression, survival will be improved from the current dismal median of 3.5 years following diagnosis. In Europe and Asia, a milestone has recently been reached with the licensing of the first IPF-specific drug, pirfenidone. This review assesses the current treatment modalities available for IPF, including pirfenidone. It also turns an eye to the future and discusses the growing number of promising compounds currently in development that it is hoped, in time, will make their way into the clinic as treatments for IPF.
Collapse
Affiliation(s)
| | - Toby M Maher
- Interstitial Lung Disease Unit, Royal Brompton Hospital, London, UK
| |
Collapse
|
25
|
Naik PN, Horowitz JC, Moore TA, Wilke CA, Toews GB, Moore BB. Pulmonary fibrosis induced by γ-herpesvirus in aged mice is associated with increased fibroblast responsiveness to transforming growth factor-β. J Gerontol A Biol Sci Med Sci 2011; 67:714-25. [PMID: 22193547 DOI: 10.1093/gerona/glr211] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Young (4 month) and aged (15-18 months) mice were given intranasal saline or γ--herpesvirus-68 infection. After 21 days, aged, but not young mice, showed significant increases in collagen content and fibrosis. There were no differences in viral clearance or inflammatory cells (including fibrocytes) between infected aged and young mice. Enzyme-linked immunosorbent assays showed increased transforming growth factor-β in whole lung homogenates of infected aged mice compared with young mice. When fibroblasts from aged and young mice were infected in vitro, aged, but not young, fibroblasts upregulate alpha-smooth muscle actin and collagen I protein. Infection with virus in vivo also demonstrates increased alpha-smooth muscle actin and collagen I protein and collagen I, collagen III, and fibronectin messenger RNA in aged fibroblasts. Furthermore, evaluation revealed that aged fibroblasts at baseline have increased transforming growth factor-β receptor 1 and 2 levels compared with young fibroblasts and are resistant to apoptosis. Increased responsiveness to transforming growth factor-β was verified by increased collagen III and fibronectin messenger RNA after treatment in vitro with transforming growth factor-β.
Collapse
Affiliation(s)
- Payal N Naik
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | |
Collapse
|
26
|
Kropski JA, Lawson WE, Blackwell TS. Right place, right time: the evolving role of herpesvirus infection as a "second hit" in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2011; 302:L441-4. [PMID: 22180659 DOI: 10.1152/ajplung.00335.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Over the course of the past decade, increasing evidence has implicated alveolar epithelial cell injury and dysfunction in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Genetic factors, cigarette smoking, and other environmental exposures have been identified as potential factors leading to a population of vulnerable alveolar epithelial cells. In addition, molecular techniques have demonstrated herpesviruses are commonly detectable in the lungs of patients with IPF, raising suspicion that, in the setting of a vulnerable alveolar epithelium, lytic (or latent) herpesvirus infection may act as a "second hit" leading to the development of pulmonary fibrosis. Intriguingly, in vivo modeling has shown that herpesvirus infection induces or worsens lung fibrosis when combined with immunodeficiency or other injurious stimuli. Here, we discuss potential mechanisms through which herpesvirus infection may contribute to the pathogenesis of IPF. Ultimately, antiviral therapy may hold promise for halting the progression of this deadly disease.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | |
Collapse
|
27
|
|
28
|
Coomes SM, Farmen S, Wilke CA, Laouar Y, Moore BB. Severe gammaherpesvirus-induced pneumonitis and fibrosis in syngeneic bone marrow transplant mice is related to effects of transforming growth factor-β. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2382-96. [PMID: 21924228 DOI: 10.1016/j.ajpath.2011.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 06/29/2011] [Accepted: 08/01/2011] [Indexed: 11/17/2022]
Abstract
Pulmonary infections and pneumonitis occur frequently after hematopoietic stem cell transplantation. Using a syngeneic mouse model of bone marrow transplantation (BMT), we have previously demonstrated that BMT mice are more susceptible to acute gammaherpesvirus 68 (MHV-68) replication at day 7 after infection. By day 21, the virus is latent in lungs of BMT and control mice, and there is no difference in viral load. Despite similar latent viral load, BMT mice develop severe pneumonitis associated with reduced oxygen saturation, fibrosis, peripheral inflammation, hyaline membranes, and foamy alveolar macrophages, a phenotype that persists for 7 weeks after infection. BMT mice demonstrate increased bronchoalveolar lavage (BAL) cells, and this population is enriched in neutrophils and T cells. Alternatively, activated macrophages appear earlier than do classically activated macrophages. BAL fluid from BMT mice at day 21 after infection contains increased levels of hydrogen peroxide, nitrite, and transforming growth factor-β (TGF-β). Mice expressing the dominant-negative transgene dn-TGFβRII in multiple cell types were used as BMT donors. BMT mice with T-cell dnTGFβRII are largely protected from the pneumonitis phenotype, whereas mice with CD11c-dnTGFβRII BMT mice are only modestly protected from pneumonitis. Protection in BMT mice with T-cell dnTGFβRII is associated with decreased TGF-β derived from parenchymal cells in the BAL fluid, lower nitrite levels, and reduced apoptosis, whereas alternatively activated macrophage markers are unchanged.
Collapse
Affiliation(s)
- Stephanie M Coomes
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | |
Collapse
|
29
|
TLR9-induced interferon β is associated with protection from gammaherpesvirus-induced exacerbation of lung fibrosis. FIBROGENESIS & TISSUE REPAIR 2011; 4:18. [PMID: 21810214 PMCID: PMC3163187 DOI: 10.1186/1755-1536-4-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 08/02/2011] [Indexed: 11/10/2022]
Abstract
Background We have shown previously that murine gammaherpesvirus 68 (γHV68) infection exacerbates established pulmonary fibrosis. Because Toll-like receptor (TLR)-9 may be important in controlling the immune response to γHV68 infection, we examined how TLR-9 signaling effects exacerbation of fibrosis in response to viral infection, using models of bleomycin- and fluorescein isothiocyanate-induced pulmonary fibrosis in wild-type (Balb/c) and TLR-9-/- mice. Results We found that in the absence of TLR-9 signaling, there was a significant increase in collagen deposition following viral exacerbation of fibrosis. This was not associated with increased viral load in TLR-9-/- mice or with major alterations in T helper (Th)1 and Th2 cytokines. We examined alveolar epithelial-cell apoptosis in both strains, but this could not explain the altered fibrotic outcomes. As expected, TLR-9-/- mice had a defect in the production of interferon (IFN)-β after viral infection. Balb/c fibroblasts infected with γHV68 in vitro produced more IFN-β than did infected TLR-9-/- fibroblasts. Accordingly, in vitro infection of Balb/c fibroblasts resulted in reduced proliferation rates whereas infection of TLR-9-/- fibroblasts did not. Finally, therapeutic administration of CpG oligodeoxynucleotides ameliorated bleomycin-induced fibrosis in wild-type mice. Conclusions These results show a protective role for TLR-9 signaling in murine models of lung fibrosis, and highlight differences in the biology of TLR-9 between mice and humans.
Collapse
|