1
|
Mota PC, Soares ML, Ferreira AC, Santos RF, Rufo JC, Vasconcelos D, Carvalho A, Guimarães S, Vasques-Nóvoa F, Cardoso C, Melo N, Alexandre AT, Coelho D, Novais-Bastos H, Morais A. Polymorphisms and haplotypes of TOLLIP and MUC5B are associated with susceptibility and survival in patients with fibrotic hypersensitivity pneumonitis. Pulmonology 2025; 31:2416788. [PMID: 38309995 DOI: 10.1016/j.pulmoe.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Hypersensitivity pneumonitis (HP) is an interstitial lung disease with diverse clinical features that can present a fibrotic phenotype similar to idiopathic pulmonary fibrosis (IPF) in genetically predisposed individuals. While several single nucleotide polymorphisms (SNPs) have been associated with IPF, the genetic factors contributing to fibrotic HP (fHP) remain poorly understood. This study investigated the association of MUC5B and TOLLIP variants with susceptibility, clinical presentation and survival in Portuguese patients with fHP. MATERIAL AND METHODS A case-control study was undertaken with 97 fHP patients and 112 controls. Six SNPs residing in the MUC5B and TOLLIP genes and their haplotypes were analyzed. Associations with risk, survival, and clinical, radiographic, and pathological features of fHP were probed through comparisons among patients and controls. RESULTS MUC5B rs35705950 and three neighboring TOLLIP variants (rs3750920, rs111521887, and rs5743894) were associated with increased susceptibility to fHP. Minor allele frequencies were greater among fHP patients than in controls (40.7% vs 12.1%, P<0.0001; 52.6% vs 40.2%, P = 0.011; 22.7% vs 13.4%, P = 0.013; and 23.2% vs 12.9%, P = 0.006, respectively). Haplotypes formed by these variants were also linked to fHP susceptibility. Moreover, carriers of a specific haplotype (G-T-G-C) had a significant decrease in survival (adjusted hazard ratio 6.92, 95% CI 1.73-27.64, P = 0.006). Additional associations were found between TOLLIP rs111521887 and rs5743894 variants and decreased lung function at baseline, and the MUC5B SNP and radiographic features, further highlighting the influence of genetic factors in fHP. CONCLUSION These findings suggest that TOLLIP and MUC5B variants and haplotypes may serve as valuable tools for risk assessment and prognosis in fibrotic hypersensitivity pneumonitis, potentially contributing to its patient stratification, and offer insights into the genetic factors influencing the clinical course of the condition.
Collapse
Affiliation(s)
- P C Mota
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - M L Soares
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- LAIMM, Núcleo de Recursos Laboratoriais, Unidade de Gestão de Conhecimento, Departamento de Recursos Comuns, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A C Ferreira
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - R F Santos
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Escola Superior de Saúde - Instituto Politécnico do Porto, Portugal
| | - J C Rufo
- Indoor Air Quality and Respiratory Health Lab, Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research (T.Bio), Escola Superior de Saúde, Instituto Politécnico do Porto, Porto, Portugal
| | - D Vasconcelos
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A Carvalho
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Radiologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - S Guimarães
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Anatomia Patológica, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - F Vasques-Nóvoa
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Medicina Interna, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Portugal
| | - C Cardoso
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - N Melo
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - A T Alexandre
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - D Coelho
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - H Novais-Bastos
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A Morais
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Cerri S, Manzini E, Nori O, Pacchetti L, Rossi L, Turchiano MG, Samarelli AV, Raineri G, Andrisani D, Gozzi F, Beghè B, Clini E, Tonelli R. Genetic Risk Factors in Idiopathic and Non-Idiopathic Interstitial Lung Disease: Similarities and Differences. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1967. [PMID: 39768847 PMCID: PMC11677115 DOI: 10.3390/medicina60121967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Recent advances in genetics and epigenetics have provided critical insights into the pathogenesis of both idiopathic and non-idiopathic interstitial lung diseases (ILDs). Mutations in telomere-related genes and surfactant proteins have been linked to familial pulmonary fibrosis, while variants in MUC5B and TOLLIP increase the risk of ILD, including idiopathic pulmonary fibrosis and rheumatoid arthritis-associated ILD. Epigenetic mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs such as miR-21 and miR-29, regulate fibrotic pathways, influencing disease onset and progression. Although no standardized genetic panel for ILD exists, understanding the interplay of genetic mutations and epigenetic alterations could aid in the development of personalized therapeutic approaches. This review highlights the genetic and epigenetic factors driving ILD, emphasizing their potential for refining diagnosis and treatment.
Collapse
Affiliation(s)
- Stefania Cerri
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Elisa Manzini
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Respiratory Disease Unit, Hospital of Sassuolo, 41049 Sassuolo, Italy
| | - Ottavia Nori
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- U.O. Pneumologia, Presidio Ospedaliero di Arco, APSS Provincia Autonoma di Trento, 38062 Trento, Italy
| | - Lucia Pacchetti
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Division of Pneumology, MultiMedica IRCCS, 20099 Milan, Italy
| | - Laura Rossi
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Respiratory Disease Unit, Arcispedale Santa Maria Nuova, 42123 Reggio Emilia, Italy
| | - Maria Giulia Turchiano
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
| | - Anna Valeria Samarelli
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Giulia Raineri
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Dario Andrisani
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Filippo Gozzi
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Bianca Beghè
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
| | - Enrico Clini
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Roberto Tonelli
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| |
Collapse
|
3
|
Han J, Song JW. Dyskeratosis congenita with heterozygous RTEL1 mutations presenting with fibrotic hypersensitivity pneumonitis. Respir Med Case Rep 2023; 42:101810. [PMID: 36655009 PMCID: PMC9841051 DOI: 10.1016/j.rmcr.2023.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/01/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023] Open
Abstract
Dyskeratosis congenita is a rare genetic disorder of telomere insufficiency characterized by a mucocutaneous triad of nail dystrophy, abnormal skin pigmentation, and mucosal leukoplakia. Early diagnosis is important for multidisciplinary approach to its complications including bone marrow failure, malignancy, interstitial lung disease, and liver disease which cause significant morbidity and mortality. We report a genetically confirmed case of dyskeratosis congenita who presented with fibrotic hypersensitivity pneumonitis, highlighting non-mucocutaneous features of dyskeratosis congenita and the need to consider genetic predisposition in a patient with interstitial lung disease and combined unusual manifestations.
Collapse
Affiliation(s)
- Jinhee Han
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea,Corresponding author. Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
4
|
Abstract
Childhood interstitial lung disease (ChILD) is an umbrella term encompassing a diverse group of diffuse lung diseases affecting infants and children. Although the timely and accurate diagnosis of ChILD is often challenging, it is optimally achieved through the multidisciplinary integration of imaging findings with clinical data, genetics, and potentially lung biopsy. This article reviews the definition and classification of ChILD; the role of imaging, pathology, and genetics in ChILD diagnosis; treatment options; and future goals. In addition, a practical approach to ChILD imaging based on the latest available research and the characteristic imaging appearance of ChILD entities are presented.
Collapse
|
5
|
Kellogg DL, Kellogg DL, Musi N, Nambiar AM. Cellular Senescence in Idiopathic Pulmonary Fibrosis. CURRENT MOLECULAR BIOLOGY REPORTS 2021; 7:31-40. [PMID: 34401216 PMCID: PMC8358258 DOI: 10.1007/s40610-021-00145-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/28/2022]
Abstract
Cellular senescence (CS) is increasingly implicated in the etiology of age-related diseases. While CS can facilitate physiological processes such as tissue repair and wound healing, senescent cells also contribute to pathophysiological processes involving macromolecular damage and metabolic dysregulation that characterize multiple morbid and prevalent diseases, including Alzheimer's disease, osteoarthritis, atherosclerotic vascular disease, diabetes mellitus, and idiopathic pulmonary fibrosis (IPF). Preclinical studies targeting senescent cells and the senescence-associated secretory phenotype (SASP) with "senotherapeutics" have demonstrated improvement in age-related morbidity associated with these disease states. Despite promising results from these preclinical trials, few human clinical trials have been conducted. A first-in-human, open-label, pilot study of the senolytic combination of dasatinib and quercetin (DQ) in patients with IPF showed improved physical function and mobility. In this review, we will discuss our current understanding of cellular senescence, its role in age-associated diseases, with a specific focus on IPF, and potential for senotherapeutics in the treatment of fibrotic lung diseases.
Collapse
Affiliation(s)
- D L Kellogg
- University of Texas Health San Antonio, San Antonio, USA
| | - D L Kellogg
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| | - N Musi
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| | - A M Nambiar
- University of Texas Health San Antonio, San Antonio, USA
- South Texas Veterans Health Care System, San Antonio, TX USA
| |
Collapse
|
6
|
Spagnolo P, Distler O, Ryerson CJ, Tzouvelekis A, Lee JS, Bonella F, Bouros D, Hoffmann-Vold AM, Crestani B, Matteson EL. Mechanisms of progressive fibrosis in connective tissue disease (CTD)-associated interstitial lung diseases (ILDs). Ann Rheum Dis 2021; 80:143-150. [PMID: 33037004 PMCID: PMC7815631 DOI: 10.1136/annrheumdis-2020-217230] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
Abstract
Interstitial lung diseases (ILDs), which can arise from a broad spectrum of distinct aetiologies, can manifest as a pulmonary complication of an underlying autoimmune and connective tissue disease (CTD-ILD), such as rheumatoid arthritis-ILD and systemic sclerosis (SSc-ILD). Patients with clinically distinct ILDs, whether CTD-related or not, can exhibit a pattern of common clinical disease behaviour (declining lung function, worsening respiratory symptoms and higher mortality), attributable to progressive fibrosis in the lungs. In recent years, the tyrosine kinase inhibitor nintedanib has demonstrated efficacy and safety in idiopathic pulmonary fibrosis (IPF), SSc-ILD and a broad range of other fibrosing ILDs with a progressive phenotype, including those associated with CTDs. Data from phase II studies also suggest that pirfenidone, which has a different-yet largely unknown-mechanism of action, may also have activity in other fibrosing ILDs with a progressive phenotype, in addition to its known efficacy in IPF. Collectively, these studies add weight to the hypothesis that, irrespective of the original clinical diagnosis of ILD, a progressive fibrosing phenotype may arise from common, underlying pathophysiological mechanisms of fibrosis involving pathways associated with the targets of nintedanib and, potentially, pirfenidone. However, despite the early proof of concept provided by these clinical studies, very little is known about the mechanistic commonalities and differences between ILDs with a progressive phenotype. In this review, we explore the biological and genetic mechanisms that drive fibrosis, and identify the missing evidence needed to provide the rationale for further studies that use the progressive phenotype as a target population.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova School of Medicine and Surgery, Padova, Italy
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Christopher J Ryerson
- Department of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Argyris Tzouvelekis
- Department of Respiratory and Internal Medicine, University of Patras Faculty of Medicine, Patras, Greece
| | - Joyce S Lee
- School of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Disease Unit, University of Duisburg-Essen, Ruhrlandklinik, Essen, Germany
| | - Demosthenes Bouros
- Department of Pneumonology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Bruno Crestani
- Inserm U1152, Université de Paris, F-75018, Paris, France
- Department of Pneumonology, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, F-75018, Paris, France
| | - Eric L Matteson
- Division of Rheumatology and Department of Health Sciences Research, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Zhang F, Yang T, Liu Z, Jia X, Yang L, Wu L, Tang L. Clinical Features of Hypersensitivity Pneumonitis in Children: A Single Center Study. Front Pediatr 2021; 9:789183. [PMID: 35127590 PMCID: PMC8811457 DOI: 10.3389/fped.2021.789183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Hypersensitivity pneumonia (HP) is an interstitial lung disease (ILD) mainly involving small airways and lung parenchyma that is caused by the inhalation of antigens in susceptible people to stimulate the body's immune response. METHODS A total of 6 Chinese children with HP treated in our center from July 2017 to July 2021 were included in our study. RESULTS Among the children, there were 4 males and 2 females, ranging in age from 4 to 14 years. Three cases had chest tightness and shortness of breath, 2 cases had cough, 1 case had chest pain, and 1 case had fever. Two cases of children had a history of close contact with pet dogs, 1 case had a history of contact with pigeons, 2 cases lived in a moldy house recently, and 1 case recently played a saxophone that had been idle for more than 2 years. The parents of two cases also had similar symptoms recently. The specific signs of chest HRCT of 6 cases all were in line with the characteristics of HP. After avoiding the sensitization environment, 2 children quickly recovered, 4 patients received low-dose glucocorticoid oral treatment, and after symptom control the dose was gradually reduced. The course of treatment was about 3-6 months. CONCLUSIONS Exposure to a potential antigen has been found in all 6 HP children. The clinical manifestations are heterogeneous and easy to confuse with other diseases. A clear history of exposure to the antigens, respiratory symptoms associated with HP, signs of HP on HRCT, and improvement after removal from the antigenic environment constitute the cornerstone of the diagnosis of HP children in our unit. Avoiding exposure to antigenic environment is the first step in treatment, and glucocorticoid use is necessary in children with persistent symptoms.
Collapse
Affiliation(s)
- Feizhou Zhang
- Department of Pulmonology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| | - Tongyu Yang
- Department of Pulmonology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| | | | - Xuan Jia
- Department of Radiology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| | - Li Yang
- Department of Radiology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| | - Lei Wu
- Department of Pulmonology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| | - Lanfang Tang
- Department of Pulmonology, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, The Children's Hospital, Hangzhou, China
| |
Collapse
|
8
|
Sanchez-Gonzalez MA, Moskowitz D, Issuree PD, Yatzkan G, Rizvi SAA, Day K. A Pathophysiological Perspective on COVID-19's Lethal Complication: From Viremia to Hypersensitivity Pneumonitis-like Immune Dysregulation. Infect Chemother 2020; 52:335-344. [PMID: 32537960 PMCID: PMC7533209 DOI: 10.3947/ic.2020.52.3.335] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/19/2020] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus responsible for our recent coronavirus disease 2019 pandemic, is driving a lung immunopathology that strongly resembles a severe form of hypersensitivity pneumonitis (HP). A review of recent Severe acute respiratory syndrome-related coronavirus (SARS-CoV) and SARS-CoV-2 medical reports, as well as described characteristics of HP, lead us to postulate a theory for SARS-CoV-2 severe disease. We propose that the novel SARS-CoV-2 can act as a trigger and substrate of an HP-like severe immune reaction especially in genetically vulnerable individuals in addition to those with immune senescence and dysregulation. Accordingly, the purpose of our letter is to shift the emphasis of concern surrounding immune activity from viral infection to an HP-like severe immune reaction. We review similarities in disease presentation between infection and allergy, relevant immunopathology, and outline phases of SARS-CoV-2 disease with perspectives on therapy and critical care. Altogether, the favored course is to begin treatments that address the disease at the earliest phase before immune dysregulation leading to uncontrolled pulmonary inflammation.
Collapse
Affiliation(s)
| | | | - Priya D Issuree
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - George Yatzkan
- Intensive & Critical Care Unit, Larkin Health System, South Miami, FL, USA
| | - Syed A A Rizvi
- Division of Clinical & Translational Research, Larkin Health System, South Miami, Florida, USA
| | - Kenneth Day
- Zymo Research Corporation, Irvine, California, USA
| |
Collapse
|
9
|
Boutros J, Benzaquen J, Delin M, Padovani B, Marquette CH, Leroy S. Exuberant cystic destruction of lung parenchyma. Respir Med Res 2020; 78:100755. [PMID: 32473555 DOI: 10.1016/j.resmer.2020.100755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 10/24/2022]
Affiliation(s)
- J Boutros
- FHU OncoAge, department of pulmonary medicine, université Côte d'Azur, CHU de Nice, Nice, France.
| | - J Benzaquen
- FHU OncoAge, department of pulmonary medicine, université Côte d'Azur, CHU de Nice, Nice, France; FHU OncoAge, CNRS UMR7284, Inserm U1081, Institute of research on cancer and ageing (IRCAN), université Côte d'Azur, Nice, France
| | - M Delin
- FHU OncoAge, department of pulmonary medicine, université Côte d'Azur, CHU de Nice, Nice, France
| | - B Padovani
- Department of radiology, université Côte d'Azur, CHU de Nice, Nice, France
| | - C-H Marquette
- FHU OncoAge, department of pulmonary medicine, université Côte d'Azur, CHU de Nice, Nice, France; FHU OncoAge, CNRS UMR7284, Inserm U1081, Institute of research on cancer and ageing (IRCAN), université Côte d'Azur, Nice, France
| | - S Leroy
- FHU OncoAge, department of pulmonary medicine, université Côte d'Azur, CHU de Nice, Nice, France; CNRS UMR 7275, institut de pharmacologie moléculaire et cellulaire, université Côte d'Azur, Sophia-Antipolis, France
| |
Collapse
|