1
|
Zhang S, Duitman J, Artigas A, Bos LD. The Complex Immune Cell Composition and Cellular Interaction in the Alveolar Compartment of Patients with Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2025; 72:233-243. [PMID: 39383858 PMCID: PMC11890076 DOI: 10.1165/rcmb.2024-0176tr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/09/2024] [Indexed: 10/11/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by protein-rich edema due to alveolar-capillary barrier dysfunction caused by inflammatory processes. Currently, our understanding of the inflammatory response in patients with ARDS is mainly based on assessment of the systemic compartment and preclinical studies. Investigations into the intricate network of immune cells and their critical functions in the alveolar compartment remain limited. However, with recent improvements in single-cell analyses, our comprehensive understanding of the interactions between immune cells in the lungs has improved. In this review, we summarize the current knowledge about the cellular composition and interactions of different immune cell types within the alveolar space of patients with ARDS. Neutrophils and macrophages are the predominant immune cells in the alveolar space of patients with ARDS. Yet, all immune cells present, including lymphocytes, participate in complex interactions, coordinate recruitment, modulate the lifespan, and control apoptosis through various signaling pathways. Moreover, the cellular composition of alveolar immune cells is associated with the clinical outcomes of patients with ARDS. In conclusion, this synthesis advances our understanding of ARDS immunology, emphasizing the crucial role of immune cells within the alveolar space. Associations between cellular composition and clinical outcomes highlight the significance of exploring distinct alveolar immune cell subsets. Such exploration holds promise for uncovering novel therapeutic targets in ARDS pathophysiology, presenting avenues for enhancing clinical management and treatment strategies for patients with ARDS.
Collapse
Affiliation(s)
| | - JanWillem Duitman
- Department of Pulmonary Medicine
- Department of Experimental Immunology, and
| | - Antonio Artigas
- Corporacion Sanitaria Universitaria Parc Taulí, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), CIBER Enfermedades Respiratorias, Universitat Autónoma de Barcelona, Sabadell, Spain
| | - Lieuwe D.J. Bos
- Department of Intensive Care Medicine
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands; and
| |
Collapse
|
2
|
Kita K, Gawinowska M, Chełmińska M, Niedoszytko M. The Role of Exhaled Breath Condensate in Chronic Inflammatory and Neoplastic Diseases of the Respiratory Tract. Int J Mol Sci 2024; 25:7395. [PMID: 39000502 PMCID: PMC11242091 DOI: 10.3390/ijms25137395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are among the most common chronic respiratory diseases. Chronic inflammation of the airways leads to an increased production of inflammatory markers by the effector cells of the respiratory tract and lung tissue. These biomarkers allow the assessment of physiological and pathological processes and responses to therapeutic interventions. Lung cancer, which is characterized by high mortality, is one of the most frequently diagnosed cancers worldwide. Current screening methods and tissue biopsies have limitations that highlight the need for rapid diagnosis, patient differentiation, and effective management and monitoring. One promising non-invasive diagnostic method for respiratory diseases is the assessment of exhaled breath condensate (EBC). EBC contains a mixture of volatile and non-volatile biomarkers such as cytokines, leukotrienes, oxidative stress markers, and molecular biomarkers, providing significant information about inflammatory and neoplastic states in the lungs. This article summarizes the research on the application and development of EBC assessment in diagnosing and monitoring respiratory diseases, focusing on asthma, COPD, and lung cancer. The process of collecting condensate, potential issues, and selected groups of markers for detailed disease assessment in the future are discussed. Further research may contribute to the development of more precise and personalized diagnostic and treatment methods.
Collapse
Affiliation(s)
- Karolina Kita
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marika Gawinowska
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marta Chełmińska
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
3
|
Wang W, He Z. Gasdermins in sepsis. Front Immunol 2023; 14:1203687. [PMID: 38022612 PMCID: PMC10655013 DOI: 10.3389/fimmu.2023.1203687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is a hyper-heterogeneous syndrome in which the systemic inflammatory response persists throughout the course of the disease and the inflammatory and immune responses are dynamically altered at different pathogenic stages. Gasdermins (GSDMs) proteins are pore-forming executors in the membrane, subsequently mediating the release of pro-inflammatory mediators and inflammatory cell death. With the increasing research on GSDMs proteins and sepsis, it is believed that GSDMs protein are one of the most promising therapeutic targets in sepsis in the future. A more comprehensive and in-depth understanding of the functions of GSDMs proteins in sepsis is important to alleviate the multi-organ dysfunction and reduce sepsis-induced mortality. In this review, we focus on the function of GSDMs proteins, the molecular mechanism of GSDMs involved in sepsis, and the regulatory mechanism of GSDMs-mediated signaling pathways, aiming to provide novel ideas and therapeutic strategies for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Wenhua Wang
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhihui He
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Seyfinejad B, Nemutlu E, Taghizadieh A, Khoubnasabjafari M, Ozkan SA, Jouyban A. Biomarkers in exhaled breath condensate as fingerprints of asthma, chronic obstructive pulmonary disease and asthma-chronic obstructive pulmonary disease overlap: a critical review. Biomark Med 2023; 17:811-837. [PMID: 38179966 DOI: 10.2217/bmm-2023-0420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024] Open
Abstract
Asthma, chronic obstructive pulmonary disease (COPD) and asthma-COPD overlap are the third leading cause of mortality around the world. They share some common features, which can lead to misdiagnosis. To properly manage these conditions, reliable markers for early and accurate diagnosis are needed. Over the past 20 years, many molecules have been investigated in the exhaled breath condensate to better understand inflammation pathways and mechanisms related to these disorders. Recently, more advanced techniques, such as sensitive metabolomic and proteomic profiling, have been used to obtain a more comprehensive understanding. This article reviews the use of targeted and untargeted metabolomic methodology to study asthma, COPD and asthma-COPD overlap.
Collapse
Affiliation(s)
- Behrouz Seyfinejad
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkiye
| | - Ali Taghizadieh
- Tuberculosis & Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Internal Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Khoubnasabjafari
- Tuberculosis & Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anesthesiology & Intensive Care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sibel A Ozkan
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, 06560, Turkiye
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Near East University, PO Box 99138 Nicosia, North Cyprus, Mersin 10, Turkiye
| |
Collapse
|
5
|
Rojas DA, Ponce CA, Bustos A, Cortés V, Olivares D, Vargas SL. Pneumocystis Exacerbates Inflammation and Mucus Hypersecretion in a Murine, Elastase-Induced-COPD Model. J Fungi (Basel) 2023; 9:jof9040452. [PMID: 37108906 PMCID: PMC10142929 DOI: 10.3390/jof9040452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Inflammation and mucus hypersecretion are frequent pathology features of chronic respiratory diseases such as asthma and COPD. Selected bacteria, viruses and fungi may synergize as co-factors in aggravating disease by activating pathways that are able to induce airway pathology. Pneumocystis infection induces inflammation and mucus hypersecretion in immune competent and compromised humans and animals. This fungus is a frequent colonizer in patients with COPD. Therefore, it becomes essential to identify whether it has a role in aggravating COPD severity. This work used an elastase-induced COPD model to evaluate the role of Pneumocystis in the exacerbation of pathology, including COPD-like lung lesions, inflammation and mucus hypersecretion. Animals infected with Pneumocystis developed increased histology features of COPD, inflammatory cuffs around airways and lung vasculature plus mucus hypersecretion. Pneumocystis induced a synergic increment in levels of inflammation markers (Cxcl2, IL6, IL8 and IL10) and mucins (Muc5ac/Muc5b). Levels of STAT6-dependent transcription factors Gata3, FoxA3 and Spdef were also synergically increased in Pneumocystis infected animals and elastase-induced COPD, while the levels of the mucous cell-hyperplasia transcription factor FoxA2 were decreased compared to the other groups. Results document that Pneumocystis is a co-factor for disease severity in this elastase-induced-COPD model and highlight the relevance of STAT6 pathway in Pneumocystis pathogenesis.
Collapse
Affiliation(s)
- Diego A Rojas
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Carolina A Ponce
- Programa de Microbiología y Micología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
| | - Adriel Bustos
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Vicente Cortés
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Daniela Olivares
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Sergio L Vargas
- Programa de Microbiología y Micología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
| |
Collapse
|
6
|
Nourian YH, Salimian J, Ahmadi A, Salehi Z, Karimi M, Emamvirdizadeh A, Azimzadeh Jamalkandi S, Ghanei M. cAMP-PDE signaling in COPD: Review of cellular, molecular and clinical features. Biochem Biophys Rep 2023; 34:101438. [PMID: 36865738 PMCID: PMC9971187 DOI: 10.1016/j.bbrep.2023.101438] [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: 11/26/2022] [Revised: 01/21/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death among non-contagious diseases in the world. PDE inhibitors are among current medicines prescribed for COPD treatment of which, PDE-4 family is the predominant PDE isoform involved in hydrolyzing cyclic adenosine monophosphate (cAMP) that regulates the inflammatory responses in neutrophils, lymphocytes, macrophages and epithelial cells The aim of this study is to investigate the cellular and molecular mechanisms of cAMP-PDE signaling, as an important pathway in the treatment management of patients with COPD. In this review, a comprehensive literature review was performed about the effect of PDEs in COPD. Generally, PDEs are overexpressed in COPD patients, resulting in cAMP inactivation and decreased cAMP hydrolysis from AMP. At normal amounts, cAMP is one of the essential agents in regulating metabolism and suppressing inflammatory responses. Low amount of cAMP lead to activation of downstream inflammatory signaling pathways. PDE4 and PDE7 mRNA transcript levels were not altered in polymorphonuclear leukocytes and CD8 lymphocytes originating from the peripheral venous blood of stable COPD subjects compared to healthy controls. Therefore, cAMP-PDE signaling pathway is one of the most important signaling pathways involved in COPD. By examining the effects of different drugs in this signaling pathway critical steps can be taken in the treatment of this disease.
Collapse
Affiliation(s)
- Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Emamvirdizadeh
- Department of Molecular Genetics, Faculty of Bio Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran,Corresponding author.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Yang S, Tong Y, Chen L, Yu W. Human Identical Sequences, hyaluronan, and hymecromone ─ the new mechanism and management of COVID-19. MOLECULAR BIOMEDICINE 2022; 3:15. [PMID: 35593963 PMCID: PMC9120813 DOI: 10.1186/s43556-022-00077-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/04/2022] [Indexed: 02/08/2023] Open
Abstract
COVID-19 caused by SARS-CoV-2 has created formidable damage to public health and market economy. Currently, SARS-CoV-2 variants has exacerbated the transmission from person-to-person. Even after a great deal of investigation on COVID-19, SARS-CoV-2 is still rampaging globally, emphasizing the urgent need to reformulate effective prevention and treatment strategies. Here, we review the latest research progress of COVID-19 and provide distinct perspectives on the mechanism and management of COVID-19. Specially, we highlight the significance of Human Identical Sequences (HIS), hyaluronan, and hymecromone ("Three-H") for the understanding and intervention of COVID-19. Firstly, HIS activate inflammation-related genes to influence COVID-19 progress through NamiRNA-Enhancer network. Accumulation of hyaluronan induced by HIS-mediated HAS2 upregulation is a substantial basis for clinical manifestations of COVID-19, especially in lymphocytopenia and pulmonary ground-glass opacity. Secondly, detection of plasma hyaluronan can be effective for evaluating the progression and severity of COVID-19. Thirdly, spike glycoprotein of SARS-CoV-2 may bind to hyaluronan and further serve as an allergen to stimulate allergic reaction, causing sudden adverse effects after vaccination or the aggravation of COVID-19. Finally, antisense oligonucleotides of HIS or inhibitors of hyaluronan synthesis (hymecromone) or antiallergic agents could be promising therapeutic agents for COVID-19. Collectively, Three-H could hold the key to understand the pathogenic mechanism and create effective therapeutic strategies for COVID-19.
Collapse
Affiliation(s)
- Shuai Yang
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Ying Tong
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Lu Chen
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Wenqiang Yu
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
8
|
Lorente L, Martín MM, Ortiz-López R, González-Rivero AF, Gómez-Bernal F, González-Mesa A, Jiménez A, Pérez-Cejas A. High mortality rate of septic patients with high blood granzyme B concentrations. Diagn Microbiol Infect Dis 2022; 103:115694. [DOI: 10.1016/j.diagmicrobio.2022.115694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 11/03/2022]
|
9
|
Goplen NP, Cheon IS, Sun J. Age-Related Dynamics of Lung-Resident Memory CD8 + T Cells in the Age of COVID-19. Front Immunol 2021; 12:636118. [PMID: 33854506 PMCID: PMC8039372 DOI: 10.3389/fimmu.2021.636118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Following respiratory viral infections or local immunizations, lung resident-memory T cells (TRM) of the CD8 lineage provide protection against the same pathogen or related pathogens with cross-reactive T cell epitopes. Yet, it is now clear that, if homeostatic controls are lost following viral pneumonia, CD8 TRM cells can mediate pulmonary pathology. We recently showed that the aging process can result in loss of homeostatic controls on CD8 TRM cells in the respiratory tract. This may be germane to treatment modalities in both influenza and coronavirus disease 2019 (COVID-19) patients, particularly, the portion that present with symptoms linked to long-lasting lung dysfunction. Here, we review the developmental cues and functionalities of CD8 TRM cells in viral pneumonia models with a particular focus on their capacity to mediate heterogeneous responses of immunity and pathology depending on immune status.
Collapse
Affiliation(s)
- Nick P Goplen
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - In Su Cheon
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jie Sun
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States.,The Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
10
|
Srinivasan K, Sathiyaseelan M, Raj J, Ranganadin P, Subramanian B. Potential health impacts and lung microbiome changes among smoking and smokeless tobacco use : A technical scan. EURASIAN JOURNAL OF PULMONOLOGY 2021. [DOI: 10.4103/ejop.ejop_108_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
11
|
van Daalen KR, Reijneveld JF, Bovenschen N. Modulation of Inflammation by Extracellular Granzyme A. Front Immunol 2020; 11:931. [PMID: 32508827 PMCID: PMC7248576 DOI: 10.3389/fimmu.2020.00931] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Granzyme A (GrA) has long been recognized as one of the key players in the induction of cell death of neoplastic, foreign or infected cells after granule delivery by cytotoxic cells. While the cytotoxic potential of GrA is controversial in current literature, accumulating evidence now indicates roles for extracellular GrA in modulating inflammation and inflammatory diseases. This paper aims to explore the literature presenting current knowledge on GrA as an extracellular modulator of inflammation by summarizing (i) the presence and role of extracellular GrA in several inflammatory diseases, and (ii) the potential molecular mechanisms of extracellular GrA in augmenting inflammation.
Collapse
Affiliation(s)
- Kim R. van Daalen
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
12
|
Hikichi M, Mizumura K, Maruoka S, Gon Y. Pathogenesis of chronic obstructive pulmonary disease (COPD) induced by cigarette smoke. J Thorac Dis 2019; 11:S2129-S2140. [PMID: 31737341 DOI: 10.21037/jtd.2019.10.43] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease that is characterized by functional and structural alterations primarily caused by long-term inhalation of harmful particles. Cigarette smoke (CS) induces airway inflammation in COPD, which is known to persist even after smoking cessation. This review discusses the basic pathogenesis of COPD, with particular focus on an endogenous protective mechanism against oxidative stress via Nrf2, altered immune response of the airway inflammatory cells, exaggerated cellular senescence of the lung structural cells, and cell death with expanded inflammation. Recently, CS-induced mitochondria autophagy is reported to initiate programmed necrosis (necroptosis). Necroptosis is a new concept of cell death which is driven by a defined molecular pathway along with exaggerated inflammation. This new cell death mechanism is of importance due to its ability to produce more inflammatory substances during the process of epithelial death, contributing to persistent airway inflammation that cannot be explained by apoptosis-derived cell death. Autophagy is an auto-cell component degradation system executed by lysosomes that controls protein and organelle degradation for successful homeostasis. As well as in the process of necroptosis, autophagy is also observed during cellular senescence. Aging of the lungs results in the acquisition of senescence-associated secretory phenotypes (SASP) that are known to secrete inflammatory cytokines, chemokines, growth factors, and matrix metalloproteinases resulting in chronic low-grade inflammation. In future research, we intend to highlight the genetic and epigenetic approaches that can facilitate the understanding of disease susceptibility. The goal of precision medicine is to establish more accurate diagnosis and treatment methods based on the patient-specific pathogenic characteristics. This review provides insights into CS-induced COPD pathogenesis, which contributes to a very complex disease. Investigating the mechanism of developing COPD, along with the availability of the particular inhibitors, will lead to new therapeutic approaches in COPD treatment.
Collapse
Affiliation(s)
- Mari Hikichi
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenji Mizumura
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Abstract
Regulated cell death is a major mechanism to eliminate damaged, infected, or superfluous cells. Previously, apoptosis was thought to be the only regulated cell death mechanism; however, new modalities of caspase-independent regulated cell death have been identified, including necroptosis, pyroptosis, and autophagic cell death. As an understanding of the cellular mechanisms that mediate regulated cell death continues to grow, there is increasing evidence that these pathways are implicated in the pathogenesis of many pulmonary disorders. This review summarizes our understanding of regulated cell death as it pertains to the pathogenesis of chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Maor Sauler
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA;
| | - Isabel S Bazan
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA;
| | - Patty J Lee
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
14
|
Alcamo AM, Pang D, Bashir DA, Carcillo JA, Nguyen TC, Aneja RK. Role of Damage-Associated Molecular Patterns and Uncontrolled Inflammation in Pediatric Sepsis-Induced Multiple Organ Dysfunction Syndrome. J Pediatr Intensive Care 2018; 8:25-31. [PMID: 31073505 DOI: 10.1055/s-0038-1675639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/19/2018] [Indexed: 01/20/2023] Open
Abstract
The incidence of multiple organ dysfunction syndrome (MODS) in sepsis varies from 17 to 73% and furthermore, increases the risk of death by 60% when controlled for the number of dysfunctional organs. Several MODS phenotypes exist, each unique in presentation and pathophysiology. Common to the phenotypes is the stimulation of the immune response by pathogen-associated molecular patterns (PAMPs), or danger-associated molecular patterns (DAMPs) causing an unremitting inflammation. Two of the MODS phenotypes are discussed in detail, thrombocytopenia-associated multiple organ failure (TAMOF) and the hyperinflammatory phenotype-macrophage activating syndrome (MAS) and hemophagocytic lymphohistiocytosis (HLH). In the end, we will briefly review the role of mitochondrial dysfunction as a significant contributor to the pathogenesis of MODS.
Collapse
Affiliation(s)
- Alicia M Alcamo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Diana Pang
- Department of Critical Care Medicine, Children's Hospital of the King's Daughters, Norfolk, Virginia, United States
| | - Dalia A Bashir
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Joseph A Carcillo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Trung C Nguyen
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
15
|
Notch signaling pathway suppresses CD8 + T cells activity in patients with lung adenocarcinoma. Int Immunopharmacol 2018; 63:129-136. [PMID: 30086535 DOI: 10.1016/j.intimp.2018.07.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/28/2022]
Abstract
Evolution and progression of cancer always leads to CD8+ T cells dysfunction/exhaustion. Controversy remains as to the role of Notch signaling pathway in CD8+ T cells regulation in tumorigenesis. Thus, the aim of this study was to investigate the immunomodulatory activity of Notch signaling pathway to peripheral and lung-resident CD8+ T cells in patients with lung adenocarcinoma. Forty-eight lung adenocarcinoma patients and twenty healthy individuals were enrolled in the current study, and CD8+ T cells were purified from both peripheral bloods and bronchoalveolar lavage fluids. Notch receptor mRNA expression was semi-quantified by real-time PCR. Cytolytic and noncytolytic activity of CD8+ T cells evaluated in direct and indirect contact co-culture with A549 cells in response to Notch signaling inhibition by measuring of lactate dehydrogenase release and cytokines production. Expression of Fas ligand (FasL), perforin, and granzyme B were also assessed by flow cytometry. Notch2 mRNA expression was elevated in both peripheral and lung-resident CD8+ T cells in lung adenocarcinoma patients, however, did not correlated with tumor stages or epidermal growth factor receptor mutation. Peripheral CD8+ T cells from healthy individuals exhibited stronger cytotoxicity in direct contact co-culture system, which was not influenced by Notch signaling inhibition. Moreover, suppression of Notch signaling augmented cytotoxicity of peripheral and lung-resident CD8+ T cells from lung adenocarcinoma patients in direct contact co-culture system, and promoted interferon-γ production in both systems. This process was accompanied by increased expression of FasL and perforin within CD8+ T cells. The current data revealed a potential immunosuppressive property of Notch signaling pathway to CD8+ T cells probably via inhibition of FasL and perforin in lung adenocarcinoma patients.
Collapse
|
16
|
Spadaro S, Kozhevnikova I, Casolari P, Ruggeri P, Bellini T, Ragazzi R, Barbieri F, Marangoni E, Caramori G, Volta CA. Lower airways inflammation in patients with ARDS measured using endotracheal aspirates: a pilot study. BMJ Open Respir Res 2017; 4:e000222. [PMID: 29071081 PMCID: PMC5647481 DOI: 10.1136/bmjresp-2017-000222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/17/2017] [Indexed: 11/07/2022] Open
Abstract
Introduction Our knowledge of acute respiratory distress syndrome (ARDS) pathogenesis is incomplete. The goal of this pilot study is to investigate the feasibility of measuring lower airways inflammation in patients with ARDS using repeated endotracheal aspirates (ETAs). Methods ETAs were obtained within 24 hours by intensive care unit admission from 25 mechanically ventilated patients with ARDS and 10 of them underwent a second ETA within 96 hours after the first sampling. In each sample, cell viability was assessed using trypan blue exclusion method and the total and differential cell counts were measured using Neubauer-improved cell counting chamber and cytospins stained with Diff-Quik. Results The median cell viability was 89 (IQR 80–93)%, with a median total cell count of 305 (IQR 130–1270)×103/mL and a median macrophage, neutrophil, lymphocyte and eosinophil count, respectively, of 19.8 (IQR 5.4–71.6)×103/mL; 279 (IQR 109–1213)×103/mL; 0 (IQR 0–0.188)×103/mL; 0 (IQR 0–1.050)×103/mL. Eosinophil count in the ETA correlated with the number of blood eosinophils (r=0.4840, p=0.0142). Cell viability and total and differential cell counts were neither significantly different in the second ETA compared with the first ETA nor were unaffected by the presence or absence of bacteria in the blood and/or ETA, or by the ARDS aetiology, apart from the macrophage count which was significantly increased in patients with ARDS associated with acute pancreatitis compared with those associated with pneumonia (p=0.0143). Conclusions ETA can be used to investigate the cellularity of the lower airways in patients with ARDS and it is an easy-to-perform and non-invasive procedure. Eosinophil counts in ETA and blood are significantly correlated. The number of macrophages in ETA may be affected by the aetiology of the ARDS.
Collapse
Affiliation(s)
- Savino Spadaro
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| | - Iryna Kozhevnikova
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| | - Paolo Casolari
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-Correlate (CEMICEF), Dipartimento di Scienze Mediche, Sezione di Medicina Interna e Cardiorespiratoria, Università di Ferrara, Ferrara, Italy
| | - Paolo Ruggeri
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Messina, Italy
| | - Tiziana Bellini
- Department of Biomedical and Specialty Surgical Sciences, Medical Biochemistry, Molecular Biology and Genetics Section, University of Ferrara, Ferrara, Italy
| | - Riccardo Ragazzi
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| | - Federica Barbieri
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| | - Elisabetta Marangoni
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| | - Gaetano Caramori
- Unità Operativa Complessa di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Messina, Italy
| | - Carlo Alberto Volta
- Unità Operativa di Anestesia e Rianimazione Universitaria dell'Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, University of Ferrara, Ferrara, Italy
| |
Collapse
|
17
|
Maknitikul S, Luplertlop N, Grau GER, Ampawong S. Dysregulation of pulmonary endothelial protein C receptor and thrombomodulin in severe falciparum malaria-associated ARDS relevant to hemozoin. PLoS One 2017; 12:e0181674. [PMID: 28732053 PMCID: PMC5521846 DOI: 10.1371/journal.pone.0181674] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 07/04/2017] [Indexed: 12/20/2022] Open
Abstract
To investigate the role of the protein C system, endothelial protein C receptor (EPCR) and thrombomodulin (TM) in the pathogenesis of malaria-associated acute respiratory distress syndrome (ARDS) in relation to hemozoin and proinflammatory cytokines-induced type II pneumocyte injury and -aggravated pulmonary resolution. A total of 29 left-over lung specimens that were obtained from patients who died from severe falciparum malaria were examined. Histopathological, immunohistochemical and electron microscopic analyses revealed that ARDS coexisted with pulmonary edema and systemic bleeding; the severity was dependent on the level of hemozoin deposition in the lung and internal alveolar hemorrhaging. The loss of EPCR and TM was primarily identified in ARDS patients and was related to the level of hemozoin, parasitized red blood cell (PRBC) and white blood cell accumulation in the lung. Moreover, an in vitro analysis demonstrated that interleukin-13 and -31 and hemozoin induced pneumocytic cell injury and apoptosis, as assessed by EB/AO staining, electron microscopy and the up-regulation of CARD-9 mRNA (caspase recruitment domain-9 messenger-ribonucleic acid). The dysregulation of EPCR and TM in the lung, especially in those with increased levels of hemozoin, may play an important role in the pathogenesis of malaria-associated ARDS through an apoptotic pathway.
Collapse
Affiliation(s)
- Sitang Maknitikul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Natthanej Luplertlop
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | - Georges E. R. Grau
- Vascular Immunology, Department of Pathology, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
18
|
García-Laorden MI, Lorente JA, Flores C, Slutsky AS, Villar J. Biomarkers for the acute respiratory distress syndrome: how to make the diagnosis more precise. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:283. [PMID: 28828358 DOI: 10.21037/atm.2017.06.49] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The acute respiratory distress syndrome (ARDS) is an acute inflammatory process of the lung caused by a direct or indirect insult to the alveolar-capillary membrane. Currently, ARDS is diagnosed based on a combination of clinical and physiological variables. The lack of a specific biomarker for ARDS is arguably one of the most important obstacles to progress in developing novel treatments for ARDS. In this article, we will review the current understanding of some appealing biomarkers that have been measured in human blood, bronchoalveolar lavage fluid (BALF) or exhaled gas that could be used for identifying patients with ARDS, for enrolling ARDS patients into clinical trials, or for better monitoring of patient's management. After a literature search, we identified several biomarkers that are associated with the highest sensitivity and specificity for the diagnosis or outcome prediction of ARDS: receptor for advanced glycation end-products (RAGE), angiopoietin-2 (Ang-2), surfactant protein D (SP-D), inteleukin-8, Fas and Fas ligand, procollagen peptide (PCP) I and III, octane, acetaldehyde, and 3-methylheptane. In general, these are cell-specific for epithelial or endothelial injury or involved in the inflammatory or infectious response. No biomarker or biomarkers have yet been confirmed for the diagnosis of ARDS or prediction of its prognosis. However, it is anticipated that in the near future, using biomarkers for defining ARDS, or for determining those patients who are more likely to benefit from a given therapy will have a major effect on clinical practice.
Collapse
Affiliation(s)
- M Isabel García-Laorden
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - José A Lorente
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Intensive Care Unit, Hospital Universitario de Getafe, Madrid, Spain.,Department of Medicine, Universidad Europea, Madrid, Spain
| | - Carlos Flores
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Research Unit, Hospital NS de Candelaria, Santa Cruz de Tenerife, Spain
| | - Arthur S Slutsky
- Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain.,Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Intratracheal Administration of Small Interfering RNA Targeting Fas Reduces Lung Ischemia-Reperfusion Injury. Crit Care Med 2017; 44:e604-13. [PMID: 26963318 DOI: 10.1097/ccm.0000000000001601] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Lung ischemia-reperfusion injury is the main cause of primary graft dysfunction after lung transplantation and results in increased morbidity and mortality. Fas-mediated apoptosis is one of the pathologic mechanisms involved in the development of ischemia-reperfusion injury. We hypothesized that the inhibition of Fas gene expression in lungs by intratracheal administration of small interfering RNA could reduce lung ischemia-reperfusion injury in an ex vivo model reproducing the procedural sequence of lung transplantation. DESIGN Prospective, randomized, controlled experimental study. SETTING University research laboratory. SUBJECTS C57/BL6 mice weighing 28-30 g. INTERVENTIONS Ischemia-reperfusion injury was induced in lungs isolated from mice, 48 hours after treatment with intratracheal small interfering RNA targeting Fas, control small interfering RNA, or vehicle. Isolated lungs were exposed to 6 hours of cold ischemia (4°C), followed by 2 hours of warm (37°C) reperfusion with a solution containing 10% of fresh whole blood and mechanical ventilation with constant low driving pressure. MEASUREMENTS AND MAIN RESULTS Fas gene expression was significantly silenced at the level of messenger RNA and protein after ischemia-reperfusion in lungs treated with small interfering RNA targeting Fas compared with lungs treated with control small interfering RNA or vehicle. Silencing of Fas gene expression resulted in reduced edema formation (bronchoalveolar lavage protein concentration and lung histology) and improvement in lung compliance. These effects were associated with a significant reduction of pulmonary cell apoptosis of lungs treated with small interfering RNA targeting Fas, which did not affect cytokine release and neutrophil infiltration. CONCLUSIONS Fas expression silencing in the lung by small interfering RNA is effective against ischemia-reperfusion injury. This approach represents a potential innovative strategy of organ preservation before lung transplantation.
Collapse
|
20
|
Svedova J, Ménoret A, Mittal P, Ryan JM, Buturla JA, Vella AT. Therapeutic blockade of CD54 attenuates pulmonary barrier damage in T cell-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2017; 313:L177-L191. [PMID: 28473322 DOI: 10.1152/ajplung.00050.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious, often fatal condition without available pharmacotherapy. Although the role of innate cells in ARDS has been studied extensively, emerging evidence suggests that T cells may be involved in disease etiology. Staphylococcus aureus enterotoxins are potent T-cell mitogens capable of triggering life-threatening shock. We demonstrate that 2 days after inhalation of S. aureus enterotoxin A, mice developed T cell-mediated increases in vascular permeability, as well as expression of injury markers and caspases in the lung. Pulmonary endothelial cells underwent sequential phenotypic changes marked by rapid activation coinciding with inflammatory events secondary to T-cell priming, followed by reductions in endothelial cell number juxtaposing simultaneous T-cell expansion and cytotoxic differentiation. Although initial T-cell activation influenced the extent of lung injury, CD54 (ICAM-1) blocking antibody administered well after enterotoxin exposure substantially attenuated pulmonary barrier damage. Thus CD54-targeted therapy may be a promising candidate for further exploration into its potential utility in treating ARDS patients.
Collapse
Affiliation(s)
- Julia Svedova
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - Antoine Ménoret
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut.,Institute for Systems Genomics, UConn Health, Farmington, Connecticut; and
| | - Payal Mittal
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - Joseph M Ryan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - James A Buturla
- Department of Internal Medicine, UConn Health, Farmington, Connecticut
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut;
| |
Collapse
|
21
|
Abstract
OBJECTIVE To describe the pathophysiology associated with multiple organ dysfunction syndrome in children. DATA SOURCES Literature review, research data, and expert opinion. STUDY SELECTION Not applicable. DATA EXTRACTION Moderated by an experienced expert from the field, pathophysiologic processes associated with multiple organ dysfunction syndrome in children were described, discussed, and debated with a focus on identifying knowledge gaps and research priorities. DATA SYNTHESIS Summary of presentations and discussion supported and supplemented by relevant literature. CONCLUSIONS Experiment modeling suggests that persistent macrophage activation may be a pathophysiologic basis for multiple organ dysfunction syndrome. Children with multiple organ dysfunction syndrome have 1) reduced cytochrome P450 metabolism inversely proportional to inflammation; 2) increased circulating damage-associated molecular pattern molecules from injured tissues; 3) increased circulating pathogen-associated molecular pattern molecules from infection or endogenous microbiome; and 4) cytokine-driven epithelial, endothelial, mitochondrial, and immune cell dysfunction. Cytochrome P450s metabolize endogenous compounds and xenobiotics, many of which ameliorate inflammation, whereas damage-associated molecular pattern molecules and pathogen-associated molecular pattern molecules alone and together amplify the cytokine production leading to the inflammatory multiple organ dysfunction syndrome response. Genetic and environmental factors can impede inflammation resolution in children with a spectrum of multiple organ dysfunction syndrome pathobiology phenotypes. Thrombocytopenia-associated multiple organ dysfunction syndrome patients have extensive endothelial activation and thrombotic microangiopathy with associated oligogenic deficiencies in inhibitory complement and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13. Sequential multiple organ dysfunction syndrome patients have soluble Fas ligand-Fas-mediated hepatic failure with associated oligogenic deficiencies in perforin and granzyme signaling. Immunoparalysis-associated multiple organ dysfunction syndrome patients have impaired ability to resolve infection and have associated environmental causes of lymphocyte apoptosis. These inflammation phenotypes can lead to macrophage activation syndrome. Resolution of multiple organ dysfunction syndrome requires elimination of the source of inflammation. Full recovery of organ functions is noted 6-18 weeks later when epithelial, endothelial, mitochondrial, and immune cell regeneration and reprogramming is completed.
Collapse
|
22
|
Fujishima S. Organ dysfunction as a new standard for defining sepsis. Inflamm Regen 2016; 36:24. [PMID: 29259697 PMCID: PMC5725936 DOI: 10.1186/s41232-016-0029-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/01/2016] [Indexed: 01/20/2023] Open
Abstract
Despite advances in intensive care and the widespread use of standardized care included in the Surviving Sepsis Campaign Guidelines, sepsis remains a leading cause of death, and the prevalence of sepsis increases concurrent with the aging process. The diagnosis of sepsis was originally based on the evidence of persistent bacteremia (septicemia) but was modified in 1992 to incorporate systemic inflammatory response syndrome (SIRS). Since then, SIRS has become the gold standard for the diagnosis of sepsis. In 2016, the Society of Critical Care Medicine and the European Society of Intensive Care Medicine published a new clinical definition of sepsis that is called Sepsis-3. In contrast to previous definitions, Sepsis-3 is based on organ dysfunctions and uses a sequential organ failure (SOFA) score as an index. Thus, patients diagnosed with respect to Sepsis-3 will inevitably represent a different population than those previously diagnosed. We assume that this drastic change in clinical definition will affect not only clinical practice but also the viewpoint and focus of basic research. This review intends to summarize the pathophysiology of sepsis and organ dysfunction and discusses potential directions for future research.
Collapse
Affiliation(s)
- Seitaro Fujishima
- Center for General Medicine Education, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Arnold N, Girke T, Sureshchandra S, Nguyen C, Rais M, Messaoudi I. Genomic and functional analysis of the host response to acute simian varicella infection in the lung. Sci Rep 2016; 6:34164. [PMID: 27677639 PMCID: PMC5039758 DOI: 10.1038/srep34164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/08/2016] [Indexed: 01/19/2023] Open
Abstract
Varicella Zoster Virus (VZV) is the causative agent of varicella and herpes zoster. Although it is well established that VZV is transmitted via the respiratory route, the host-pathogen interactions during acute VZV infection in the lungs remain poorly understood due to limited access to clinical samples. To address these gaps in our knowledge, we leveraged a nonhuman primate model of VZV infection where rhesus macaques are intrabronchially challenged with the closely related Simian Varicella Virus (SVV). Acute infection is characterized by immune infiltration of the lung airways, a significant up-regulation of genes involved in antiviral-immunity, and a down-regulation of genes involved in lung development. This is followed by a decrease in viral loads and increased expression of genes associated with cell cycle and tissue repair. These data provide the first characterization of the host response required to control varicella virus replication in the lung and provide insight into mechanisms by which VZV infection can cause lung injury in an immune competent host.
Collapse
Affiliation(s)
- Nicole Arnold
- Graduate Program in Microbiology, University of California-Riverside, CA, USA
| | - Thomas Girke
- Department of Botany and Plant Sciences, University of California-Riverside, CA, USA
| | - Suhas Sureshchandra
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California-Riverside, CA, USA
| | - Christina Nguyen
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| | - Ilhem Messaoudi
- Graduate Program in Microbiology, University of California-Riverside, CA, USA
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California-Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| |
Collapse
|
24
|
Levitt JE, Rogers AJ. Proteomic study of acute respiratory distress syndrome: current knowledge and implications for drug development. Expert Rev Proteomics 2016; 13:457-69. [PMID: 27031735 DOI: 10.1586/14789450.2016.1172481] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The acute respiratory distress syndrome (ARDS) is a common cause of acute respiratory failure, and is associated with substantial mortality and morbidity. Dozens of clinical trials targeting ARDS have failed, with no drug specifically targeting lung injury in widespread clinical use. Thus, the need for drug development in ARDS is great. Targeted proteomic studies in ARDS have identified many key pathways in the disease, including inflammation, epithelial injury, endothelial injury or activation, and disordered coagulation and repair. Recent studies reveal the potential for proteomic changes to identify novel subphenotypes of ARDS patients who may be most likely to respond to therapy and could thus be targeted for enrollment in clinical trials. Nontargeted studies of proteomics in ARDS are just beginning and have the potential to identify novel drug targets and key pathways in the disease. Proteomics will play an important role in phenotyping of patients and developing novel therapies for ARDS in the future.
Collapse
Affiliation(s)
- Joseph E Levitt
- a Division of Pulmonary and Critical Care Medicine , Stanford University , Stanford , CA , USA
| | - Angela J Rogers
- a Division of Pulmonary and Critical Care Medicine , Stanford University , Stanford , CA , USA
| |
Collapse
|
25
|
Wang L, Qin W, Zhang J, Bao C, Zhang H, Che Y, Sun C, Gu J, Feng X, Du C, Han W, Richard PL, Lei L. Adh enhances Actinobacillus pleuropneumoniae pathogenicity by binding to OR5M11 and activating p38 which induces apoptosis of PAMs and IL-8 release. Sci Rep 2016; 6:24058. [PMID: 27046446 PMCID: PMC4820727 DOI: 10.1038/srep24058] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/21/2016] [Indexed: 12/16/2022] Open
Abstract
Members of the Trimeric Autotransporter Adhesin (TAA) family play a crucial role in the adhesion of Gram-negative pathogens to host cells, but the immunopathogenesis of TAAs remains unknown. Our previous studies demonstrated that Adh from Actinobacillus pleuropneumoniae (A. pleuropneumoniae) is required for full bacterial pathogenicity. Alveolar macrophages are the first line of defense against respiratory infections. This study compared the interactions between porcine alveolar macrophages (PAMs) and wild-type A. pleuropneumoniae (5b WT) or an Adh-deletion strain (5b ΔAdh) via gene microarray, immunoprecipitation and other technologies. We found that Adh was shown to interact with the PAMs membrane protein OR5M11, an olfactory receptor, resulting in the high-level secretion of IL-8 by activation of p38 MAPK signaling pathway. Subsequently, PAMs apoptosis via the activation of the Fax and Bax signaling pathways was observed, followed by activation of caspases 8, 9, and 3. The immunological pathogenic roles of Adh were also confirmed in both murine and piglets infectious models in vivo. These results identify a novel immunological strategy for TAAs to boost the pathogenicity of A. pleuropneumoniae. Together, these datas reveal the high versatility of the Adh protein as a virulence factor and provide novel insight into the immunological pathogenic role of TAAs.
Collapse
Affiliation(s)
- Lei Wang
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China.,College of Animal Science, Henan Institute of Science and Technology, Xinxiang, P. R. China
| | - Wanhai Qin
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Jing Zhang
- Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Chuntong Bao
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Hu Zhang
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Yanyi Che
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Changjiang Sun
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Jingmin Gu
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Xin Feng
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Chongtao Du
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | - Wenyu Han
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| | | | - Liancheng Lei
- College of Veterinary Medicine, JiLin University, Changchun, P. R. China
| |
Collapse
|
26
|
Nelin LD, White HA, Jin Y, Trittmann JK, Chen B, Liu Y. The Src family tyrosine kinases src and yes have differential effects on inflammation-induced apoptosis in human pulmonary microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2016; 310:L880-8. [PMID: 26919896 DOI: 10.1152/ajplung.00306.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/19/2016] [Indexed: 01/11/2023] Open
Abstract
Endothelial cells are essential for normal lung function: they sense and respond to circulating factors and hemodynamic alterations. In inflammatory lung diseases such as acute respiratory distress syndrome, endothelial cell apoptosis is an inciting event in pathogenesis and a prominent pathological feature. Endothelial cell apoptosis is mediated by circulating inflammatory factors, which bind to receptors on the cell surface, activating signal transduction pathways, leading to caspase-3-mediated apoptosis. We hypothesized that yes and src have differential effects on caspase-3 activation in human pulmonary microvascular endothelial cells (hPMVEC) due to differential downstream signaling effects. To test this hypothesis, hPMVEC were treated with siRNA against src (siRNAsrc), siRNA against yes (siRNAyes), or their respective scramble controls. After recovery, the hPMVEC were treated with cytomix (LPS, IL-1β, TNF-α, and IFN-γ). Treatment with cytomix induced activation of the extracellular signal-regulated kinase (ERK) pathway and caspase-3-mediated apoptosis. Treatment with siRNAsrc blunted cytomix-induced ERK activation and enhanced cleaved caspase-3 levels, while treatment with siRNAyes enhanced cytomix-induced ERK activation and attenuated levels of cleaved caspase-3. Inhibition of the ERK pathway using U0126 enhanced cytomix-induced caspase-3 activity. Treatment of hPMVEC with cytomix induced Akt activation, which was inhibited by siRNAsrc. Inhibition of the phosphatidylinositol 3-kinase/Akt pathway using LY294002 prevented cytomix-induced ERK activation and augmented cytomix-induced caspase-3 cleavage. Together, our data demonstrate that, in hPMVEC, yes activation blunts the ERK cascade in response to cytomix, resulting in greater apoptosis, while cytomix-induced src activation induces the phosphatidylinositol 3-kinase pathway, which leads to activation of Akt and ERK and attenuation of apoptosis.
Collapse
Affiliation(s)
- Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Hilary A White
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
27
|
Song J, Lu H, Zheng X, Huang X. Effects of Vascular Endothelial Growth Factor in Recovery Phase of Acute Lung Injury in Mice. Lung 2015; 193:1029-36. [PMID: 26415949 DOI: 10.1007/s00408-015-9803-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
AIM To test the hypothesis that exogenous administration of vascular endothelial growth factor (VEGF) promotes lung repair in acute lung injury (ALI). METHODS ALI was induced by intranasal lipopolysaccharide (LPS) administration in mice, followed by different treatment protocols for 7 days in 3 groups (n = 6, each) including the LPS, the VEGF and the anti-VEGF group. At day 7, peripheral blood and lungs were collected. Lung wet-to-dry (W/D) ratio and lung injury score were measured. Immunohistochemistry assay was employed to detect the number of pulmonary vessels. Circulating endothelial progenitor cells (EPCs) was detected using flow cytometric analysis, and the apoptosis of lung cells was determined by TUNEL staining. RESULTS VEGF treatment reduced W/D ratio and pulmonary neutrophil infiltration in the VEGF group compared with the LPS group. The treatment of VEGF increased the number of pulmonary vessels, and significantly increased the number of circulating EPC cells. Moreover, administration of VEGF decreased the percentage of apoptotic cells in the VEGF group. CONCLUSIONS Our results suggest that VEGF may contribute to vascular endothelial repair and function as a protective factor against ALI.
Collapse
Affiliation(s)
- Junfeng Song
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China.
| | - Hui Lu
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| | - Xuyang Zheng
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| | - Xianmei Huang
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| |
Collapse
|
28
|
Jin Z, Pan X, Zhou K, Bi H, Wang L, Yu L, Wang Q. Biological effects and mechanisms of action of mesenchymal stem cell therapy in chronic obstructive pulmonary disease. J Int Med Res 2015; 43:303-10. [PMID: 25834280 DOI: 10.1177/0300060514568733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most frequent chronic respiratory disease and a leading cause of morbidity and mortality, worldwide. Given that the foremost risk factor leading to the development of COPD is cigarette smoke, the initial treatment for COPD is smoking cessation. Even after smoking cessation, inflammation, apoptosis and oxidative stress can persist and continue to contribute to COPD. Although current therapies for COPD (which are primarily based on anti-inflammatory drugs such as corticosteroids, theophylline and bronchodilators) reduce airway obstruction, limit COPD exacerbation and improve the patient's health-related quality-of-life, none can prevent disease progression or reduce mortality. Recent advances in stem cell research have provided novel insight into the potential of bone marrow mesenchymal stem cells (MSCs) in the treatment of several pulmonary diseases. This review article discusses the biological effects and mechanisms of action of MSC transplantation in COPD, and highlights the foundation that MSCs provide for novel therapeutic approaches in COPD.
Collapse
Affiliation(s)
- Zhixian Jin
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Xinghua Pan
- Stem Cell Engineering Laboratory of Yunnan Province, Department of Clinical Research, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Kaihua Zhou
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Hong Bi
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Liyan Wang
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Lu Yu
- Department of Pathology, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Qing Wang
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| |
Collapse
|
29
|
Arias M, Jiménez de Bagües M, Aguiló N, Menao S, Hervás-Stubbs S, de Martino A, Alcaraz A, Simon M, Froelich C, Pardo J. Elucidating Sources and Roles of Granzymes A and B during Bacterial Infection and Sepsis. Cell Rep 2014; 8:420-9. [DOI: 10.1016/j.celrep.2014.06.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/05/2014] [Accepted: 06/10/2014] [Indexed: 10/25/2022] Open
|
30
|
Jeong EK, Kim YK, Kim SH, Lee CH, Kim JS. Systemic capillary leak syndrome under general anesthesia: a case report. Korean J Anesthesiol 2014; 66:462-6. [PMID: 25006371 PMCID: PMC4085268 DOI: 10.4097/kjae.2014.66.6.462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/27/2013] [Accepted: 05/28/2013] [Indexed: 11/27/2022] Open
Abstract
Systemic capillary leak syndrome (SCLS) is very rare and lethal disease and only 150 cases have been reported after the first publication of its report in 1960 by Clarkson. SCLS is characterized by hemoconcentation and hypoalbuminemia caused by reversible plasma extravasation. Its mechanism is unknown, but transient dysfunction of the endothelium is the most suspected cause and trigger of this event may cause immunologic disarrangement. After recovery of endothelial function, fluid injected during the shock period is redistributed and can cause severe pulmonary edema. SCLS should be considered in patients with acute and severe hypotension with hemoconcentration and hypoalbuminemia without obvious cardiac dysfunction. Especially we should take into account the possibility of SCLS if fluid replacement does not work or the shock state is aggravated despite aggressive fluid resuscitation and vasopressor administration. SCLS itself is a very rare disease; furthermore, SCLS that develops during well-controlled surgery is even more rare. So we report this case with review of the literature.
Collapse
Affiliation(s)
- Eui-Kyun Jeong
- Department of Anesthesiology and Pain Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Young-Ki Kim
- Department of Anesthesiology and Pain Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Se-Hun Kim
- Department of Anesthesiology and Pain Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Chang-Hee Lee
- Department of Anesthesiology and Pain Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Jin-Sun Kim
- Department of Anesthesiology and Pain Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| |
Collapse
|
31
|
Angelis N, Porpodis K, Zarogoulidis P, Spyratos D, Kioumis I, Papaiwannou A, Pitsiou G, Tsakiridis K, Mpakas A, Arikas S, Tsiouda T, Katsikogiannis N, Kougioumtzi I, Machairiotis N, Argyriou M, Kessisis G, Zarogoulidis K. Airway inflammation in chronic obstructive pulmonary disease. J Thorac Dis 2014; 6 Suppl 1:S167-72. [PMID: 24672691 DOI: 10.3978/j.issn.2072-1439.2014.03.07] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 03/04/2014] [Indexed: 11/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory airway disease whose incidence and mortality increases every year. It is associated with an abnormal inflammatory response of the lung to toxic particles or gases (usually cigarette smoke). A central role in the pathophysiology has been shown to play a chronic inflammation of the airways that is expressed primarily by hypersecretion of mucus, stenosis of the smaller airways and the establishment of pulmonary emphysema. There is an increasing trend for assessing the inflammatory pattern of inflammatory airway diseases through mediators measured by noninvasive techniques. Markers in biological fluids and exhaled air have been the subject of intense evaluation over the past few years, with some of them reaching their introduction into clinical practice, while others remain as research tools. Of particular interest for the scientific community is the discovery of clinically exploitable biomarkers associated with specific phenotypes of the disease. Studying the effects of therapeutic interventions in these biomarkers may lead to targeted therapy based on phenotype and this is perhaps the future of therapeutics in COPD.
Collapse
Affiliation(s)
- Nikolaos Angelis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Konstantinos Porpodis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Paul Zarogoulidis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Dionysios Spyratos
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Ioannis Kioumis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Antonis Papaiwannou
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Georgia Pitsiou
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Kosmas Tsakiridis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Andreas Mpakas
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Stamatis Arikas
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Theodora Tsiouda
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Nikolaos Katsikogiannis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Ioanna Kougioumtzi
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Nikolaos Machairiotis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Michael Argyriou
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - George Kessisis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| | - Konstantinos Zarogoulidis
- 1 Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; 2 Cardiology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece ; 3 Internal Medicine Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece ; 4 Surgery Department (NHS), University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece ; 5 2nd Cardiac Surgery Department, "Evangelismos" General Hospital, Athens, Greece ; 6 Oncology Department, "Saint Luke" Private Clinic, Thessaloniki, Panorama, Greece
| |
Collapse
|
32
|
Wang X, Luo F, Zhao H. Paraquat-induced reactive oxygen species inhibit neutrophil apoptosis via a p38 MAPK/NF-κB-IL-6/TNF-α positive-feedback circuit. PLoS One 2014; 9:e93837. [PMID: 24714343 PMCID: PMC3979731 DOI: 10.1371/journal.pone.0093837] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 03/06/2014] [Indexed: 12/31/2022] Open
Abstract
Paraquat (PQ), a widely used herbicide and potent reactive oxygen species (ROS) inducer, can injure multiple tissues and organs, especially the lung. However, the underlying mechanism is still poorly understood. According to previous reports, neutrophil aggregation and excessive ROS production might play pivotal pathogenetic roles. In the present study, we found that PQ could prolong neutrophil lifespan and induce ROS generation in a concentration-independent manner. Activated nuclear factor-κB (NF-κB), p38 mitogen-activated kinase (p38 MAPK), and myeloid cell leukemia sequence 1 (Mcl-1) but not Akt signaling pathways were involved in this process, as well as increasing levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and IL-1β. Furthermore, the proinflammatory mediators IL-6 and TNF-α could in turn promote ROS generation, creating a vicious cycle. The existence of such a feedback loop is supported by our finding that neutrophil apoptosis is attenuated by PQ in a concentration-independent manner and could partially explain the clinical dilemma why oxygen therapy will exacerbate PQ induced tissue injury.
Collapse
Affiliation(s)
- Xiaolong Wang
- Emergency department, the 2nd affiliated hospital of Chongqing Medical University, Chongqing, China
| | - Fuling Luo
- Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hengguang Zhao
- Department of Dermatology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Simpson JL, Gibson PG, Yang IA, Upham J, James A, Reynolds PN, Hodge S. Altered sputum granzyme B and granzyme B/proteinase inhibitor-9 in patients with non-eosinophilic asthma. Respirology 2013; 19:280-287. [PMID: 24372772 DOI: 10.1111/resp.12213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/30/2013] [Accepted: 10/04/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVE The non-eosinophilic phenotype of asthma (NEA) is associated with chronic airway inflammation and airway neutrophilia. An accumulation of apoptotic airway epithelial cells, if not efficiently cleared by efferocytosis, can undergo secondary necrosis, with the potential for inflammation of surrounding tissues. Apoptosis may occur via the T cell granzyme B pathway. The role of granzyme B in NEA is not known. The aim of this study was to investigate production of granzyme B and its inhibitor proteinase inhibitor (PI)-9 by T cells from induced sputum and compare expression between eosinophilic, NEA and healthy controls. METHODS We investigated T cell intracellular granzyme B and its inhibitor, PI-9, in sputum from healthy control subjects (n = 10), and patients with NEA (n = 22) or eosinophilic asthma (EA) (n = 15) using flow cytometry. RESULTS Granzyme B expression and the ratio of granzyme B to PI-9 positive cells were highest in those with NEA for both CD3+ and CD4+ T cells. The expression of granzyme B was not statistically different between patients with NEA and EA; however, the ratio of granzyme B to PI-9 positive cells for CD3+ T cells was significantly higher in those with NEA compared with EA. CONCLUSIONS Induced sputum provides a non-invasive tool for investigating T cell cytotoxic mediators in the various asthma subtypes. Granzyme B expression is increased in NEA and the contribution of granzyme B to chronic inflammation requires further study.
Collapse
Affiliation(s)
- Jodie L Simpson
- Centre for Asthma and Respiratory Disease, The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter G Gibson
- Centre for Asthma and Respiratory Disease, The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Ian A Yang
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - John Upham
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Alan James
- Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Paul N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Sandra Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | | |
Collapse
|
34
|
Hecquet CM, Zhang M, Mittal M, Vogel SM, Di A, Gao X, Bonini MG, Malik AB. Cooperative interaction of trp melastatin channel transient receptor potential (TRPM2) with its splice variant TRPM2 short variant is essential for endothelial cell apoptosis. Circ Res 2013; 114:469-79. [PMID: 24337049 DOI: 10.1161/circresaha.114.302414] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Oxidants generated by activated endothelial cells are known to induce apoptosis, a pathogenic feature of vascular injury and inflammation from multiple pathogeneses. The melastatin-family transient receptor potential 2 (TRPM2) channel is an oxidant-sensitive Ca2+ permeable channel implicated in mediating apoptosis; however, the mechanisms of gating of the supranormal Ca2+ influx required for initiating of apoptosis are not understood. OBJECTIVE Here, we addressed the role of TRPM2 and its interaction with the short splice variant TRPM2 short variant (TRPM2-S) in mediating the Ca2+ entry burst required for induction of endothelial cell apoptosis. METHODS AND RESULTS We observed that TRPM2-S was basally associated with TRPM2 in the endothelial plasmalemma, and this interaction functioned to suppress TRPM2-dependent Ca2+ gating constitutively. Reactive oxygen species production in endothelial cells or directly applying reactive oxygen species induced protein kinase C-α activation and phosphorylation of TRPM2 at Ser 39. This in turn stimulated a large entry of Ca2+ and activated the apoptosis pathway. A similar TRPM2-dependent endothelial apoptosis mechanism was seen in intact vessels. The protein kinase C-α-activated phosphoswitch opened the TRPM2 channel to allow large Ca2+ influx by releasing TRPM2-S inhibition of TRPM2, which in turn activated caspase-3 and cleaved the caspase substrate poly(ADP-ribose) polymerase. CONCLUSIONS Here, we describe a fundamental mechanism by which activation of the trp superfamily TRPM2 channel induces apoptosis of endothelial cells. The signaling mechanism involves reactive oxygen species-induced protein kinase C-α activation resulting in phosphorylation of TRPM2-S that allows enhanced TRPM2-mediated gating of Ca2+ and activation of the apoptosis program. Strategies aimed at preventing the uncoupling of TRPM2-S from TRPM2 and subsequent Ca2+ gating during oxidative stress may mitigate endothelial apoptosis and its consequences in mediating vascular injury and inflammation.
Collapse
Affiliation(s)
- Claudie M Hecquet
- From the Department of Pharmacology and the Center for Lung and Vascular Biology (C.M.H., M.Z., M.M., S.M.V., A.D., X.G., M.G.B., A.B.M.) and Section of Cardiology (M.G.B.), College of Medicine, University of Illinois, Chicago
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Hirota JA, Hiebert PR, Gold M, Wu D, Graydon C, Smith JA, Ask K, McNagny K, Granville DJ, Knight DA. Granzyme B deficiency exacerbates lung inflammation in mice after acute lung injury. Am J Respir Cell Mol Biol 2013; 49:453-62. [PMID: 23642129 DOI: 10.1165/rcmb.2012-0512oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Granzyme B (GzmB) is a serine protease with intracellular and extracellular activities capable of regulating inflammation through cytokine processing and the apoptosis of effector cells. We tested the hypothesis that GzmB expression in T regulatory cells (Tregs) is required for the control of inflammatory responses and pathology during acute lung injury. To substantiate the clinical relevance of GzmB during lung injury, we performed GzmB immunohistochemistry on lung tissue from patients with acute respiratory distress syndrome (ARDS) and healthy control subjects. We also performed in vivo experiments with wild-type (WT) C57BL/6 and GzmB(-/-) mice exposed to a single intranasal instillation of bleomycin to model lung injury. Our results demonstrate that the expression of GzmB was elevated in ARDS lung sections, relative to healthy control samples. Bleomycin-exposed GzmB(-/-) mice exhibited greater morbidity and mortality, which was associated with increased numbers of lung lymphocytes. Bleomycin induced an equal increase in CD4(+)/CD25(+)/FoxP3(+) Treg populations in WT and GzmB(-/-) mice. GzmB expression was not significant in Tregs, with the majority of the expression localized to natural killer (NK)-1.1(+) cells. The expression of GzmB in NK cells of bleomycin-exposed WT mice was associated with greater lymphocyte apoptosis, reduced total lymphocyte numbers, and reduced pathology relative to GzmB(-/-) mice. Our data demonstrate that GzmB deficiency results in the exacerbation of lymphocytic inflammation during bleomycin-induced acute lung injury, which is associated with pathology, morbidity, and mortality.
Collapse
Affiliation(s)
- Jeremy A Hirota
- University of British Columbia James Hogg Research Centre, St. Paul's Hospital, 1081 Burrard St., Vancouver, BC, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Predescu DN, Bardita C, Tandon R, Predescu SA. Intersectin-1s: an important regulator of cellular and molecular pathways in lung injury. Pulm Circ 2013; 3:478-98. [PMID: 24618535 PMCID: PMC4070809 DOI: 10.1086/674439] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe syndromes resulting from the diffuse damage of the pulmonary parenchyma. ALI and ARDS are induced by a plethora of local or systemic insults, leading to the activation of multiple pathways responsible for injury, resolution, and repair or scarring of the lungs. Despite the large efforts aimed at exploring the roles of different pathways in humans and animal models and the great strides made in understanding the pathogenesis of ALI/ARDS, the only viable treatment options are still dependent on ventilator and cardiovascular support. Investigation of the pathophysiological mechanisms responsible for initiation and resolution or advancement toward lung scarring in ALI/ARDS animal models led to a better understanding of the disease's complexity and helped in elucidating the links between ALI and systemic multiorgan failure. Although animal models of ALI/ARDS have pointed out a variety of new ideas for study, there are still limited data regarding the initiating factors, the critical steps in the progression of the disease, and the central mechanisms dictating its resolution or progression to lung scarring. Recent studies link deficiency of intersectin-1s (ITSN-1s), a prosurvival protein of lung endothelial cells, to endothelial barrier dysfunction and pulmonary edema as well as to the repair/recovery from ALI. This review discusses the effects of ITSN-1s deficiency on pulmonary endothelium and its significance in the pathology of ALI/ARDS.
Collapse
Affiliation(s)
- Dan N Predescu
- 1 Department of Pharmacology, Rush University, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
37
|
Apoptotic and inflammatory signaling via Fas and tumor necrosis factor receptor I contribute to the development of chest trauma-induced septic acute lung injury. J Trauma Acute Care Surg 2013; 74:792-800. [PMID: 23425737 DOI: 10.1097/ta.0b013e31827a3655] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Direct acute lung injury (ALI) is still associated with a high mortality, whereas the underlying pathomechanisms are not yet fully understood. In this regard, epithelial cell death in the lungs has been attributed an important role in the pathogenesis of this clinical entity. Based on this background here, we hypothesized that signaling through Fas and tumor necrosis factor receptor 1 (TNFR-1) is involved in mediating apoptosis and inflammation in chest trauma induced septic ALI. METHODS Male C57BL/6 mice (wild-type [WT]), male mutant mice expressing nonfunctional Fas receptor (B6.MRL-Faslpr/J [lpr]) (lpr) and male TNFR-1-deficient mice (TNFR-1(-/-)) were subjected to a model of direct ALI consisting of blunt chest trauma followed by cecal ligation and puncture.Cytokine/chemokine concentrations of plasma, bronchoalveolar lavage (BAL) fluids, and lung tissue were investigated as well as BAL protein and lung myeloperoxidase. Lung histology was assessed; lung caspase 3, TUNEL-positive cells, and apoptotic polymorphonuclear neutrophil were measured, followed by a survival study. RESULTS Cytokine/chemokine levels in plasma, BAL, and lung tissue were markedly increased in WT animals following ALI, whereas lpr and TNFR-1((-/-) showed significantly decreased levels. BAL protein levels were substantially elevated following ALI, but lpr animals presented markedly diminished protein levels compared with WT and TNFR-1(-/-) animals. Lung myeloperoxidase level was only increased 12 hours after ALI in WT animals, whereas lung myeloperoxidase levels in lpr and TNFR-1(-/-) animals were not increased compared with sham. Lung histology revealed beneficial effects in lpr and TNFR-1(-/-). Lung active caspase 3 after ALI was substantially decreased in lpr and TNFR-1(-/-) mice compared with WT. Interestingly, an early but not persisting survival benefit was observed in lpr and TNFR-1 animals(-/-). CONCLUSION Pathomechanistically, Fas and TNFR-1 signaling contributed to the apoptotic and inflammatory response in a clinically relevant double-hit model of trauma-induced septic ALI. Moreover, this was associated with a temporary survival benefit.
Collapse
|
38
|
Herrero R, Tanino M, Smith LS, Kajikawa O, Wong VA, Mongovin S, Matute-Bello G, Martin TR. The Fas/FasL pathway impairs the alveolar fluid clearance in mouse lungs. Am J Physiol Lung Cell Mol Physiol 2013; 305:L377-88. [PMID: 23812636 DOI: 10.1152/ajplung.00271.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alveolar epithelial damage is a critical event that leads to protein-rich edema in acute lung injury (ALI), but the mechanisms leading to epithelial damage are not completely understood. Cell death by necrosis and apoptosis occurs in alveolar epithelial cells in the lungs of patients with ALI. Fas activation induces apoptosis of alveolar epithelial cells, but its role in the formation of lung edema is unclear. The main goal of this study was to determine whether activation of the Fas/Fas ligand pathway in the lungs could alter the function of the lung epithelium, and the mechanisms involved. The results show that Fas activation alters the alveolar barrier integrity and impairs the ability of the lung alveolar epithelium to reabsorb fluid from the air spaces. This result was dependent on the presence of a normal Fas receptor and was not affected by inflammation induced by Fas activation. Alteration of the fluid transport properties of the alveolar epithelium was partially restored by β-adrenergic stimulation. Fas activation also caused apoptosis of alveolar endothelial cells, but this effect was less pronounced than the effect on the alveolar epithelium. Thus, activation of the Fas pathway impairs alveolar epithelial function in mouse lungs by mechanisms involving caspase-dependent apoptosis, suggesting that targeting apoptotic pathways could reduce the formation of lung edema in ALI.
Collapse
Affiliation(s)
- Raquel Herrero
- Medical Research Service of the Veterans Affairs Puget Sound Health Care Center, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Compton C, McBryan D, Bucchioni E, Patalano F. The Novartis view on emerging drugs and novel targets for the treatment of chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2013; 26:562-73. [PMID: 23748050 DOI: 10.1016/j.pupt.2013.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/27/2013] [Indexed: 10/26/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease characterized by airflow limitation and chronic inflammation in the lungs. The mainstay of drug therapy for COPD is represented by long-acting bronchodilators, an important aspect of Novartis' development program. Novel once-daily dosing bronchodilators, such as the long-acting muscarinic antagonist (LAMA) glycopyrronium and the LAMA/long-acting β2-agonist (LABA) fixed-dose combination QVA149, have been shown to provide significant benefits to patients with COPD in terms of improvement in lung function, exercise tolerance, health-related quality of life, symptoms and reduction in the rate of exacerbations. Despite the benefits provided by these new treatment options, prevention of disease progression and control of exacerbations in certain patient phenotypes remain key challenges in the treatment of COPD. In order to address these needs and gain new insights into the complexity of COPD, Novartis is, in addition to bronchodilator-only therapies, developing LABA/inhaled corticosteroids (ICS) combinations to target inflammation, such as QMF149, as well as non-steroid based anti-inflammatory agents against key novel targets. These commitments are central to the Novartis' final goal of improving the standard of care in respiratory medicine and offering a better quality of life to patients with COPD.
Collapse
Affiliation(s)
- C Compton
- Novartis Pharma, Basel, Switzerland.
| | | | | | | |
Collapse
|
40
|
Deroost K, Tyberghein A, Lays N, Noppen S, Schwarzer E, Vanstreels E, Komuta M, Prato M, Lin JW, Pamplona A, Janse CJ, Arese P, Roskams T, Daelemans D, Opdenakker G, Van den Steen PE. Hemozoin Induces Lung Inflammation and Correlates with Malaria-Associated Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2013; 48:589-600. [DOI: 10.1165/rcmb.2012-0450oc] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
41
|
Wang X, Wang Y, Lee SJ, Kim HP, Choi AM, Ryter SW. Carbon monoxide inhibits Fas activating antibody-induced apoptosis in endothelial cells. Med Gas Res 2011; 1:8. [PMID: 22146483 PMCID: PMC3231877 DOI: 10.1186/2045-9912-1-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 05/18/2011] [Indexed: 12/18/2022] Open
Abstract
Background The extrinsic apoptotic pathway initiates when a death ligand, such as the Fas ligand, interacts with its cell surface receptor (ie., Fas/CD95), forming a death-inducing signaling complex (DISC). The Fas-dependent apoptotic pathway has been implicated in several models of lung or vascular injury. Carbon monoxide, an enzymatic product of heme oxygenase-1, exerts antiapoptotic effects at low concentration in vitro and in vivo. Methods Using mouse lung endothelial cells (MLEC), we examined the antiapoptotic potential of carbon monoxide against apoptosis induced by the Fas/CD95-activating antibody (Jo2). Carbon monoxide was applied to cell cultures in vitro. The expression and/or activation of apoptosis-related proteins and signaling intermediates were determined using Western Immunoblot and co-immunoprecipitation assays. Cell death was monitored by lactate dehydrogenase (LDH) release assays. Statistical significance was determined by student T-test and a value of P < 0.05 was considered significant. Results Treatment of MLEC with Fas-activating antibody (Jo2) induced cell death associated with the formation of the DISC, and activation of caspases (-8, -9, and -3), as well as the pro-apoptotic Bcl-2 family protein Bax. Exposure of MLEC to carbon monoxide inhibited Jo2-induced cell death, which correlated with the inhibition of DISC formation, cleavage of caspases-8, -9, and -3, and Bax activation. Carbon monoxide inhibited the phosphorylation of the Fas-associated death domain-containing protein, as well as its association with the DISC. Furthermore, carbon monoxide induced the expression of the antiapoptotic protein FLIP and increased its association with the DISC. CO-dependent cytoprotection against Fas mediated apoptosis in MLEC depended in part on activation of ERK1/2-dependent signaling. Conclusions Carbon monoxide has been proposed as a potential therapy for lung and other diseases based in part on its antiapoptotic effects in endothelial cells. In vitro, carbon monoxide may inhibit both Fas/caspase-8 and Bax-dependent apoptotic signaling pathways induced by Fas-activating antibody in endothelial cells. Strategies to block Fas-dependent apoptotic pathways may be useful in development of therapies for lung or vascular disorders.
Collapse
Affiliation(s)
- Xue Wang
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Herrero R, Kajikawa O, Matute-Bello G, Wang Y, Hagimoto N, Mongovin S, Wong V, Park DR, Brot N, Heinecke JW, Rosen H, Goodman RB, Fu X, Martin TR. The biological activity of FasL in human and mouse lungs is determined by the structure of its stalk region. J Clin Invest 2011; 121:1174-90. [PMID: 21285513 DOI: 10.1172/jci43004] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 12/08/2010] [Indexed: 01/20/2023] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition in critically ill patients. Injury to the alveolar epithelium is a critical event in ALI, and accumulating evidence suggests that it is linked to proapoptotic Fas/FasL signals. Active soluble FasL (sFasL) is detectable in the bronchoalveolar lavage (BAL) fluid of patients with ALI, but the mechanisms controlling its bioactivity are unclear. We therefore investigated how the structure of sFasL influences cellular activation in human and mouse lungs and the role of oxidants and proteases in modifying sFasL activity. The sFasL in BAL fluid from patients with ALI was bioactive and present in high molecular weight multimers and aggregates. Oxidants generated from neutrophil myeloperoxidase in BAL fluid promoted aggregation of sFasL in vitro and in vivo. Oxidation increased the biological activity of sFasL at low concentrations but degraded sFasL at high concentrations. The amino-terminal extracellular stalk region of human sFasL was required to induce lung injury in mice, and proteolytic cleavage of the stalk region by MMP-7 reduced the bioactivity of sFasL in human cells in vitro. The sFasL recovered from the lungs of patients with ALI contained both oxidized methionine residues and the stalk region. These data provide what we believe to be new insights into the structural determinants of sFasL bioactivity in the lungs of patients with ALI.
Collapse
Affiliation(s)
- Raquel Herrero
- Medical Research Service of the VA Puget Sound Health Center, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Perl M, Lomas-Neira J, Venet F, Chung CS, Ayala A. Pathogenesis of indirect (secondary) acute lung injury. Expert Rev Respir Med 2011; 5:115-26. [PMID: 21348592 PMCID: PMC3108849 DOI: 10.1586/ers.10.92] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
At present, therapeutic interventions to treat acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) remain largely limited to lung-protective strategies, as no real molecular-pathophysiologic-driven therapeutic intervention has yet become available. This is in part the result of the heterogeneous nature of the etiological processes that contribute to the state of ALI/ARDS. This article sets out to understand the development of ALI resulting from indirect pulmonary insults, such as extrapulmonary sepsis and trauma, shock, burn injury or mass transfusion, as opposed to direct pulmonary challenges, such as pneumonia, aspiration or lung contusion. Here, we consider not only the experimental and clinical data concerning the roles of various immune (neutrophil, macrophage, lymphocyte and dendritic) as well as nonimmune (epithelial and endothelial) cells in orchestrating the development of ALI resulting from indirect pulmonary stimuli, but also how these cell populations might be targeted therapeutically.
Collapse
Affiliation(s)
- Mario Perl
- Department of Traumatology, Hand and Reconstructive Surgery, University of Ulm Medical School, Ulm, Germany
| | - Joanne Lomas-Neira
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital and Brown University, Providence, RI, USA
| | - Fabienne Venet
- Hospices Civils de Lyon, Hôpital E. Herriot, Laboratoire d’Immunologie, Lyon, France
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital and Brown University, Providence, RI, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital and Brown University, Providence, RI, USA
| |
Collapse
|
44
|
Bahgat MM, Błazejewska P, Schughart K. Inhibition of lung serine proteases in mice: a potentially new approach to control influenza infection. Virol J 2011; 8:27. [PMID: 21251300 PMCID: PMC3034701 DOI: 10.1186/1743-422x-8-27] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 01/20/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Host serine proteases are essential for the influenza virus life cycle because the viral haemagglutinin is synthesized as a precursor which requires proteolytic maturation. Therefore, we studied the activity and expression of serine proteases in lungs from mice infected with influenza and evaluated the effect of serine protease inhibitors on virus replication both in cell culture and in infected mice. RESULTS Two different inbred mouse strains were investigated: DBA/2J as a highly susceptible and C57Bl/6J as a more resistant strain to influenza virus infection. The serine proteases from lung homogenates of mice exhibited pH optima of 10.00. Using the substrate Bz-Val-Gly-Arg-p-nitroanilide or in zymograms, the intensities of proteolysis increased in homogenates from both mouse strains with time post infection (p.i.) with the mouse-adapted influenza virus A/Puerto Rico/8/34 (H1N1; PR8). In zymograms at day 7 p.i., proteolytic bands were stronger and numerous in lung homogenates from DBA/2J than C57Bl/6J mice. Real-time PCR results confirmed differential expression of several lung proteases before and after infecting mice with the H1N1 virus. The most strongly up-regulated proteases were Gzma, Tmprss4, Elane, Ctrl, Gzmc and Gzmb. Pretreatment of mouse and human lung cell lines with the serine protease inhibitors AEBSF or pAB or a cocktail of both prior to infection with the H1N1 or the A/Seal/Massachusetts/1/80 (H7N7; SC35M) virus resulted in a decrease in virus replication. Pretreatment of C57Bl/6J mice with either AEBSF or a cocktail of AEBSF and pAB prior to infection with the H1N1 virus significantly reduced weight loss and led to a faster recovery of treated versus untreated mice while pAB alone exerted a very poor effect. After infection with the H7N7 virus, the most significant reduction of weight loss was obtained upon pretreatment with either the protease inhibitor cocktail or pAB. Furthermore, pretreatment of C57BL/6J mice with AEBSF prior to infection resulted in a significant reduction in the levels of both the H1N1 and H7N7 nucleoproteins in mice lungs and also a significant reduction in the levels of the HA transcript in the lungs of the H1N1--but not the H7N7-infected mice. CONCLUSION Multiple serine protease activities might be implicated in mediating influenza infection. Blocking influenza A virus infection in cultured lung epithelia and in mice by the used serine protease inhibitors may provide an alternative approach for treatment of influenza infection.
Collapse
Affiliation(s)
- Mahmoud M Bahgat
- Department of Infection Genetics and University of Veterinary Medicine Hannover, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | | | | |
Collapse
|
45
|
Urbanowicz RA, Lamb JR, Todd I, Corne JM, Fairclough LC. Enhanced effector function of cytotoxic cells in the induced sputum of COPD patients. Respir Res 2010; 11:76. [PMID: 20540777 PMCID: PMC2891678 DOI: 10.1186/1465-9921-11-76] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 06/11/2010] [Indexed: 11/25/2022] Open
Abstract
Background We have previously shown that NK (CD56+CD3-) and NKT-like (CD56+CD3+) cells are reduced in both numbers and cytotoxicity in peripheral blood. The aim of the present study was to investigate their numbers and function within induced sputum. Methods Induced sputum cell numbers and intracellular granzyme B and perforin were analysed by flow cytometry. Immunomagnetically selected CD56+ cells (NK and NKT-like cells) were used in an LDH release assay to determine cytotoxicity. Results The proportion of NK cells and NKT-like cells in smokers with COPD (COPD subjects) was significantly higher (12.7% and 3%, respectively) than in healthy smokers (smokers) (5.7%, p < 0.01; 1%, p < 0.001) and non-smoking healthy subjects (HNS) (4.2%, p < 0.001; 0.8%, p < 0.01). The proportions of NK cells and NKT-like cells expressing both perforin and granzyme B were also significantly higher in COPD subjects compared to smokers and HNS. CD56+ cells from COPD subjects were significantly more cytotoxic (1414 biological lytic activity) than those from smokers (142.5; p < 0.01) and HNS (3.8; p < 0.001) and were inversely correlated to FEV1. (r = -0.75; p = 0.0098). Conclusion We have shown an increased proportion of NK and NKT-like cells in the induced sputum of COPD subjects and have demonstrated that these cells are significantly more cytotoxic in COPD subjects than smokers and HNS.
Collapse
Affiliation(s)
- Richard A Urbanowicz
- COPD Research Group, Nottingham Respiratory Biomedical Research Unit, The University of Nottingham, UK
| | | | | | | | | |
Collapse
|
46
|
Therapeutic accessibility of caspase-mediated cell death as a key pathomechanism in indirect acute lung injury*. Crit Care Med 2010; 38:1179-86. [DOI: 10.1097/ccm.0b013e3181d4563f] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
47
|
Galani V, Tatsaki E, Bai M, Kitsoulis P, Lekka M, Nakos G, Kanavaros P. The role of apoptosis in the pathophysiology of Acute Respiratory Distress Syndrome (ARDS): An up-to-date cell-specific review. Pathol Res Pract 2010; 206:145-50. [DOI: 10.1016/j.prp.2009.12.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 12/03/2009] [Indexed: 01/01/2023]
|
48
|
Lopez AD, Avasarala S, Grewal S, Murali AK, London L. Differential role of the Fas/Fas ligand apoptotic pathway in inflammation and lung fibrosis associated with reovirus 1/L-induced bronchiolitis obliterans organizing pneumonia and acute respiratory distress syndrome. THE JOURNAL OF IMMUNOLOGY 2010; 183:8244-57. [PMID: 20007588 DOI: 10.4049/jimmunol.0901958] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bronchiolitis obliterans organizing pneumonia (BOOP) and acute respiratory distress syndrome (ARDS) are two clinically and histologically distinct syndromes sharing the presence of an inflammatory and fibrotic component. Apoptosis via the Fas/Fas ligand (FasL) pathway plays an important role in the development of acute lung injury and fibrosis characteristic of these and other pulmonary inflammatory and fibrotic syndromes. We evaluated the role of apoptosis via the Fas/FasL pathway in the development of pulmonary inflammation and fibrosis in reovirus 1/L-induced BOOP and ARDS. CBA/J mice were intranasally inoculated with saline, 1 x 10(6) (BOOP), or 1 x 10(7) (ARDS) PFU reovirus 1/L, and evaluated at various days postinoculation for in situ apoptosis by TUNEL analysis and Fas/FasL expression. Our results demonstrate the presence of apoptotic cells and up-regulation of Fas/FasL expression in alveolar epithelium and in infiltrating cells during the inflammatory and fibrotic stages of both reovirus 1/L-induced ARDS and BOOP. Treatment of mice with the caspase 8 inhibitor, zIETD-fmk, inhibited apoptosis, inflammation, and fibrotic lesion development in reovirus 1/L-induced BOOP and ARDS. However, CBA/KlJms-Fas(lpr-cg)/J mice, which carry a point mutation in the Fas cytoplasmic region that abolishes the ability of Fas to transduce an apoptotic signal, do not develop pulmonary inflammation and fibrotic lesions associated with reovirus 1/L-induced BOOP, but still develop inflammation and fibrotic lesions associated with reovirus 1/L-induced ARDS. These results suggest a differential role for the Fas/FasL apoptotic pathway in the development of inflammation and fibrotic lesions associated with BOOP and ARDS.
Collapse
Affiliation(s)
- Andrea D Lopez
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
49
|
Bem RA, van Woensel JBM, Lutter R, Domachowske JB, Medema JP, Rosenberg HF, Bos AP. Granzyme A- and B-cluster deficiency delays acute lung injury in pneumovirus-infected mice. THE JOURNAL OF IMMUNOLOGY 2009; 184:931-8. [PMID: 20018616 DOI: 10.4049/jimmunol.0903029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lower respiratory tract infection by the human pneumovirus respiratory syncytial virus is a frequent cause of acute lung injury in children. Severe pneumovirus disease in humans is associated with activation of the granzyme pathway by effector lymphocytes, which may promote pathology by exaggerating proapoptotic caspase activity and proinflammatory activity. The main goal of this study was to determine whether granzymes contribute to the development of acute lung injury in pneumovirus-infected mice. Granzyme-expressing mice and granzyme A- and B-cluster single- and double-knockout mice were inoculated with the rodent pneumovirus pneumonia virus of mice strain J3666, and were studied for markers of lung inflammation and injury. Expression of granzyme A and B is detected in effector lymphocytes in mouse lungs in response to pneumovirus infection. Mice deficient for granzyme A and the granzyme B cluster have unchanged virus titers in the lungs but show a significantly delayed clinical response to fatal pneumovirus infection, a feature that is associated with delayed neutrophil recruitment, diminished activation of caspase-3, and reduced lung permeability. We conclude that granzyme A- and B-cluster deficiency delays the acute progression of pneumovirus disease by reducing alveolar injury.
Collapse
Affiliation(s)
- Reinout A Bem
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
50
|
|