1
|
Wang T, Shuai P, Wang Q, Guo C, Huang S, Li Y, Wu W, Yi L. α‑1 Antitrypsin is a potential target of inflammation and immunomodulation (Review). Mol Med Rep 2025; 31:107. [PMID: 40017119 PMCID: PMC11881679 DOI: 10.3892/mmr.2025.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/12/2024] [Indexed: 03/01/2025] Open
Abstract
α‑1 Antitrypsin (AAT) is an acute phase protein encoded by the serine protease inhibitor family A member 1 gene. This multifunctional protein serves several roles, including anti‑inflammatory, antibacterial, antiapoptotic and immune regulatory functions. The primary role of AAT is to protect tissues and organs from protease‑induced damage due to its function as a serine protease inhibitor. AAT is associated with the development of lung inflammation, liver inflammation and immune‑mediated inflammatory diseases, which are influenced by environmental and genetic factors. For instance, AAT acts as an anti‑inflammatory protein to prevent and reverse type I diabetes. The present study briefly reviewed the molecular properties and mechanisms of AAT, as well as advances in the study of lung, liver and inflammatory diseases associated with AAT. The potential of AAT as a diagnostic and therapeutic target for inflammatory and immune‑mediated inflammatory diseases was reviewed. In addition, the damaging and protective effects of AAT, and its effects on organ function were discussed.
Collapse
Affiliation(s)
- Tiantian Wang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Peimeng Shuai
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qingyu Wang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Caimao Guo
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuqi Huang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuanyuan Li
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wenyu Wu
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lan Yi
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
2
|
Shen Y, Leng L, Hu Y. Exploring Core Genes Associated with Sepsis and Systemic Inflammatory Response Syndrome Using Single-Cell Sequencing Technology. J Inflamm Res 2025; 18:1815-1838. [PMID: 39935525 PMCID: PMC11811729 DOI: 10.2147/jir.s448900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Purpose As a crucial aspect of emergency critical medicine, sepsis has been in a difficult stage. As its "preparatory stage", SIRS has attracted the attention of the medical workers all over the world. The frequency of occurrence is on the rise, but there is a lack of certain indicators for the timely detection and recognition of illnesses. Methods By virtue of scRNA-seq, this research has analyzed single-cell transcriptome data from samples taken from groups with septic death and systemic inflammatory response syndrome so as to identify the unique markers and patterns in immune response. Results By revealing the status of twelve cell clusters of four major cell types in blood samples through UMAP cell clustering and the differences of major cell populations between the dead and SIRS patients, the results have elucidated the components of different cells and their marker genes in two disease states, and the response mechanism beneficial to disease diagnosis in blood samples. Conclusion By establishing a theoretical framework centered on cellular and molecular regulation, the study has introduced a novel approach for diagnosing and treating sepsis death group and SIRS patients early, as well as differentiating and preventing these conditions.
Collapse
Affiliation(s)
- YuZhou Shen
- Department of Emergency Medicine, the Affiliated Hospital of Southwest Medical University, Lu Zhou, Sichuan, People’s Republic of China
| | - LingHan Leng
- Department of Intensive Care Unit, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, People’s Republic of China
| | - YingChun Hu
- Department of Emergency Medicine, the Affiliated Hospital of Southwest Medical University, Lu Zhou, Sichuan, People’s Republic of China
| |
Collapse
|
3
|
Rojas-Quintero J, Ochsner SA, Lee HS, Cong C, Waich Cohen A, Colborg AS, Tsoyi K, Basil MC, Cantu E, Rosas IO, McKenna NJ, San-José Estépar R, Barjaktarevic I, Wilson AA, Polverino F. Skewed adaptive immune responses are involved in alpha-1 antitrypsin deficiency emphysema. Eur Respir J 2025; 65:2400839. [PMID: 39401853 PMCID: PMC11695135 DOI: 10.1183/13993003.00839-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024]
Affiliation(s)
- Joselyn Rojas-Quintero
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Christine Cong
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Alan Waich Cohen
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Adrianne S Colborg
- Department of Pulmonary Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Konstantin Tsoyi
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Maria C Basil
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan O Rosas
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Raúl San-José Estépar
- Applied Chest Imaging Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Andrew A Wilson
- Center for Regenerative Medicine (CreM) of Boston University and Boston Medical Center, Boston, MA, USA
| | - Francesca Polverino
- Pulmonary Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Khedoe PPSJ, van 't Wout EFA. Buddy, bystander or betrayer: B cells in lymphoid aggregates in AATD emphysema. Eur Respir J 2025; 65:2402163. [PMID: 39746767 DOI: 10.1183/13993003.02163-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025]
Affiliation(s)
- P Padmini S J Khedoe
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emily F A van 't Wout
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Abo KM, Merritt C, Basil MC, Lin SM, Cantu E, Morley MP, Bawa P, Gallagher M, Byers DE, Morrisey EE, Wilson AA. Pulmonary Cellular Toxicity in Alpha-1 Antitrypsin Deficiency. Chest 2024; 166:472-479. [PMID: 38360172 PMCID: PMC11443245 DOI: 10.1016/j.chest.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/09/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024] Open
Affiliation(s)
- Kristine M Abo
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Carly Merritt
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Susan M Lin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Edward Cantu
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Pushpinder Bawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Marissa Gallagher
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA.
| |
Collapse
|
6
|
Pintanel-Raymundo M, Menao-Guillén S, Perales-Afán JJ, García-Gutiérrez A, Moreno-Gázquez I, Julián-Ansón M, Ramos-Álvarez M, Olivera-González S, Gutiérrez-Cía I, Torralba-Cabeza MA. Analysis of the expression of the Serpina1 gene in SARS-CoV-2 infection: study of a new biomarker. Rev Clin Esp 2024; 224:253-258. [PMID: 38608729 DOI: 10.1016/j.rceng.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/13/2024] [Indexed: 04/14/2024]
Abstract
INTRODUCTION The SERPINA1 gene encodes the protein Alpha-1 Antitrypsin (AAT1). Possible imbalances between the concentrations of proteases and antiproteases (AAT1) can lead to the development of serious pulmonary and extrapulmonary pathologies. In this work we study the importance of this possible imbalance in patients with COVID-19. OBJECTIVES To correlate the severity of the symptoms of SARS-COV-2 infection with the AAT1 concentrations at diagnosis of the disease. METHODS An observational, prospective, cross-sectional, non-interventional, analytical study was carried out where 181 cases with COVID-19 admitted to the "Lozano Blesa" University Clinical Hospital of Zaragoza were selected. The concentration of AAT1 was studied in all of them and this was correlated with the clinical aspects and biochemical parameters at hospital admission. RESULTS 141 cases corresponded to patients with severe COVID and 40 patients with mild COVID. AAT1 levels were positively correlated with the days of hospitalization, severity, C-Reactive Protein, ferritin, admission to Intensive Care, and death, and presented a negative correlation with the number of lymphocytes/mm3. AAT1 concentrations higher than 237.5 mg/dL allowed the patient to be classified as "severe" (S72%; E78%) and 311.5 mg/dL were associated with the risk of admission to Intensive Care or Exitus (S67%; E79%). CONCLUSIONS Levels of the SERPINA1 gene expression product, AAT1, correlate with the severity of COVID-19 patients at diagnosis of the disease, being useful as a prognostic biomarker.
Collapse
Affiliation(s)
| | - S Menao-Guillén
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España
| | - J J Perales-Afán
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España
| | - A García-Gutiérrez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - I Moreno-Gázquez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M Julián-Ansón
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M Ramos-Álvarez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - S Olivera-González
- Servicio de Medicina Interna, Hospital Marina Salud de Denia, Alicante, Spain
| | - I Gutiérrez-Cía
- Servicio de Cuidados Intensivos, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M A Torralba-Cabeza
- Facultad de Medicina, Universidad de Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España; Unidad de Enfermedades Minoritarias, Servicio de Medicina Interna, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Grupo de Trabajo de Enfermedades Minoritarias de la Sociedad Española de Medicina Interna, España.
| |
Collapse
|
7
|
Maslakova AA, Golyshev SA, Potashnikova DM, Moisenovich AM, Orlovsky IV, Smirnova OV, Rubtsov MA. SERPINA1 long transcripts produce non-secretory alpha1-antitrypsin isoform: In vitro translation in living cells. Int J Biol Macromol 2023; 241:124433. [PMID: 37086761 DOI: 10.1016/j.ijbiomac.2023.124433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/24/2023]
Abstract
SERPINA1 is a well-studied serpin gene due to its dramatic impact on human health. Translation initiation at the main SERPINA1 start codon produces the only known alpha1-antitrypsin (AAT) isoform intended for secretion. AAT performs essential functions by inhibiting proteases and modulating immunity. However, SERPINA1 expression at the level of translation is not sufficiently studied. Here we hypothesize that the main SERPINA1 ORF can be alternatively translated, producing a non-secretory AAT isoform by either masking or excluding a signal peptide. We defined SERPINA1 long mRNA isoforms specific for prostate (DU145) and liver (HepG2) cell lines and studied their individual expression by in vitro assay. We found that all long transcripts produce both glycosylated secretory AAT-eGFP fusion protein and non-glycosylated intracellular AAT-eGFP (initiated from an alternative AUG-2 start codon), with the proportion regulated by the SERPINA1 5'-UTR. Both fusion proteins localize to distinct cellular compartments: in contrast to a fusion with the secretory AAT accumulating in the ER, the intracellular one exhibits nuclear-cytoplasmic shuttling. We detected putative endogenous AAT isoform enriching the nuclear speckles. CONCLUSION: Alternative translation initiation might be a mechanism through which SERPINA1 expands the biological diversity of its protein products. Our findings open up new prospects for the study of SERPINA1 gene expression.
Collapse
Affiliation(s)
- A A Maslakova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia.
| | - S A Golyshev
- A.N. Belozersky Institute of Physical and Chemical Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119992, Russia
| | - D M Potashnikova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
| | - A M Moisenovich
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
| | - I V Orlovsky
- Research Institute of Molecular and Cellular Medicine, Рeoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya, Moscow 117198, Russia
| | - O V Smirnova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
| | - M A Rubtsov
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia; Center for Industrial Technologies and Entrepreneurship, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya, Moscow 119991, Russia
| |
Collapse
|
8
|
Kokturk N, Khodayari N, Lascano J, Riley EL, Brantly ML. Lung Inflammation in alpha-1-antitrypsin deficient individuals with normal lung function. Respir Res 2023; 24:40. [PMID: 36732772 PMCID: PMC9893669 DOI: 10.1186/s12931-023-02343-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alpha-1-antitrypsin deficient (AATD) individuals are prone to develop early age of onset chronic obstructive pulmonary disease (COPD) more severe than non-genetic COPD. Here, we investigated the characteristics of lower respiratory tract of AATD individuals prior to the onset of clinically significant COPD. METHODS Bronchoalveolar lavage was performed on 22 AATD with normal lung function and 14 healthy individuals. Cell counts and concentrations of proteases, alpha-1-antitrypsin and proinflammatory mediators were determined in the bronchoalveolar lavage fluid from study subjects. In order to determine the airway inflammation, we also analyzed immune cell components of the large airways from bronchial biopsies using immunohistochemistry in both study subjects. Finally, we made comparisons between airway inflammation and lung function rate of decline using four repeated lung function tests over one year in AATD individuals. RESULTS AATD individuals with normal lung function had 3 folds higher neutrophil counts, 2 folds increase in the proteases levels, and 2-4 folds higher levels of IL-8, IL-6, IL-1β, and leukotriene B4 in their epithelial lining fluid compared to controls. Neutrophil elastase levels showed a positive correlation with the levels of IL-8 and neutrophils in AATD epithelial lining fluid. AATD individuals also showed a negative correlation of baseline FEV1 with neutrophil count, neutrophil elastase, and cytokine levels in epithelial lining fluid (p < 0.05). In addition, we observed twofold increase in the number of lymphocytes, macrophages, neutrophils, and mast cells of AATD epithelial lining fluid as compared to controls. CONCLUSION Mild inflammation is present in the lower respiratory tract and airways of AATD individuals despite having normal lung function. A declining trend was also noticed in the lung function of AATD individuals which was correlated with pro-inflammatory phenotype of their lower respiratory tract. This results suggest the presence of proinflammatory phenotype in AATD lungs. Therefore, early anti-inflammatory therapies may be a potential strategy to prevent progression of lung disease in AATD individuals.
Collapse
Affiliation(s)
- Nurdan Kokturk
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
- Department of Pulmonary and Critical Care, Gazi University School of Medicine, Ankara, Turkey
| | - Nazli Khodayari
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
| | - Jorge Lascano
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
| | | | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA.
| |
Collapse
|
9
|
Izquierdo M, Rawal H, Armstrong M, Marion CR. Alpha-1 Asthma Overlap Syndrome: a Clinical Overview. Curr Allergy Asthma Rep 2022; 22:101-111. [PMID: 35596100 DOI: 10.1007/s11882-022-01036-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Alpha-1 antitrypsin deficiency (AATD) is one of the most common genetic diseases that is associated with severe complications and yet remains underdiagnosed. The pulmonary symptoms of both AATD and asthma include cough, excessive sputum production, dyspnea, and wheezing. These symptoms overlap significantly leading to difficulty distinguishing between these two conditions and suspicion that there may be an overlap syndrome. We aim to discuss the pathophysiology, clinical manifestations, and treatment of both alpha-1 antitrypsin and asthma and how they may overlap. RECENT FINDINGS Recent literature suggests that there is an association between asthma and AATD. This association has been hypothesized to be secondary to an imbalance of elastase and anti-elastase leading to a pro-inflammatory state in patients with AATD. This review serves to overview the pathophysiology, clinical manifestations, and treatment of alpha-1 antitrypsin, asthma, and the increasingly recognized intersection of the two, AATD-asthma overlap syndrome.
Collapse
Affiliation(s)
- Manuel Izquierdo
- Department of Internal Medicine, Section On Pulmonary, Critical Care, Immunologic, and Asthma Medicine, Wake Forest Baptist Hospital, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Himanshu Rawal
- Department of Internal Medicine, Section On Pulmonary, Critical Care, Immunologic, and Asthma Medicine, Wake Forest Baptist Hospital, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Michael Armstrong
- Department of Internal Medicine, Wake Forest Baptist Hospital, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Chad R Marion
- Department of Internal Medicine, Section On Pulmonary, Critical Care, Immunologic, and Asthma Medicine, Wake Forest Baptist Hospital, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA. .,Department On Internal Medicine, Section On Pulmonary, Critical Care and Sleep Medicine, W. G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, NC, USA.
| |
Collapse
|
10
|
Li MH, Marty-Santos LM, van Ginkel PR, McDermott AE, Rasky AJ, Lukacs NW, Wellik DM. The Lung Elastin Matrix Undergoes Rapid Degradation Upon Adult Loss of Hox5 Function. Front Cell Dev Biol 2021; 9:767454. [PMID: 34901011 PMCID: PMC8662386 DOI: 10.3389/fcell.2021.767454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Hox genes encode transcription factors that are critical for embryonic skeletal patterning and organogenesis. The Hoxa5, Hoxb5, and Hoxc5 paralogs are expressed in the lung mesenchyme and function redundantly during embryonic lung development. Conditional loss-of-function of these genes during postnatal stages leads to severe defects in alveologenesis, specifically in the generation of the elastin network, and animals display bronchopulmonary dysplasia (BPD) or BPD-like phenotype. Here we show the surprising results that mesenchyme-specific loss of Hox5 function at adult stages leads to rapid disruption of the mature elastin matrix, alveolar enlargement, and an emphysema-like phenotype. As the elastin matrix of the lung is considered highly stable, adult disruption of the matrix was not predicted. Just 2 weeks after deletion, adult Hox5 mutant animals show significant increases in alveolar space and changes in pulmonary function, including reduced elastance and increased compliance. Examination of the extracellular matrix (ECM) of adult Tbx4rtTA; TetOCre; Hox5a f a f bbcc lungs demonstrates a disruption of the elastin network although the underlying fibronectin, interstitial collagen and basement membrane appear unaffected. An influx of macrophages and increased matrix metalloproteinase 12 (MMP12) are observed in the distal lung 3 days after Hox5 deletion. In culture, fibroblasts from Hox5 mutant lungs exhibit reduced adhesion. These findings establish a novel role for Hox5 transcription factors as critical regulators of lung fibroblasts at adult homeostasis.
Collapse
Affiliation(s)
- Mu-Hang Li
- Genetics Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Leilani M. Marty-Santos
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Paul R. van Ginkel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Aubrey E. McDermott
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Andrew J. Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
11
|
Aust AC, Benesova E, Vidova V, Coufalikova K, Smetanova S, Borek I, Janku P, Budinska E, Klanova J, Thon V, Spacil Z. Profiling Tryptophan Catabolites of Human Gut Microbiota and Acute-Phase Protein Levels in Neonatal Dried Blood Specimens. Front Microbiol 2021; 12:665743. [PMID: 34777268 PMCID: PMC8581761 DOI: 10.3389/fmicb.2021.665743] [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: 02/08/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022] Open
Abstract
National screening programs use dried blood specimens to detect metabolic disorders or aberrant protein functions that are not clinically evident in the neonatal period. Similarly, gut microbiota metabolites and immunological acute-phase proteins may reveal latent immune aberrations. Microbial metabolites interact with xenobiotic receptors (i.e., aryl hydrocarbon and pregnane-X) to maintain gastrointestinal tissue health, supported by acute-phase proteins, functioning as sensors of microbial immunomodulation and homeostasis. The delivery (vaginal or cesarean section) shapes the microbial colonization, which substantially modulates both the immune system’s response and mucosal homeostasis. This study profiled microbial metabolites of the kynurenine and tryptophan pathway and acute-phase proteins in 134 neonatal dried blood specimens. We newly established neonatal blood levels of microbial xenobiotic receptors ligands (i.e., indole-3-aldehyde, indole-3-butyric acid, and indole-3-acetamide) on the second day of life. Furthermore, we observed diverse microbial metabolic profiles in neonates born vaginally and via cesarean section, potentially due to microbial immunomodulatory influence. In summary, these findings suggest the supportive role of human gut microbiota in developing and maintaining immune system homeostasis.
Collapse
Affiliation(s)
| | - Eliska Benesova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Veronika Vidova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | | | - Sona Smetanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Ivo Borek
- Department of Pediatrics, University Hospital Brno and Masaryk University Medical School, Brno, Czechia
| | - Petr Janku
- Department of Gynecology and Obstetrics, University Hospital Brno and Masaryk University Medical School, Brno, Czechia
| | - Eva Budinska
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Jana Klanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Vojtech Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Zdenek Spacil
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
12
|
Vass L, Fisk M, Cheriyan J, Mohan D, Forman J, Oseni A, Devaraj A, Mäki-Petäjä KM, McEniery CM, Fuld J, Hopkinson NS, Lomas DA, Cockcroft JR, Tal-Singer R, Polkey MI, Wilkinson IB. Quantitative 18F-fluorodeoxyglucose positron emission tomography/computed tomography to assess pulmonary inflammation in COPD. ERJ Open Res 2021; 7:00699-2020. [PMID: 34476245 PMCID: PMC8405867 DOI: 10.1183/23120541.00699-2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/12/2021] [Indexed: 11/07/2022] Open
Abstract
Rationale COPD and smoking are characterised by pulmonary inflammation. 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) imaging may improve knowledge of pulmonary inflammation in COPD patients and aid early development of novel therapies as an imaging biomarker. Objectives To evaluate pulmonary inflammation, assessed by FDG uptake, in whole and regional lung in “usual” (smoking-related) COPD patients, alpha-1 antitrypsin deficiency (α1ATD) COPD patients, smokers without COPD and never-smokers using FDG PET/CT. Secondly, to explore cross-sectional associations between FDG PET/CT and systemic inflammatory markers in COPD patients and repeatability of the technique in COPD patients. Methods Data from two imaging studies were evaluated. Pulmonary FDG uptake (normalised Ki; nKi) was measured by Patlak graphical analysis in four subject groups: 84 COPD patients, 11 α1ATD-COPD patients, 12 smokers and 10 never-smokers. Within the COPD group, associations between nKi and systemic markers of inflammation were assessed. Repeatability was evaluated in 32 COPD patients comparing nKi values at baseline and at 4-month follow-up. Results COPD patients, α1ATD-COPD patients and smokers had increased whole lung FDG uptake (nKi) compared with never-smokers (0.0037±0.001, 0.0040±0.001, 0.0040±0.001 versus 0.0028±0.001 mL·cm−3·min−1, respectively, p<0.05 for all). Similar results were observed in upper and middle lung regions. In COPD participants, plasma fibrinogen was associated with whole lung nKi (β=0.30, p=0.02) in multivariate analysis adjusted for current smoking, forced expiratory volume in 1 s % predicted, systemic neutrophils and C-reactive protein levels. Mean percentage difference in nKi between the baseline and follow-up was 3.2%, and the within subject coefficient of variability was 7.7%. Conclusions FDG PET/CT has potential as a noninvasive tool to enable whole lung and regional quantification of FDG uptake to assess smoking- and COPD-related pulmonary inflammation. FDG PET/CT has potential utility to noninvasively evaluate pulmonary inflammation in COPD. Pulmonary FDG uptake is increased in COPD patients, positively associated with systemic inflammatory markers and shows low inter-occasion variability.https://bit.ly/3dELYAW
Collapse
Affiliation(s)
- Laurence Vass
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.,These authors contributed equally
| | - Marie Fisk
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.,These authors contributed equally
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.,Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Julia Forman
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Adelola Oseni
- Dept of Radiology, St George's Hospital NHS Trust, London, UK
| | - Anand Devaraj
- National Heart and Lung Institute, Imperial College, London, UK
| | - Kaisa M Mäki-Petäjä
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Jonathan Fuld
- Division of Respiratory Medicine, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - David A Lomas
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - John R Cockcroft
- Dept of Cardiology, Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | | | | | - Ian B Wilkinson
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| |
Collapse
|
13
|
Chen YH, Cheadle CE, Rice LV, Pfeffer PE, Dimeloe S, Gupta A, Bush A, Gooptu B, Hawrylowicz CM. The Induction of Alpha-1 Antitrypsin by Vitamin D in Human T Cells Is TGF-β Dependent: A Proposed Anti-inflammatory Role in Airway Disease. Front Nutr 2021; 8:667203. [PMID: 34458299 PMCID: PMC8397538 DOI: 10.3389/fnut.2021.667203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Vitamin D upregulates anti-inflammatory and antimicrobial pathways that promote respiratory health. Vitamin D synthesis is initiated following skin exposure to sunlight, however nutritional supplementation can be required to address deficiency, for example during the winter months or due to cultural constraints. We recently reported that 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) treatment induced alpha-1 antitrypsin (AAT) expression in CD4+, but not CD8+ T cells, with evidence supporting an immunoregulatory role. Research Question: To understand the relationship between vitamin D, lung AAT levels and T lymphocytes further we investigated whether TGF-β is required as a co-factor for 1,25(OH)2D3-induced upregulation of AAT by vitamin D in CD8+ T cells in vitro and correlated circulating vitamin D levels with lung AAT levels in vivo. Results: 1,25(OH)2D3 in combination with TGF-β1 increased AAT expression by CD8+ T cells, as well as VDR and RXRα gene expression, which may partly explain the requirement for TGF-β. CD4+ T cells may also require autocrine stimulation with TGF-β as a co-factor since 1,25(OH)2D3 was associated with increased TGF-β bioactivity and neutralisation of TGF-β partially abrogated 1,25(OH)2D3-induced SERPINA1 gene expression. Neither CD4+ nor CD8+ T cells responded to the circulating vitamin D precursor, 25-hydroxyvitamin D3 for induction of SERPINA1, suggesting that local generation of 1,25(OH)2D3 is required. Transcriptional gene profiling studies previously demonstrated that human bronchial epithelial cells rapidly increased TGF-β2 gene expression in response to 1,25(OH)2D3. Here, human epithelial cells responded to precursor 25(OH)D3 to increase bioactive TGF-β synthesis. CD8+ T cells responded comparably to TGF-β1 and TGF-β2 to increase 1,25(OH)2D3-induced AAT. However, CD8+ T cells from adults with AAT-deficiency, homozygous for the Z allele of SERPINA1, were unable to mount this response. AAT levels in the airways of children with asthma and controls correlated with circulating 25(OH)D3. Conclusions: Vitamin D increases AAT expression in human T cells and this response is impaired in T cells from individuals homozygous for the Z allele of SERPINA1 in a clinic population. Furthermore, a correlation between circulating vitamin D and airway AAT is reported. We propose that vitamin D-induced AAT contributes to local immunomodulation and airway health effects previously attributed to vitamin D.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Charlotte E Cheadle
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Medical Research Council and Asthma UK Centre for Allergic Mechanisms of Asthma, Guy's Hospital, King's College London, London, United Kingdom
| | - Louise V Rice
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Medical Research Council and Asthma UK Centre for Allergic Mechanisms of Asthma, Guy's Hospital, King's College London, London, United Kingdom
| | - Paul E Pfeffer
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Medical Research Council and Asthma UK Centre for Allergic Mechanisms of Asthma, Guy's Hospital, King's College London, London, United Kingdom
| | - Sarah Dimeloe
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Atul Gupta
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,National Heart and Lung Institute, Royal Brompton & Harefield National Health Service Foundation Trust, London, United Kingdom
| | - Andrew Bush
- Centre for Paediatrics and Child Health, National Heart and Lung Institute, Imperial College, Royal Brompton Hospital, London, United Kingdom
| | - Bibek Gooptu
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,National Institute for Health Research Leicester Biomedical Research Centre-Respiratory and Leicester Institute of Structural & Chemical Biology, University of Leicester, Leicester, United Kingdom.,London Alpha-1 Antitrypsin Deficiency Service, Royal Free Hospital, London, United Kingdom
| | - Catherine M Hawrylowicz
- Peter Gorer Department of Immunobiology (Formerly Asthma, Allergy and Lung Biology), School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Medical Research Council and Asthma UK Centre for Allergic Mechanisms of Asthma, Guy's Hospital, King's College London, London, United Kingdom
| |
Collapse
|
14
|
Boreika R, Sitkauskiene B. Interleukin-32 in Pathogenesis of Atopic Diseases: Proinflammatory or Anti-Inflammatory Role? J Interferon Cytokine Res 2021; 41:235-243. [PMID: 34280028 DOI: 10.1089/jir.2020.0230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Atopic diseases, such as atopic dermatitis (AD), allergic asthma (AA), and allergic rhinitis (AR), are increasingly becoming a worldwide issue. This atopic triad originates at an early age and on a multifactorial basis, causing significant discomfort to susceptible individuals. The global case number is now reaching new highs, so exploring immune system regulation and its components is becoming critical. One cytokine, interleukin-32 (IL-32), is involved in inflammation and regulation of the immune system. It has nine isoforms that show varying degrees of expression, both intracellularly and extracellularly. IL-32 is secreted by immune cells, such as monocytes, macrophages, natural killer cells, and T cells, and by nonimmune cells, including fibroblasts, keratinocytes, and endothelial cells. Its production is regulated and augmented by microorganisms, mitogens, and other cytokines. Early studies demonstrated that IL-32 was an immune regulator that functioned to protect against inflammatory diseases, including AD, AA, and AR, and proposed a proinflammatory role for IL-32 in immune regulation and symptom exacerbation. However, several later reports suggested that IL-32 is downregulated in inflammatory diseases and exerts an anti-inflammatory effect. This review article focuses on recent findings regarding the detrimental and protective roles of IL-32 in development and management of inflammatory diseases. The exact role of IL-32 in AD, AA, and AR still remains to be elucidated. Future research should explore new avenues of IL-32 functionality in human inflammatory diseases.
Collapse
Affiliation(s)
- Rytis Boreika
- Department of Immunology and Allergology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Brigita Sitkauskiene
- Department of Immunology and Allergology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
15
|
Ostermann L, Maus R, Stolper J, Schütte L, Katsarou K, Tumpara S, Pich A, Mueller C, Janciauskiene S, Welte T, Maus UA. Alpha-1 antitrypsin deficiency impairs lung antibacterial immunity in mice. JCI Insight 2021; 6:140816. [PMID: 33554955 PMCID: PMC7934856 DOI: 10.1172/jci.insight.140816] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is a major inhibitor of serine proteases in mammals. Therefore, its deficiency leads to protease–antiprotease imbalance and a risk for developing lung emphysema. Although therapy with human plasma-purified AAT attenuates AAT deficiency–related emphysema, its impact on lung antibacterial immunity is poorly defined. Here, we examined the effect of AAT therapy on lung protective immunity in AAT-deficient (KO) mice challenged with Streptococcus pneumoniae. AAT-KO mice were highly susceptible to S. pneumoniae, as determined by severe lobar pneumonia and early mortality. Mechanistically, we found that neutrophil-derived elastase (NE) degraded the opsonophagocytically important collectins, surfactant protein A (SP-A) and D (SP-D), which was accompanied by significantly impaired lung bacterial clearance in S. pneumoniae–infected AAT-KO mice. Treatment of S. pneumoniae–infected AAT-KO mice with human AAT protected SP-A and SP-D from NE-mediated degradation and corrected the pulmonary pathology observed in these mice. Likewise, treatment with Sivelestat, a specific inhibitor of NE, also protected collectins from degradation and significantly decreased bacterial loads in S. pneumoniae–infected AAT-KO mice. Our findings show that NE is responsible for the degradation of lung SP-A and SP-D in AAT-KO mice affecting lung protective immunity in AAT deficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andreas Pich
- Institute of Toxicology and Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sabina Janciauskiene
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Tobias Welte
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Ulrich A Maus
- Division of Experimental Pneumology.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| |
Collapse
|
16
|
Menon M, Hussell T, Ali Shuwa H. Regulatory B cells in respiratory health and diseases. Immunol Rev 2021; 299:61-73. [PMID: 33410165 PMCID: PMC7986090 DOI: 10.1111/imr.12941] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
B cells are critical mediators of humoral immune responses in the airways through antibody production, antigen presentation, and cytokine secretion. In addition, a subset of B cells, known as regulatory B cells (Bregs), exhibit immunosuppressive functions via diverse regulatory mechanisms. Bregs modulate immune responses via the secretion of IL‐10, IL‐35, and tumor growth factor‐β (TGF‐β), and by direct cell contact. The balance between effector and regulatory B cell functions is critical in the maintenance of immune homeostasis. The importance of Bregs in airway immune responses is emphasized by the different respiratory disorders associated with abnormalities in Breg numbers and function. In this review, we summarize the role of immunosuppressive Bregs in airway inflammatory diseases and highlight the importance of this subset in the maintenance of respiratory health. We propose that improved understanding of signals in the lung microenvironment that drive Breg differentiation can provide novel therapeutic avenues for improved management of respiratory diseases.
Collapse
Affiliation(s)
- Madhvi Menon
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Tracy Hussell
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Halima Ali Shuwa
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
17
|
Tanabe N, Vasilescu DM, Hague CJ, Ikezoe K, Murphy DT, Kirby M, Stevenson CS, Verleden SE, Vanaudenaerde BM, Gayan-Ramirez G, Janssens W, Coxson HO, Paré PD, Hogg JC. Pathological Comparisons of Paraseptal and Centrilobular Emphysema in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2020; 202:803-811. [PMID: 32485111 DOI: 10.1164/rccm.201912-2327oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although centrilobular emphysema (CLE) and paraseptal emphysema (PSE) are commonly identified on multidetector computed tomography (MDCT), little is known about the pathology associated with PSE compared with that of CLE.Objectives: To assess the pathological differences between PSE and CLE in chronic obstructive pulmonary disease (COPD).Methods: Air-inflated frozen lung specimens (n = 6) obtained from patients with severe COPD treated by lung transplantation were scanned with MDCT. Frozen tissue cores were taken from central (n = 8) and peripheral (n = 8) regions of each lung, scanned with micro-computed tomography (microCT), and processed for histology. The core locations were registered to the MDCT, and a percentage of PSE or CLE was assigned by radiologists to each of the regions. MicroCT scans were used to measure number and structural change of terminal bronchioles. Furthermore, microCT-based volume fractions of CLE and PSE allowed classifying cores into mild emphysema, CLE-dominant, and PSE-dominant.Measurements and Main Results: The percentages of PSE measured on MDCT and microCT were positively associated (P = 0.015). The number of terminal bronchioles per milliliter of lung and cross-sectional lumen area were significantly lower and wall area percentage was significantly higher in CLE-dominant regions compared with mild emphysema and PSE-dominant regions (all P < 0.05), whereas no difference was found between PSE-dominant and mild emphysema samples (all P > 0.5). Immunohistochemistry showed significantly higher infiltration of neutrophils (P = 0.002), but not of macrophages, CD4, CD8, or B cells, in PSE compared with CLE regions.Conclusions: The terminal bronchioles are relatively preserved, whereas neutrophilic inflammation is increased in PSE-dominant regions compared with CLE-dominant regions in patients with COPD.
Collapse
Affiliation(s)
- Naoya Tanabe
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and.,Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Cameron J Hague
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kohei Ikezoe
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and
| | - Darra T Murphy
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Miranda Kirby
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and.,Department of Physics, Ryerson University, Toronto, Ontario, Canada
| | - Christopher S Stevenson
- Janssen Disease Interception Accelerator, Janssen Pharmaceutical Companies of Johnson and Johnson, Beerse, Belgium; and
| | - Stijn E Verleden
- Department of Chronic Disease, Metabolism and Aging, Laboratory of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Chronic Disease, Metabolism and Aging, Laboratory of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Department of Chronic Disease, Metabolism and Aging, Laboratory of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Wim Janssens
- Department of Chronic Disease, Metabolism and Aging, Laboratory of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Harvey O Coxson
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and
| | - Peter D Paré
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and
| | - James C Hogg
- Centre for Heart and Lung Innovation, St. Paul's Hospital, and
| |
Collapse
|
18
|
Williams M, Todd I, Fairclough LC. The role of CD8 + T lymphocytes in chronic obstructive pulmonary disease: a systematic review. Inflamm Res 2020; 70:11-18. [PMID: 33037881 PMCID: PMC7806561 DOI: 10.1007/s00011-020-01408-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/23/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE AND DESIGN This systematic review aims to establish the role of CD8 + T lymphocytes in COPD. METHODS Forty-eight papers published in the last 15 years were identified for inclusion. RESULTS CD8 + T-cells are increased in the lungs of patients with COPD (17 studies, 16 positive) whereas in the circulation, findings were inconclusive. Activation of CD8 + T-cells was enhanced in lungs (four studies, three positive) but cell phenotype was unclear. There was substantial evidence of a higher proportion of type 1 CD8 + (Tc1) cells in COPD (11 studies, 9 positive), though the population of type 2 (Tc2) cells was also increased (5 studies, 4 positive). CD8 + T-cells in COPD exhibited greater expression of cytotoxic proteins (five studies, five positive). Studies assessed a variety of questions so evidence was insufficient to draw firm conclusions. The role of CD8 + T-cells at acute exacerbation of COPD and also their contribution to alveolar destruction can only be hypothesised at this stage. CONCLUSIONS Not only is the number of CD8 + T-cells increased in COPD, these cells have increased capacity to exert effector functions and are likely to contribute to disease pathogenesis. Several mechanisms highlighted show promise for future investigation to consolidate current knowledge.
Collapse
Affiliation(s)
- Maya Williams
- School of Life Sciences, The University of Nottingham, Life Sciences Building, University Park, Nottingham, NG7 2RD, UK
| | - Ian Todd
- School of Life Sciences, The University of Nottingham, Life Sciences Building, University Park, Nottingham, NG7 2RD, UK
| | - Lucy C Fairclough
- School of Life Sciences, The University of Nottingham, Life Sciences Building, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
19
|
Khodayari N, Oshins R, Holliday LS, Clark V, Xiao Q, Marek G, Mehrad B, Brantly M. Alpha-1 antitrypsin deficient individuals have circulating extracellular vesicles with profibrogenic cargo. Cell Commun Signal 2020; 18:140. [PMID: 32887613 PMCID: PMC7487708 DOI: 10.1186/s12964-020-00648-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background Alpha-1 antitrypsin deficiency (AATD)-mediated liver disease is a toxic “gain-of-function” inflammation in the liver associated with intracellular retention of mutant alpha-1 antitrypsin. The clinical presentation of the disease includes fibrosis, cirrhosis and liver failure. However, the pathogenic mechanism of AATD-mediated liver disease is not well understood. Here, we investigated the role of plasma extracellular vesicles (EVs) in progression of AATD-mediated liver disease. Methods EVs were isolated from plasma of AATD individuals with liver disease and healthy controls. Their cytokines and miRNA content were examined by multiplex assay and small RNA sequencing. The bioactivity of EVs was assessed by qPCR, western blot analysis and immunofluorescent experiments using human hepatic stellate cells (HSCs) treated with EVs isolated from control or AATD plasma samples. Results We have found that AATD individuals have a distinct population of EVs with pathological cytokine and miRNA contents. When HSCs were cultured with AATD plasma derived-EVs, the expression of genes related to the development of fibrosis were significantly amplified compared to those treated with healthy control plasma EVs. Conclusion AATD individuals have a distinct population of EVs with abnormal cytokine and miRNA contents and the capacity to activate HSCs and mediate fibrosis. Better understanding of the components which cause liver inflammation and fibrogenesis, leading to further liver injury, has the potential to lead to the development of new treatments or preventive strategies to prevent AATD-mediated liver disease. Video abstract
Collapse
Affiliation(s)
- Nazli Khodayari
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, USA.
| | - Regina Oshins
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, USA
| | | | - Virginia Clark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, USA
| | | | - George Marek
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, USA
| | - Mark Brantly
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, USA.
| |
Collapse
|
20
|
Sapey E. Neutrophil Modulation in Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:247-259. [PMID: 32697897 DOI: 10.15326/jcopdf.7.3.2019.0164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Neutrophils have been implicated in the pathogenesis of alpha-1 antitrypsin deficiency (AATD) since the first descriptions of the disease. Neutrophil proteinases can cause all lung manifestations of AATD, from small airways destruction, to emphysema, to chronic bronchitis and airflow obstruction. Initially, it was proposed that neutrophil functions were normal in AATD, responding in an initially physiological manner to a high burden of pulmonary inflammation. More recent studies have shed new light on this, describing changes in neutrophil responses (a modulation of usual cellular functions) in the presence of inflammation or infection which might enhance tissue damage while impeding bacterial clearance, providing some evidence to support there being an AATD neutrophil phenotype. Many facets of neutrophil function in AATD can be explained by the loss of alpha-1 antitrypsin (AAT) in diverse biological processes. If this were the only reason for altered neutrophil functions, one would predict similar disease presentation across affected people. However, this is not the case. Despite similar (low) levels of AAT, lung disease is extremely variable in AATD, with some patients suffering a significant burden of lung disease and some much less, irrespective of smoking habits and, in some cases, despite augmentation therapy. This review will explore how complex neutrophil responses are and how they are altered with age, inflammation and AATD. Further, it will discuss the need to understand more completely which aspects of AATD-associated disease are driven by neutrophils and how patients more susceptible to neutrophil dysfunction could be identified to potentially stratify treatment approaches.
Collapse
Affiliation(s)
- Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
21
|
Alpha-1 Antitrypsin Deficiency and Pulmonary Morbidity in Patients with Primary Immunodeficiency Disease: A Single-Center Experience. Can Respir J 2020; 2020:4019608. [PMID: 32566054 PMCID: PMC7273390 DOI: 10.1155/2020/4019608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/22/2020] [Accepted: 05/06/2020] [Indexed: 01/20/2023] Open
Abstract
Background Alpha-1 antitrypsin deficiency (AATD) is of importance in the pathogenesis of pulmonary emphysema, chronic obstructive pulmonary diseases (COPD), and bronchiectasis. Various pulmonary disorders are a typical feature of primary immunodeficiency disease (PID). This includes recurrent pulmonary infections, immunodysregulation, and autoinflammatory diseases. As a result, incidence of acute and chronic pulmonary diseases is higher. Interestingly, pulmonary morbidity in PID and AATD share similar features. To study the coexistence of AATD in patients suffering from PID, we performed the underlying investigation. Methods We evaluated a study group of 149 patients (n = 149) with PID. In total, serum AAT concentrations were available for 110 patients (n = 110). For the identified patients, we analyzed both clinical associations and interactions. Results Among the investigated patients, reduced serum AAT levels were detected in 7 patients. With regard to the genotype, PI∗ZZ was found in 2 patients, whereas PI∗MZ was observed in 5 patients. Independent of the underlying phenotype, obstructive lung diseases were found in 2 patients with PI∗ZZ and 2 patients with PI∗MZ. Conclusions In Germany, the estimated percentage for PI∗ZZ and PI∗MZ is 0.01% and 1.9%, respectively. As demonstrated, the ratio in our study group was even higher. We identified seven patients with AATD. Since AATD contributes to pulmonary morbidity in PID patients, systematic underdiagnosis of the coexistence might yield a strong clinical impact. Hence, AAT analysis should be offered to all patients with confirmed PID diagnoses. To strengthen this finding, we suggest the investigation of larger databases.
Collapse
|
22
|
Tam A, Tanabe N, Churg A, Wright JL, Hogg JC, Sin DD. Sex differences in lymphoid follicles in COPD airways. Respir Res 2020; 21:46. [PMID: 32033623 PMCID: PMC7006095 DOI: 10.1186/s12931-020-1311-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/30/2020] [Indexed: 01/20/2023] Open
Abstract
Background Female smokers have increased risk for chronic obstructive pulmonary disease (COPD) compared with male smokers who have a similar history of cigarette smoke exposure. Tertiary lymphoid follicles are often found in the lungs of patients with severe COPD but sex-related differences have not been previously investigated. We determined the impact of female sex hormones on chronic cigarette smoke-induced expression of lymphoid aggregates in mice with COPD-like pathologies. Methods Lymphoid aggregate counts, total aggregate cross-sectional area and foamy macrophage counts were determined morphometrically in male, female, and ovariectomized mice exposed to air or cigarette smoke for 6 months. B-cell activating factor (BAFF) protein expression and markers of oxidative stress were evaluated in mouse lung tissues by immunofluorescence staining and gene expression analyses. Quantitative histology was performed on lung tissue sections of human COPD lungs to evaluate follicle formation. Results Lymphoid follicle and foamy macrophage counts as well as the total follicle cross-sectional area were differentially increased in lung tissues of female mice compared to male mice, and these differences were abolished by ovariectomy. These lymphoid aggregates were positive for CD45, CD20, CD21 and BAFF expression. Differential increases in Mmp12 and Cxcl2 gene expression correlated with an increase in foamy macrophages in parenchymal tissues of female but not male mice after smoke exposure. Parenchymal tissues from female mice failed to induce antioxidant-related genes in response to smoke exposure, and this effect was restored by ovariectomy. 3-nitrotyrosine, a stable marker of oxidative stress, positively correlated with Mmp12 and Cxcl2 gene expression. Hydrogen peroxide induced BAFF protein in mouse macrophage cell line. In human lung tissues, female smokers with severe COPD demonstrated increased numbers of lymphoid follicles compared with males. Conclusions Chronic smoke exposure increases the risk of lymphoid aggregate formation in female mice compared with male mice, which is mediated female sex hormones and BAFF expression in an oxidative environment.
Collapse
Affiliation(s)
- Anthony Tam
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada
| | - Naoya Tanabe
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada.,Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Andrew Churg
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne L Wright
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - James C Hogg
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada.
| |
Collapse
|
23
|
Abstract
It is well known that particulate matter suspended in the earth's atmosphere generated by tobacco smoke, automobile exhaust, industrial processes, and forest fires has been identified as a major risk factor for chronic lung disease. Particulate matter can be divided into large, intermediate, and fine particulates. When inhaled, large particulates develop sufficient momentum to leave the flowing stream of inhaled air and deposit by impaction in the nose, mouth, nasopharynx, larynx, trachea, and central bronchi. Intermediate-sized particulates that develop less momentum deposit in the smaller bronchi and larger bronchioles, and the finest particulates that develop the least momentum make it to the distal gas-exchanging tissue, where gas moves solely by diffusion. On the basis of Einstein's classic work on Brownian motion that showed particles suspended in a gas diffuse much more slowly than the gas in which they are suspended, we postulate that the small airways that accommodate the shift from bulk airflow to diffusion become the major site for deposition of fine particles, resulting in a host immune response. Much remains to be learned about the interaction between the deposition of fine particulates and the host immune and tissue responses; the purpose of this review is to examine the hypothesis that the smallest conducting airways and proximal gas-exchanging tissue are the primary sites for the deposition of the finest particulates inhaled into the lungs.
Collapse
|
24
|
Exome Sequencing Reveals Immune Genes as Susceptibility Modifiers in Individuals with α 1-Antitrypsin Deficiency. Sci Rep 2019; 9:13088. [PMID: 31511551 PMCID: PMC6739380 DOI: 10.1038/s41598-019-49409-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a genetic disorder associated to early onset emphysema, mainly imputable to Pi*ZZ genotype. In spite of the serious potential effects, many AATD individuals do not develop emphysema. To identify genes/variants potentially involved in emphysema development we studied 4 AATD families. Each family had at least one affected sibling with emphysema and one non-affected. Whole Exome Sequencing (WES) was performed on genomic DNA isolated from 9 individuals with AATD (4 affected/5 non-affected). Genetic variants confirmed at least in three families were prioritized using QueryOR and network analysis was used to verify enriched pathways. In affected subjects: 14 genes (57% immune-related) segregated in a recessive model and 21 (29% immune-related) in a dominant model. In non-affected subjects: 21 genes (43% immune-related) segregated in a recessive model and 50 (24% immune-related) in a dominant model. In affected siblings immune genes had an activating function, while where immune-suppressing in non-affected siblings involving antigen processing, MHC-I presentation, TCR and PD-1 signalling. This study describes possible genetic susceptibility factors for emphysema development in AATD, and suggests that gene variants involved in regulation of immune homeostasis and maintenance of self-tolerance contribute to the development or suppression of the disease.
Collapse
|
25
|
Alpha-1-Antitrypsin Ameliorates Pristane Induced Diffuse Alveolar Hemorrhage in Mice. J Clin Med 2019; 8:jcm8091341. [PMID: 31470606 PMCID: PMC6780888 DOI: 10.3390/jcm8091341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/21/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
Diffuse alveolar hemorrhage (DAH) is a fatal complication in patients with lupus. DAH can be induced in B6 mice by an intraperitoneal injection of pristane. Since human alpha-1-antitrypsin (hAAT) is an anti-inflammatory and immuno-regulatory protein, we investigated the protective effect of hAAT against pristane-induced DAH in B6 mice and hAAT transgenic (hAAT-Tg) mice. We first showed that hAAT Tg expression lowers TNF-α production in B cells, as well as CD4+ T cells in untreated mice. Conversely, the frequency of regulatory CD4+CD25+ and CD4+CD25-IL-10+ cells was significantly higher in hAAT-Tg than in B6 mice. This confirmed the anti-inflammatory effect of hAAT that was observed even at steady state. One week after a pristane injection, the frequency of peritoneal Ly6Chi inflammatory monocytes and neutrophils in hAAT-Tg mice was significantly lower than that in B6 mice. Importantly, pristane-induced DAH was completely prevented in hAAT-Tg mice and this was associated with a modulation of anti- to pro-inflammatory myeloid cell ratio/balance. We also showed that treatment with hAAT decreased the severity of DAH in B6 mice. These results showed for the first time that hAAT has a therapeutic potential for the treatment of DAH.
Collapse
|
26
|
Ladjemi MZ, Burgel PR, Pilette C. Reply to Polverino: Deconvoluting Chronic Obstructive Pulmonary Disease: Are B Cells the Frontrunners? Am J Respir Crit Care Med 2019; 199:1171-1172. [PMID: 30633554 PMCID: PMC6515872 DOI: 10.1164/rccm.201812-2249le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Maha Zohra Ladjemi
- 1 Université Catholique de Louvain Brussels, Belgium.,2 Walloon Excellence in Life Sciences and Biotechnology Brussels, Belgium
| | - Pierre Régis Burgel
- 3 Université Paris Descartes Paris, France.,4 Hôpital Cochin, AP-HP Paris, France and
| | - Charles Pilette
- 1 Université Catholique de Louvain Brussels, Belgium.,2 Walloon Excellence in Life Sciences and Biotechnology Brussels, Belgium.,5 Cliniques Universitaires Saint-Luc Brussels, Belgium
| |
Collapse
|
27
|
Polverino F. Deconvoluting Chronic Obstructive Pulmonary Disease: Are B Cells the Frontrunners? Am J Respir Crit Care Med 2019; 199:1170-1171. [PMID: 30633537 PMCID: PMC6515871 DOI: 10.1164/rccm.201811-2093le] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Francesca Polverino
- Asthma and Airway Disease Research CenterUniversity of ArizonaTucson, Arizona
| |
Collapse
|
28
|
Advances in chronic obstructive pulmonary disease genetics: building the picture one piece at a time. THE LANCET RESPIRATORY MEDICINE 2019; 7:371-372. [PMID: 31000474 DOI: 10.1016/s2213-2600(19)30082-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/04/2019] [Indexed: 01/08/2023]
|
29
|
Elshikha AS, Yuan Y, Lu Y, Chen MJ, Abboud G, Akbar MA, Plate H, Wolney H, Hoffmann T, Tagari E, Zeumer L, Morel L, Song S. Alpha 1 Antitrypsin Gene Therapy Extends the Lifespan of Lupus-Prone Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:131-142. [PMID: 30547047 PMCID: PMC6258868 DOI: 10.1016/j.omtm.2018.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/13/2018] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by high levels of pathogenic autoantibodies and tissue damage. Multiple studies showed that dendritic cell (DC) activation plays a critical role in SLE pathogenesis. Human alpha 1 antitrypsin (hAAT) is a serine proteinase inhibitor with potent anti-inflammatory and cytoprotective properties. In this study, we first examined the effects of hAAT on the functions of DCs from lupus-prone mice, and we showed that hAAT treatment efficiently inhibited CpG- (TLR9 agonist) induced activation of bone marrow-derived conventional and plasmacytoid DCs as well as the production of pro-inflammatory cytokines. The hAAT treatment also attenuated DC help for B cell proliferation and immunoglobulin M (IgM) production. We next tested the protective effect of hAAT protein and gene therapy using recombinant adeno-associated virus 8 (rAAV8-CB-hAAT) in a spontaneous lupus mouse model, and we showed that both treatments decreased autoantibody levels. Importantly, rAAV8-CB-hAAT did not induce an immune response to its transgene product (hAAT), but it showed more pronounced therapeutic effects in reducing urine protein levels and extending the lifespan of these mice. These results indicate that AAT has therapeutic potential in the treatment of SLE in humans.
Collapse
Affiliation(s)
- Ahmed Samir Elshikha
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Sharkia, Egypt
| | - Ye Yuan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Yuanqing Lu
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Mong-Jen Chen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mohammad Ahsanul Akbar
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Henrike Plate
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Hedwig Wolney
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Tanja Hoffmann
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Eleni Tagari
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Leilani Zeumer
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
30
|
Song S. Alpha-1 Antitrypsin Therapy for Autoimmune Disorders. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:289-301. [PMID: 30723786 DOI: 10.15326/jcopdf.5.4.2018.0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Autoimmune diseases are conditions caused by an over reactive immune system that attacks self-tissues and organs. Although the pathogenesis of autoimmune disease is complex and multi-factorial, inflammation is commonly involved. Therefore, anti-inflammatory therapies hold potential for the treatment of autoimmune diseases. However, long-term control of inflammation is challenging and most of the currently used drugs have side effects. Alpha-1 antitrypsin (AAT) is an anti-inflammatory protein with a well-known safety profile. The therapeutic potential of AAT has been tested in several autoimmune disease models. The first study using a recombinant adeno-associated viral (rAAV) vector showed that AAT gene transfer prevented the development of type 1 diabetes (T1D) in the non-obese diabetic (NOD) mouse model. Subsequent studies showed that treatment with AAT protein prevented and reversed type 1 diabetes. The beneficial effects of AAT treatment have also been observed in other autoimmune disease models such as rheumatoid arthritis and systemic lupus erythematosus. This paper reviews the therapeutic application of AAT and discusses possible mechanisms of action in various autoimmune diseases.
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville
| |
Collapse
|
31
|
α1-Antitrypsin Polymerizes in Alveolar Macrophages of Smokers With and Without α1-Antitrypsin Deficiency. Chest 2018; 154:607-616. [DOI: 10.1016/j.chest.2018.04.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/05/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022] Open
|
32
|
Abstract
α1-Antitrypsin deficiency (AATD) is an inherited metabolic disorder in which mutations in the coding sequence of the SERPINA1 gene prevent secretion of α1-antitrypsin (α1-AT) and cause predisposition to pulmonary and liver diseases. The heterogeneity of clinical manifestations in AATD is related to the complexity of biological function of α1-AT. The role of smoking is crucial in the natural history of lung damage progression in severe AATD individuals, even if it also partly explains the heterogeneity in lung disease. Lung damage progression in AATD can also be related to body mass index, exacerbation rate, sex, environmental exposure and specific mutations of SERPINA1. Recent randomised controlled trials, together with previous observational work, have provided compelling evidence for the importance of early detection and intervention in order to enable patients to receive appropriate treatment and preserve functional lung tissue. Early detection and intervention in cases of α1-antitrypsin deficiency are essential to enable appropriate treatment and preserve functional lung tissuehttp://ow.ly/Mr3P30jUEyn
Collapse
Affiliation(s)
- Ilaria Ferrarotti
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| | - Stefania Ottaviani
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| | | | - Angelo G Corsico
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| |
Collapse
|
33
|
Kawamoto T, Kanazawa H, Tochino Y, Kawaguchi T. Evaluation of the severity of small airways obstruction and alveolar destruction in chronic obstructive pulmonary disease. Respir Med 2018; 141:159-164. [PMID: 30053962 DOI: 10.1016/j.rmed.2018.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Airflow limitation in COPD is caused by a mixture of small airways obstruction and alveolar destruction. OBJECTIVE To evaluate the contributions of these factors to airflow limitation through measurement of two biomarkers, pentosidine and vascular endothelial growth factor (VEGF), which reflect pathology or function of the lower respiratory tract of COPD. METHODS We measured pentosidine and VEGF levels in induced sputum from 23 non-smokers, 26 smokers without COPD, and 43 smokers with COPD. We evaluated the correlations of two biomarkers levels with the grade of low attenuation area (LAA) in high-resolution computed tomographic scans and the Δ N2 from the nitrogen washout curve. RESULTS Pentosidine levels were significantly higher in smokers with COPD than in non-smokers and smokers without COPD. In contrast, VEGF levels were significantly lower in smokers without COPD than in non-smokers, and further decreased in smokers with COPD. In the four-stage classification of LAA grading, pentosidine levels steeply increased from grade I to Ⅳ, while VEGF levels decreased with increasing severity of LAA grade. Pentosidine levels were positively correlated with Δ N2 in COPD patients with mild emphysema. In contrast, VEGF levels were inversely correlated with Δ N2 in COPD patients with severe emphysema. CONCLUSION Pentosidine level is responsible for the severity of small airways obstruction, while VEGF level reflects the magnitude of alveolar destruction. Thus, simultaneous measurement of pentosidine and VEGF levels may be a promising approach to discriminate the severity of small airways obstruction and alveolar destruction in the lower respiratory tract of COPD.
Collapse
Affiliation(s)
- Tamaki Kawamoto
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiroshi Kanazawa
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| | - Yoshihiro Tochino
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
34
|
Abstract
This review seeks to give an overview of alpha-1 antitrypsin deficiency, including the different disease phenotypes that it encompasses. We then describe the different therapeutic endeavors that have been undertaken to address these different phenotypes. Lastly we discuss future potential therapeutics, such as genome editing, and how they may play a role in treating alpha-1 antitrypsin deficiency.
Collapse
|
35
|
Tanabe N, Vasilescu DM, Kirby M, Coxson HO, Verleden SE, Vanaudenaerde BM, Kinose D, Nakano Y, Paré PD, Hogg JC. Analysis of airway pathology in COPD using a combination of computed tomography, micro-computed tomography and histology. Eur Respir J 2018; 51:51/2/1701245. [PMID: 29444912 DOI: 10.1183/13993003.01245-2017] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/30/2017] [Indexed: 01/01/2023]
Abstract
The small conducting airways are the major site of obstruction in chronic obstructive pulmonary disease (COPD). This study examined small airway pathology using a novel combination of multidetector row computed tomography (MDCT), micro-computed tomography (microCT) and histology.Airway branches visible on specimen MDCT were counted and the dimensions of the third- to fifth-generation airways were computed, while the terminal bronchioles (designated TB), preterminal bronchioles (TB-1) and pre-preterminal bronchioles (TB-2) were examined with microCT and histology in eight explanted lungs with end-stage COPD and seven unused donor lungs that served as controls.On MDCT, COPD lungs showed a decrease in the number of 2-2.5 mm diameter airways and the lumen area of fifth-generation airways, while on microCT there was a reduction in the number of terminal bronchioles as well as a decrease in the luminal areas, wall volumes and alveolar attachments to the walls of TB, TB-1 and TB-2 bronchioles. The combination of microCT and histology showed increased B-cell infiltration into the walls of TB-1 and TB-2 bronchioles, and this change was correlated with a reduced number of alveolar attachments in COPD.Small airways disease extends from 2 mm diameter airways to the terminal bronchioles in COPD. Destruction of alveolar attachments may be driven by a B-cell-mediated immune response in the preterminal bronchioles.
Collapse
Affiliation(s)
- Naoya Tanabe
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada .,Dept of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Dragoş M Vasilescu
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Miranda Kirby
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Harvey O Coxson
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Stijn E Verleden
- KU Leuven - University of Leuven, Dept of Clinical and Experimental Medicine, Division of Respiratory diseases, Leuven, Belgium
| | - Bart M Vanaudenaerde
- KU Leuven - University of Leuven, Dept of Clinical and Experimental Medicine, Division of Respiratory diseases, Leuven, Belgium
| | - Daisuke Kinose
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.,Division of Respiratory Medicine, Dept of Internal Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Yasutaka Nakano
- Division of Respiratory Medicine, Dept of Internal Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Peter D Paré
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - James C Hogg
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Suzuki M, Sze MA, Campbell JD, Brothers JF, Lenburg ME, McDonough JE, Elliott WM, Cooper JD, Spira A, Hogg JC. The cellular and molecular determinants of emphysematous destruction in COPD. Sci Rep 2017; 7:9562. [PMID: 28842670 PMCID: PMC5573394 DOI: 10.1038/s41598-017-10126-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
The introduction of microCT has made it possible to show that the terminal bronchioles are narrowed and destroyed before the onset of emphysematous destruction in COPD. This report extends those observations to the cellular and molecular level in the centrilobular phenotype of emphysematous destruction in lungs donated by persons with very severe COPD (n = 4) treated by lung transplantation with unused donor lungs (n = 4) serving as controls. These lung specimens provided companion samples to those previously examined by microCT (n = 61) that we examined using quantitative histology (n = 61) and gene expression profiling (n = 48). The histological analysis showed that remodeling and destruction of the bronchiolar and alveolar tissue is associated with macrophage, CD4, CD8, and B cell infiltration with increased formation of tertiary lymphoid organs. Moreover, gene set enrichment analysis showed that genes known to be expressed by natural killer (NK), lymphoid tissue inducer (LTi), and innate lymphoid cell 1 (ILC1) cells, but not ILC2 or ILC3 cells, were enriched in the expression profiles associated with CD4, CD8, and B cell infiltration. Based on these findings, we postulate that the centrilobular phenotype of emphysematous destruction COPD is driven by a Th1 response activated by infiltrating ILC1, NK, and LTi cells.
Collapse
Affiliation(s)
- Masaru Suzuki
- Centre for Heart Lung Innovation, St. Paul's Hospital, Departments of Medicine, and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Marc A Sze
- Centre for Heart Lung Innovation, St. Paul's Hospital, Departments of Medicine, and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joshua D Campbell
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John F Brothers
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Marc E Lenburg
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John E McDonough
- Centre for Heart Lung Innovation, St. Paul's Hospital, Departments of Medicine, and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - W Mark Elliott
- Centre for Heart Lung Innovation, St. Paul's Hospital, Departments of Medicine, and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joel D Cooper
- Division of Thoracic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Avrum Spira
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - James C Hogg
- Centre for Heart Lung Innovation, St. Paul's Hospital, Departments of Medicine, and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Kwon JW, Chang HS, Heo JS, Bae DJ, Lee JU, Jung CA, Son JH, Park JS, Kim SH, Min KU, Park CS. Characteristics of asthmatics with detectable IL-32γ in induced sputum. Respir Med 2017; 129:85-90. [DOI: 10.1016/j.rmed.2017.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/22/2017] [Accepted: 06/05/2017] [Indexed: 12/30/2022]
|
38
|
Tanabe N, Vasilescu DM, McDonough JE, Kinose D, Suzuki M, Cooper JD, Paré PD, Hogg JC. Micro-Computed Tomography Comparison of Preterminal Bronchioles in Centrilobular and Panlobular Emphysema. Am J Respir Crit Care Med 2017; 195:630-638. [PMID: 27611890 DOI: 10.1164/rccm.201602-0278oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
RATIONALE Very little is known about airways that are too small to be visible on thoracic multidetector computed tomography but larger than the terminal bronchioles. OBJECTIVES To examine the structure of preterminal bronchioles located one generation proximal to terminal bronchioles in centrilobular and panlobular emphysema. METHODS Preterminal bronchioles were identified by backtracking from the terminal bronchioles, and their centerlines were established along the entire length of their lumens. Multiple cross-sectional images perpendicular to the centerline were reconstructed to evaluate the bronchiolar wall and lumen, and the alveolar attachments to the outer airway walls in relation to emphysematous destruction in 28 lung samples from six patients with centrilobular emphysema, 20 lung samples from seven patients with panlobular emphysema associated with alpha-1 antitrypsin deficiency, and 47 samples from seven control (donor) lungs. MEASUREMENTS AND MAIN RESULTS The preterminal bronchiolar length, wall volume, total volume (wall + lumen), lumen circularity, and number of alveolar attachments were reduced in both centrilobular and panlobular emphysema compared with control lungs. In contrast, thickening of the wall and narrowing of the lumen were more severe and heterogeneous in centrilobular than in panlobular emphysema. The bronchiolar lumen was narrower in the middle than at both ends, and the decreased number of alveolar attachments was associated with increased wall thickness in centrilobular emphysema. CONCLUSIONS These results provide new information about small airways pathology in centrilobular and panlobular emphysema and show that these changes affect airways that are not visible with thoracic multidetector computed tomography scans but located proximal to the terminal bronchioles in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Naoya Tanabe
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dragoş M Vasilescu
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - John E McDonough
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,2 Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, KU Leuven-University of Leuven, Leuven, Belgium
| | - Daisuke Kinose
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masaru Suzuki
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,3 First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan; and
| | - Joel D Cooper
- 4 Division of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter D Paré
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - James C Hogg
- 1 Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
39
|
Differential regulation of muscle protein turnover in response to emphysema and acute pulmonary inflammation. Respir Res 2017; 18:75. [PMID: 28464882 PMCID: PMC5414227 DOI: 10.1186/s12931-017-0531-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/03/2017] [Indexed: 01/22/2023] Open
Abstract
Background Exacerbations in COPD are often accompanied by pulmonary and systemic inflammation, and associated with increased susceptibility to and prevalence of weight loss and muscle wasting. Muscle mass loss during disease exacerbations may contribute to emphysema-associated muscle atrophy. However, whether pulmonary inflammation in presence of emphysema differentially affects skeletal muscle, including protein synthesis and degradation signaling pathways has not previously been addressed. The aims of this study were to 1) develop a mouse model of disease exacerbation-associated muscle wasting, 2) evaluate whether emphysema and muscle wasting can be monitored non-invasively and 3) assess alterations in muscle protein turnover regulation. Methods Emphysema was induced by three, weekly intra-tracheal (IT) elastase (E) or vehicle control (vc) instillations, followed by one single IT-LPS bolus (L) or vc instillation to mimic pulmonary inflammation-driven disease exacerbation. Consequently, four experimental groups were defined: vc/vc (‘C’), E/vc (‘E’), vc/LPS (‘L’), E/LPS (‘E + L’). Using micro cone-beam CT-scans, emphysema development and muscle mass changes were monitored, and correlated to muscle weight 48 h after LPS instillation. Protein turnover signaling was assessed in muscle tissue collected 24 h post LPS instillation. Results Micro-CT imaging correlated strongly with established invasive measurements of emphysema and muscle atrophy. Pulmonary inflammation following LPS instillation developed irrespective of emphysema and body and muscle weight were similarly reduced in the ‘L’ and ‘E + L’ groups. Accordingly, mRNA and protein expression levels of genes of the ubiquitin-proteasome pathway (UPS) and the autophagy-lysosomal pathway (ALP) were upregulated in skeletal muscle following IT-LPS (‘L’ and ‘E + L’). In contrast, mTOR signaling, which controls ALP and protein synthesis, was reduced by pulmonary inflammation (‘L’ and ‘E + L’) as well as emphysema as a single insult (‘E’) compared to control. Conclusion Changes in lung tissue density and muscle mass can be monitored non-invasively to evaluate emphysema and muscle atrophy longitudinally. Acute loss of muscle mass evoked by pulmonary inflammation is similar in control and emphysematous mice. Although muscle atrophy cues in response to pulmonary inflammation are not altered by emphysema, emphysema itself affects protein synthesis and ALP signaling, which may interfere with muscle mass recovery and impair maintenance of muscle mass in emphysema. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0531-z) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Hogg JC, Paré PD, Hackett TL. The Contribution of Small Airway Obstruction to the Pathogenesis of Chronic Obstructive Pulmonary Disease. Physiol Rev 2017; 97:529-552. [PMID: 28151425 PMCID: PMC6151481 DOI: 10.1152/physrev.00025.2015] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hypothesis that the small conducting airways were the major site of obstruction to airflow in normal lungs was introduced by Rohrer in 1915 and prevailed until Weibel introduced a quantitative method of studying lung anatomy in 1963. Green repeated Rohrer's calculations using Weibels new data in 1965 and found that the smaller conducting airways offered very little resistance to airflow. This conflict was resolved by seminal experiments conducted by Macklem and Mead in 1967, which confirmed that a small proportion of the total lower airways resistance is attributable to small airways <2 mm in diameter. Shortly thereafter, Hogg, Macklem, and Thurlbeck used this technique to show that small airways become the major site of obstruction in lungs affected by emphysema. These and other observations led Mead to write a seminal editorial in 1970 that postulated the small airways are a silent zone within normal lungs where disease can accumulate over many years without being noticed. This review provides a progress report since the 1970s on methods for detecting chronic obstructive pulmonary disease, the structural nature of small airways' disease, and the cellular and molecular mechanisms that are thought to underlie its pathogenesis.
Collapse
Affiliation(s)
- James C Hogg
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| | - Peter D Paré
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| |
Collapse
|
41
|
Integrating Transcriptomics, Proteomics, and Metabolomics Profiling with System Pharmacology for the Delineation of Long-Term Therapeutic Mechanisms of Bufei Jianpi Formula in Treating COPD. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7091087. [PMID: 28424787 PMCID: PMC5382313 DOI: 10.1155/2017/7091087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/22/2017] [Indexed: 12/19/2022]
Abstract
In previous work, we identified 145 active compounds from Bufei Jianpi formula (BJF) by system pharmacology and found that BJF showed short-term effect on chronic obstructive pulmonary disease (COPD) rats. Here, we applied the transcriptomic, proteomic, and metabolomics approaches to illustrate the long-term anti-COPD action and its system mechanism of BJF. BJF has obvious anti-COPD effect through decreasing inflammatory cytokines level, preventing protease-antiprotease imbalance and collagen deposition on week 32 by continuous oral administration to rats from weeks 9 to 20. Subsequently, applying the transcriptomic, proteomic, and metabolomics techniques, we detected a number of regulated genes, proteins, and metabolites, mainly related to antioxidant activity, focal adhesion, or lipid metabolism, in lung tissues of COPD and BJF-treated rats. Afterwards, we integrated system pharmacology target, transcript, protein, and metabolite data sets and found that many genes, proteins, and metabolites in rats BJF-treated group and the target proteins of BJF were mainly attributed to lipid metabolism, inflammatory response, oxidative stress, and focal adhesion. Taken together, BJF displays long-term anti-COPD effect probably by system regulation of the lipid metabolism, inflammatory response pathways oxidative stress, and focal adhesion.
Collapse
|
42
|
Morrow JD, Zhou X, Lao T, Jiang Z, DeMeo DL, Cho MH, Qiu W, Cloonan S, Pinto-Plata V, Celli B, Marchetti N, Criner GJ, Bueno R, Washko GR, Glass K, Quackenbush J, Choi AMK, Silverman EK, Hersh CP. Functional interactors of three genome-wide association study genes are differentially expressed in severe chronic obstructive pulmonary disease lung tissue. Sci Rep 2017; 7:44232. [PMID: 28287180 PMCID: PMC5347019 DOI: 10.1038/srep44232] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 02/06/2017] [Indexed: 12/20/2022] Open
Abstract
In comparison to genome-wide association studies (GWAS), there has been poor replication of gene expression studies in chronic obstructive pulmonary disease (COPD). We performed microarray gene expression profiling on a large sample of resected lung tissues from subjects with severe COPD. Comparing 111 COPD cases and 40 control smokers, 204 genes were differentially expressed; none were at significant GWAS loci. The top differentially expressed gene was HMGB1, which interacts with AGER, a known COPD GWAS gene. Differentially expressed genes showed enrichment for putative interactors of the first three identified COPD GWAS genes IREB2, HHIP, and FAM13A, based on gene sets derived from protein and RNA binding studies, RNA-interference, a murine smoking model, and expression quantitative trait locus analyses. The gene module most highly associated for COPD in Weighted Gene Co-Expression Network Analysis (WGCNA) was enriched for B cell pathways, and shared seventeen genes with a mouse smoking model and twenty genes with previous emphysema studies. As in other common diseases, genes at COPD GWAS loci were not differentially expressed; however, using a combination of network methods, experimental studies and careful phenotype definition, we found differential expression of putative interactors of these genes, and we replicated previous human and mouse microarray results.
Collapse
Affiliation(s)
- Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Taotao Lao
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhiqiang Jiang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Suzanne Cloonan
- Department of Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | - Victor Pinto-Plata
- Department of Critical Care Medicine and Pulmonary Disease, Baystate Medical Center, Springfield, MA, USA
| | - Bartholome Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Nathaniel Marchetti
- Division of Pulmonary and Critical Care Medicine, Temple University, Philadelphia, PA, USA
| | - Gerard J Criner
- Division of Pulmonary and Critical Care Medicine, Temple University, Philadelphia, PA, USA
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John Quackenbush
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Augustine M K Choi
- Department of Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
43
|
Polverino F, Seys LJM, Bracke KR, Owen CA. B cells in chronic obstructive pulmonary disease: moving to center stage. Am J Physiol Lung Cell Mol Physiol 2016; 311:L687-L695. [PMID: 27542809 DOI: 10.1152/ajplung.00304.2016] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammatory responses in the lungs contribute to the development and progression of chronic obstructive pulmonary disease (COPD). Although research studies focused initially on the contributions of the innate immune system to the pathogenesis of COPD, more recent studies have implicated adaptive immune responses in COPD. In particular, studies have demonstrated increases in B cell counts and increases in the number and size of B cell-rich lymphoid follicles in COPD lungs that correlate directly with COPD severity. There are also increases in lung levels of mediators that promote B cell maturation, activation, and survival in COPD patients. B cell products such as autoantibodies directed against lung cells, components of cells, and extracellular matrix proteins are also present in COPD lungs. These autoantibodies may contribute to lung inflammation and injury in COPD patients, in part, by forming immune complexes that activate complement components. Studies of B cell-deficient mice and human COPD patients have linked B cells most strongly to the emphysema phenotype. However, B cells have protective activities during acute exacerbations of COPD by promoting adaptive immune responses that contribute to host defense against pathogens. This review outlines the evidence that links B cells and B cell-rich lymphoid follicles to the pathogenesis of COPD and the mechanisms involved. It also reviews the potential and limitations of B cells as therapeutic targets to slow the progression of human COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; University of Parma, Parma, Italy; and
| | - Leen J M Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico;
| |
Collapse
|
44
|
Alpha-1 Antitrypsin Deficiency: Beyond the Protease/Antiprotease Paradigm. Ann Am Thorac Soc 2016; 13 Suppl 4:S305-10. [DOI: 10.1513/annalsats.201510-671kv] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
45
|
Rationale and Design of the Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis Study. Alpha-1 Protocol. Ann Am Thorac Soc 2016; 12:1551-60. [PMID: 26153726 DOI: 10.1513/annalsats.201503-143oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Severe deficiency of alpha-1 antitrypsin has a highly variable clinical presentation. The Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis α1 Study is a prospective, multicenter, cross-sectional study of adults older than age 35 years with PiZZ or PiMZ alpha-1 antitrypsin genotypes. It is designed to better understand if microbial factors influence this heterogeneity. Clinical symptoms, pulmonary function testing, computed chest tomography, exercise capacity, and bronchoalveolar lavage (BAL) will be used to define chronic obstructive pulmonary disease (COPD) phenotypes that can be studied with an integrated systems biology approach that includes plasma proteomics; mouth, BAL, and stool microbiome and virome analysis; and blood microRNA and blood mononuclear cell RNA and DNA profiling. We will rely on global genome, transcriptome, proteome, and metabolome datasets. Matched cohorts of PiZZ participants on or off alpha-1 antitrypsin augmentation therapy, PiMZ participants not on augmentation therapy, and control participants from the Subpopulations and Intermediate Outcome Measures in COPD Study who match on FEV1 and age will be compared. In the primary analysis, we will determine if the PiZZ individuals on augmentation therapy have a difference in lower respiratory tract microbes identified compared with matched PiZZ individuals who are not on augmentation therapy. By characterizing the microbiome in alpha-1 antitrypsin deficiency (AATD), we hope to define new phenotypes of COPD that explain some of the diversity of clinical presentations. As a unique genetic cause of COPD, AATD may inform typical COPD pathogenesis, and better understanding of it may illuminate the complex interplay between environment and genetics. Although the biologic approaches are hypothesis generating, the results may lead to development of novel biomarkers, better understanding of COPD phenotypes, and development of novel diagnostic and therapeutic trials in AATD and COPD. Clinical trial registered with www.clinicaltrials.gov (NCT01832220).
Collapse
|
46
|
Marchetti N, Criner GJ. Update in Chronic Obstructive Pulmonary Disease 2015. Am J Respir Crit Care Med 2016; 193:1092-100. [DOI: 10.1164/rccm.201602-0213up] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
47
|
Baraldo S, Balestro E, Bazzan E, Tiné ME, Biondini D, Turato G, Cosio MG, Saetta M. Alpha-1 Antitrypsin Deficiency Today: New Insights in the Immunological Pathways. Respiration 2016; 91:380-5. [PMID: 27164860 DOI: 10.1159/000445692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/17/2016] [Indexed: 11/19/2022] Open
Abstract
More than 50 years ago, the observation that absence of the α1 band from protein electrophoresis is associated with severe emphysema established the link between α1-antitrypsin deficiency (AATD) and lung damage. From this discovery, the classic paradigm of protease/antiprotease imbalance was derived, linking lung destruction in patients with AATD to the unopposed effect of proteases. By extension, this paradigm was also applied to patients with 'common' chronic obstructive pulmonary disease, in whom large increases in smoke-induced proteases could overwhelm the antiprotease capability of AAT. However, it has become increasingly evident that AAT has important anti-inflammatory and immunoregulatory activities which, beside its antiprotease function, may be critically involved in lung destruction. From this perspective, we will consider recent evidence, based on epidemiological, clinical and immunopathological studies, suggesting that it is time to move on from the original protease/antiprotease paradigm toward a more complex view of the condition, which embraces its immunomodulating functions. Of importance, the potent immunoregulatory, tolerogenic role of AAT may support its therapeutic use in a number of diseases other than AATD, particularly in immune-related disorders.
Collapse
Affiliation(s)
- Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova and Padova City Hospital, Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Balestro E, Calabrese F, Turato G, Lunardi F, Bazzan E, Marulli G, Biondini D, Rossi E, Sanduzzi A, Rea F, Rigobello C, Gregori D, Baraldo S, Spagnolo P, Cosio MG, Saetta M. Immune Inflammation and Disease Progression in Idiopathic Pulmonary Fibrosis. PLoS One 2016; 11:e0154516. [PMID: 27159038 PMCID: PMC4861274 DOI: 10.1371/journal.pone.0154516] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/14/2016] [Indexed: 11/18/2022] Open
Abstract
The clinical course in idiopathic pulmonary fibrosis (IPF) is highly heterogeneous, with some patients having a slow progression and others an accelerated clinical and functional decline. This study aims to clinically characterize the type of progression in IPF and to investigate the pathological basis that might account for the observed differences in disease behavior. Clinical and functional data were analyzed in 73 IPF patients, followed long-time as candidates for lung transplantation. The forced vital capacity (FVC) change/year (< or ≥10% predicted) was used to define "slow" or "rapid" disease progression. Pathological abnormalities were quantified in the explanted lung of 41 out of 73 patients undergoing lung transplantation. At diagnosis, slow progressors (n = 48) showed longer duration of symptoms and lower FVC than rapid progressors (n = 25). Eleven slow and 3 rapid progressors developed an acute exacerbation (AE) during follow-up. Quantitative lung pathology showed a severe innate and adaptive inflammatory infiltrate in rapid progressors, markedly increased compared to slow progressors and similar to that observed in patients experiencing AE. The extent of inflammation was correlated with the yearly FVC decline (r = 0.52, p = 0.005). In conclusion an innate and adaptive inflammation appears to be a prominent feature in the lung of patients with IPF and could contribute to determining of the rate of disease progression.
Collapse
Affiliation(s)
- Elisabetta Balestro
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Erica Bazzan
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Giuseppe Marulli
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Davide Biondini
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Emanuela Rossi
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Alessandro Sanduzzi
- Department of Clinical Medicine and Surgery, Federico II University, Napoli, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Chiara Rigobello
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
- Respiratory Division Meakins-Christie Laboratories, McGill University, Montreal, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| |
Collapse
|
49
|
Solleti SK, Srisuma S, Bhattacharya S, Rangel-Moreno J, Bijli KM, Randall TD, Rahman A, Mariani TJ. Serpine2 deficiency results in lung lymphocyte accumulation and bronchus-associated lymphoid tissue formation. FASEB J 2016; 30:2615-26. [PMID: 27059719 DOI: 10.1096/fj.201500159r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/28/2016] [Indexed: 11/11/2022]
Abstract
Serine proteinase inhibitor, clade E, member 2 (SERPINE2), is a cell- and extracellular matrix-associated inhibitor of thrombin. Although SERPINE2 is a candidate susceptibility gene for chronic obstructive pulmonary disease, the physiologic role of this protease inhibitor in lung development and homeostasis is unknown. We observed spontaneous monocytic-cell infiltration in the lungs of Serpine2-deficient (SE2(-/-)) mice, beginning at or before the time of lung maturity, which resulted in lesions that resembled bronchus-associated lymphoid tissue (BALT). The initiation of lymphocyte accumulation in the lungs of SE2(-/-) mice involved the excessive expression of chemokines, cytokines, and adhesion molecules that are essential for BALT induction, organization, and maintenance. BALT-like lesion formation in the lungs of SE2(-/-) mice was also associated with a significant increase in the activation of thrombin, a recognized target of SE2, and excess stimulation of NF-κB, a major regulator of chemokine expression and inflammation. Finally, systemic delivery of thrombin rapidly stimulated lung chemokine expression in vivo These data uncover a novel mechanism whereby loss of serine protease inhibition leads to lung lymphocyte accumulation.-Solleti, S. K., Srisuma, S., Bhattacharya, S., Rangel-Moreno, J., Bijli, K. M., Randall, T. D., Rahman, A., Mariani, T. J. Serpine2 deficiency results in lung lymphocyte accumulation and bronchus-associated lymphoid tissue formation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Sorachai Srisuma
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kaiser M Bijli
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University/Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Troy D Randall
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA; Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Arshad Rahman
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA;
| |
Collapse
|
50
|
Hampson JA, Stockley RA, Turner AM. Free light chains: potential biomarker and predictor of mortality in alpha-1-antitrypsin deficiency and usual COPD. Respir Res 2016; 17:34. [PMID: 27036487 PMCID: PMC4815123 DOI: 10.1186/s12931-016-0348-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/18/2016] [Indexed: 12/15/2022] Open
Abstract
Background Circulating free light chains (FLCs) can alter neutrophil migration, apoptosis and activation and may be a biomarker of autoimmune disease and adaptive immune system activation. These pathogenic roles could be relevant to lung disease in alpha 1 antitrypsin deficiency (A1ATD) and chronic obstructive pulmonary disease (COPD). Methods Total combined (c)FLCs were measured using the FreeLite® assay in 547 patients with A1ATD and 327 patients with usual COPD in the stable state, and assessed for association with clinical phenotype, disease severity, airway bacterial colonisation and mortality. Univariate and multivariate analyses were undertaken. Results Circulating cFLCs were static in the stable state when measured on 4 occasions in A1ATD and twice in usual COPD. Levels were inversely related to renal function (A1ATD and COPD p = <0.01), and higher in patients with chronic bronchitis (p = 0.019) and airway bacterial colonisation (p = 0.008). After adjusting for renal function and age the relationship between cFLCs and lung function was weak. Kaplan Meier curves showed that cFLC > normal (43.3 mg/L) significantly associated with mortality in both cohorts (A1ATD p = 0.001, COPD p = 0.013). Conclusions cFLCs may be a promising biomarker for risk stratification in A1ATD and COPD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0348-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Judith A Hampson
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, B15 2WB, UK.,ADAPT Project, University Hospital Birmingham, Birmingham, B15 2WB, UK
| | - Robert A Stockley
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, B15 2WB, UK.,ADAPT Project, University Hospital Birmingham, Birmingham, B15 2WB, UK
| | - Alice M Turner
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, B15 2WB, UK. .,ADAPT Project, University Hospital Birmingham, Birmingham, B15 2WB, UK. .,Heart of England NHS Foundation Trust, Birmingham, B9 5SS, UK.
| |
Collapse
|