In vitro and in vivo modulation of NADPH oxidase activity and reactive oxygen species production in human neutrophils by α1-antitrypsin

Oxidative stress from innate immune cells is a driving mechanism that underlies COPD pathogenesis. Individuals with α-1 antitrypsin (AAT) deficiency (AATD) have a dramatically increased risk of developing COPD. To understand this further, the aim of this study was to investigate whether AATD presents with altered neutrophil NADPH oxidase activation, due to the specific lack of plasma AAT. Experiments were performed using circulating neutrophils isolated from healthy controls and individuals with AATD. Superoxide anion (O2−) production was determined from the rate of reduction of cytochrome c. Quantification of membrane NADPH oxidase subunits was performed by mass spectrometry and Western blot analysis. The clinical significance of our in vitro findings was assessed in patients with AATD and severe COPD receiving intravenous AAT replacement therapy. In vitro, AAT significantly inhibited O2− production by stimulated neutrophils and suppressed receptor stimulation of cyclic adenosine monophosphate and extracellular signal-regulated kinase (ERK)1/2 phosphorylation. In addition, AAT reduced plasma membrane translocation of cytosolic phox components of the NADPH oxidase. Ex vivo, AATD neutrophils demonstrated increased plasma membrane-associated p67phox and p47phox and significantly increased O2− production. The described variance in phox protein membrane assembly was resolved post-AAT augmentation therapy in vivo, the effects of which significantly reduced AATD neutrophil O2− production to that of healthy control cells. These results expand our knowledge on the mechanism of neutrophil-driven airways disease associated with AATD. Therapeutic AAT augmentation modified neutrophil NADPH oxidase assembly and reactive oxygen species production, with implications for clinical use in conditions in which oxidative stress plays a pathogenic role.

[1]  F. Blasi,et al.  Neutrophil extracellular traps, disease severity, and antibiotic response in bronchiectasis: an international, observational, multicohort study. , 2021, The Lancet. Respiratory medicine.

[2]  N. McElvaney,et al.  Altered Degranulation and pH of Neutrophil Phagosomes Impacts Antimicrobial Efficiency in Cystic Fibrosis , 2020, Frontiers in Immunology.

[3]  N. McElvaney,et al.  Anti-cytokines as a Strategy in Alpha-1 Antitrypsin Deficiency. , 2020, Chronic obstructive pulmonary diseases.

[4]  M. Henry,et al.  α1 Antitrypsin therapy modulates the neutrophil membrane proteome and secretome , 2020, European Respiratory Journal.

[5]  R. Stockley,et al.  The Biological Effects of Double-Dose Alpha-1 Antitrypsin Augmentation Therapy. A Pilot Clinical Trial , 2019, American journal of respiratory and critical care medicine.

[6]  M. Henry,et al.  Glycosylation Repurposes Alpha‐1 Antitrypsin for Resolution of Community‐acquired Pneumonia , 2017, American journal of respiratory and critical care medicine.

[7]  J. Mecsas,et al.  Neutrophils to the ROScue: Mechanisms of NADPH Oxidase Activation and Bacterial Resistance , 2017, Front. Cell. Infect. Microbiol..

[8]  N. McElvaney,et al.  The Role of Neutrophils in Alpha-1 Antitrypsin Deficiency. , 2016, Annals of the American Thoracic Society.

[9]  M. Wormald,et al.  The BLT1 Inhibitory Function of α-1 Antitrypsin Augmentation Therapy Disrupts Leukotriene B4 Neutrophil Signaling , 2015, The Journal of Immunology.

[10]  Berend C Stoel,et al.  Intravenous augmentation treatment and lung density in severe α1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial , 2015, The Lancet.

[11]  N. McElvaney,et al.  Alpha-1 Antitrypsin Augmentation Therapy Corrects Accelerated Neutrophil Apoptosis in Deficient Individuals , 2014, The Journal of Immunology.

[12]  M. Henry,et al.  A neutrophil intrinsic impairment affecting Rab27a and degranulation in cystic fibrosis is corrected by CFTR potentiator therapy. , 2014, Blood.

[13]  N. McElvaney,et al.  The Circulating Proteinase Inhibitor α-1 Antitrypsin Regulates Neutrophil Degranulation and Autoimmunity , 2014, Science Translational Medicine.

[14]  P. Kirkham,et al.  Oxidative stress in COPD. , 2013, Chest.

[15]  R. Stockley,et al.  Behavioral and structural differences in migrating peripheral neutrophils from patients with chronic obstructive pulmonary disease. , 2011, American journal of respiratory and critical care medicine.

[16]  N. McElvaney,et al.  The Cystic Fibrosis Neutrophil: A Specialized Yet Potentially Defective Cell , 2011, Archivum Immunologiae et Therapiae Experimentalis.

[17]  M. Henry,et al.  α-1 Antitrypsin regulates human neutrophil chemotaxis induced by soluble immune complexes and IL-8. , 2010, The Journal of clinical investigation.

[18]  Lin Ying Liu,et al.  Human Airway Eosinophils Respond to Chemoattractants with Greater Eosinophil-Derived Neurotoxin Release, Adherence to Fibronectin, and Activation of the Ras–ERK Pathway When Compared with Blood Eosinophils , 2010, The Journal of Immunology.

[19]  N. McElvaney,et al.  Evidence for Unfolded Protein Response Activation in Monocytes from Individuals with α-1 Antitrypsin Deficiency , 2010, The Journal of Immunology.

[20]  P. King The pathophysiology of bronchiectasis , 2009, International journal of chronic obstructive pulmonary disease.

[21]  N. McElvaney,et al.  Proteases and antiproteases in chronic neutrophilic lung disease – relevance to drug discovery , 2009, British journal of pharmacology.

[22]  I. Petrache,et al.  Mechanism of α‐1 antitrypsin endocytosis by lung endothelium , 2009, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[23]  J. Fahy,et al.  Eosinophilic and neutrophilic inflammation in asthma: insights from clinical studies. , 2009, Proceedings of the American Thoracic Society.

[24]  Naoko Nakamura,et al.  A Feeder‐Free and Efficient Production of Functional Neutrophils from Human Embryonic Stem Cells , 2009, Stem cells.

[25]  Hongwei Yao,et al.  Future therapeutic treatment of COPD: Struggle between oxidants and cytokines , 2007, International journal of chronic obstructive pulmonary disease.

[26]  J. Roca,et al.  Oxidative stress and airway inflammation in severe exacerbations of COPD , 2005, Thorax.

[27]  A. Churg,et al.  alpha-1-Antitrypsin ameliorates cigarette smoke-induced emphysema in the mouse. , 2003, American journal of respiratory and critical care medicine.

[28]  P. Vignais The superoxide-generating NADPH oxidase: structural aspects and activation mechanism , 2002, Cellular and Molecular Life Sciences CMLS.

[29]  Giorgio Gabella,et al.  Killing activity of neutrophils is mediated through activation of proteases by K+ flux , 2002, Nature.

[30]  J. Moss,et al.  Oxidation of either methionine 351 or methionine 358 in alpha 1-antitrypsin causes loss of anti-neutrophil elastase activity. , 2000, The Journal of biological chemistry.

[31]  Farshid N. Rouhani,et al.  Lung neutrophil burden correlates with increased pro-inflammatory cytokines and decreased lung function in individuals with alpha(1)-antitrypsin deficiency , 2000, Chest.

[32]  A. Mócsai,et al.  Kinase Pathways in Chemoattractant-Induced Degranulation of Neutrophils: The Role of p38 Mitogen-Activated Protein Kinase Activated by Src Family Kinases1 , 2000, The Journal of Immunology.

[33]  T. Yoneda,et al.  Airway inflammation in COPD assessed by sputum levels of interleukin-8. , 1997, Chest.

[34]  J. Potempa,et al.  Biosynthesis of α1-Proteinase Inhibitor by Human Lung-derived Epithelial Cells* , 1997, The Journal of Biological Chemistry.

[35]  D. Lomas,et al.  α1-antitrypsin deficiency , 2005, The Lancet.

[36]  R. Snyderman,et al.  Functional high efficiency expression of cloned leucocyte chemoattractant receptor cDNAs , 1992, FEBS letters.

[37]  R. Crystal,et al.  Use of a Highly Purified α1-Antitrypsin Standard to Establish Ranges for the Common Normal and Deficient α1-Antitrypsin Phenotypes , 1991 .

[38]  R. Crystal,et al.  Neutrophil accumulation in the lung in alpha 1-antitrypsin deficiency. Spontaneous release of leukotriene B4 by alveolar macrophages. , 1991, The Journal of clinical investigation.

[39]  V. Ferrans,et al.  Human neutrophils express the alpha 1-antitrypsin gene and produce alpha 1-antitrypsin. , 1991, Blood.

[40]  J. Pierce,et al.  The alpha 1-antitrypsin gene and emphysema. , 1989, The American journal of physiology.

[41]  L. Koenderman,et al.  Eosinophils do respond to fMLP. , 1987, Blood.

[42]  R. Crystal,et al.  Replacement therapy for alpha 1-antitrypsin deficiency associated with emphysema. , 1987, The New England journal of medicine.

[43]  R. Crystal,et al.  Antielastases of the human alveolar structures. Implications for the protease-antiprotease theory of emphysema. , 1981, The Journal of clinical investigation.

[44]  M. Sanak,et al.  Long-term efficacy and safety of α1 proteinase inhibitor treatment for emphysema caused by severe α1 antitrypsin deficiency: an open-label extension trial (RAPID-OLE). , 2017, The Lancet. Respiratory medicine.

[45]  Michael J Morgan,et al.  Crosstalk of reactive oxygen species and NF-κB signaling , 2011, Cell Research.

[46]  N. Porchet,et al.  Evaluation of a new Sebia isoelectrofocusing kit for α1-antitrypsin phenotyping with the Hydrasys® System , 2008, Clinical chemistry and laboratory medicine.

[47]  A. Segal,et al.  How neutrophils kill microbes. , 2005, Annual review of immunology.

[48]  J. El-Benna The Mitogen-Activated Protein Kinase Extracellular Signal-Regulated Kinase 1/2 Pathway Is Involved in formyl-Methionyl-Leucyl-Phenylalanine-Induced p47 Phosphorylation in Human Neutrophils , 2000 .

[49]  O. Götze,et al.  The biologic role of interleukin-8: functional analysis and expression of CXCR1 and CXCR2 on human eosinophils. , 1999, Blood.

[50]  D. Blake,et al.  Inhibition of neutrophil superoxide production by human plasma alpha 1-antitrypsin. , 1992, FEBS letters.

[51]  A. Solomon,et al.  Alpha 1-antitrypsin deficiency associated with panniculitis. , 1988, Journal of the American Academy of Dermatology.