Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
2.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33554955

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.


Subject(s)
Lung/immunology , Lung/microbiology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , alpha 1-Antitrypsin Deficiency/immunology , Animals , Female , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/etiology , Pneumonia, Pneumococcal/immunology , Pulmonary Emphysema/etiology , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/immunology , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin/pharmacokinetics , alpha 1-Antitrypsin/therapeutic use , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin Deficiency/genetics
3.
Curr Opin Allergy Clin Immunol ; 21(1): 46-51, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33284159

ABSTRACT

PURPOSE OF REVIEW: The aim of the article is to highlight the association between α1-antitrypsin deficiency (AATD) and asthma. RECENT FINDINGS: AATD is one of the most common and underrecognized autosomal disorders associated with an increased risk of developing liver and lung diseases. An association between α1-antitrypsin and asthma has been suggested, especially with severe forms of this disease. Many studies have shown an increased prevalence of asthma in the α1-antitrypsin-deficient population overtime (4-38%). The biological mechanism underlying these two conditions and able to bind them has not yet been well investigated. As α1-antitrypsin is the main inhibitor of the serine proteinase and it is an important anti-inflammatory protein with pronounced immunomodulatory activities, it can be hypothesized that the link between AATD and asthma might be represented by the elastase/antielastase imbalance and the proinflammatory effect that occurs because of the reduction of this protein. SUMMARY: There is a strong need for further researches to better understand the molecular mechanisms binding AATD and asthma. It is also recommendable to screen for AATD, late-onset asthma patients, and/or those with not fully reversible airways obstruction.


Subject(s)
Asthma/genetics , Leukocyte Elastase/metabolism , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin/genetics , Asthma/diagnosis , Asthma/immunology , Humans , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/immunology
4.
Front Immunol ; 11: 574410, 2020.
Article in English | MEDLINE | ID: mdl-33329539

ABSTRACT

Alpha 1 antitrypsin deficiency (AATD) is an autosomal co-dominant disorder characterized by a low level of circulating AAT, which significantly reduces protection for the lower airways against proteolytic burden caused by neutrophils. Neutrophils, which are terminally differentiated innate immune cells and play a critical role to clear pathogens, accumulate excessively in the lung of AATD individuals. The neutrophil burden in AATD individuals increases the risk for early-onset destructive lung diseases by producing neutrophil products such as reactive oxygen radicals and various proteases. The level of AAT in AATD individuals is not sufficient to inhibit the activity of neutrophil chemotactic factors such as CXCL-8 and LTB4, which could lead to alveolar neutrophil accumulation in AATD individuals. However, as neutrophils have a short lifespan, and apoptotic neutrophils are rapidly cleared by alveolar macrophages that outnumber the apoptotic neutrophils in the pulmonary alveolus, the increased chemotaxis activity does not fully explain the persistent neutrophil accumulation and the resulting chronic inflammation in AATD individuals. Here, we propose that the ability of alveolar macrophages to clear apoptotic neutrophils is impaired in AATD individuals and it could be the main driver to cause neutrophil accumulation in their lung. This study demonstrates that Z-AAT variant significantly increases the expression of pro-inflammatory cytokines including CXCL-8, CXCL1, LTB4, and TNFα in LPS-treated macrophages. These cytokines play a central role in neutrophil recruitment to the lung and in clearance of apoptotic neutrophils by macrophages. Our result shows that LPS treatment significantly reduces the efferocytosis ability of macrophages with the Z-AAT allele by inducing TNFα expression. We incubated monocyte-derived macrophages (MDMs) with apoptotic neutrophils and found that after 3 h of co-incubation, the expression level of CXCL-8 is reduced in M-MDMs but increased in Z-MDMs. This result shows that the expression of inflammatory cytokines could be increased by impaired efferocytosis. It indicates that the efferocytosis ability of macrophages plays an important role in regulating cytokine expression and resolving inflammation. Findings from this study would help us better understand the multifaceted effect of AAT on regulating neutrophil balance in the lung and the underlying mechanisms.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Neutrophils/immunology , Phagocytosis/immunology , alpha 1-Antitrypsin Deficiency/immunology , Chemotaxis, Leukocyte , Cytokines/metabolism , Genotype , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Neutrophils/metabolism , Phagocytosis/drug effects , Phagocytosis/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/immunology , alpha 1-Antitrypsin Deficiency/genetics
5.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32699193

ABSTRACT

The α-1-antitrypsin (or alpha-1-antitrypsin, A1AT) Z variant is the primary cause of severe A1AT deficiency and forms polymeric chains that aggregate in the endoplasmic reticulum of hepatocytes. Around 2%-5% of Europeans are heterozygous for the Z and WT M allele, and there is evidence of increased risk of liver disease when compared with MM A1AT individuals. We have shown that Z and M A1AT can copolymerize in cell models, but there has been no direct observation of heteropolymer formation in vivo. To this end, we developed a monoclonal antibody (mAb2H2) that specifically binds to M in preference to Z A1AT, localized its epitope using crystallography to a region perturbed by the Z (Glu342Lys) substitution, and used Fab fragments to label polymers isolated from an MZ heterozygote liver explant. Glu342 is critical to the affinity of mAb2H2, since it also recognized the mild S-deficiency variant (Glu264Val) present in circulating polymers from SZ heterozygotes. Negative-stain electron microscopy of the Fab2H2-labeled liver polymers revealed that M comprises around 6% of the polymer subunits in the MZ liver sample. These data demonstrate that Z A1AT can form heteropolymers with polymerization-inert variants in vivo with implications for liver disease in heterozygous individuals.


Subject(s)
Liver Cirrhosis/genetics , Protein Aggregates/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Alleles , Catalytic Domain/drug effects , Crystallography , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Epitopes/genetics , Epitopes/immunology , Genetic Variation/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Protein Aggregates/immunology , Protein Conformation , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/ultrastructure , alpha 1-Antitrypsin Deficiency/immunology , alpha 1-Antitrypsin Deficiency/pathology
6.
Can Respir J ; 2020: 4019608, 2020.
Article in English | MEDLINE | ID: mdl-32566054

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.


Subject(s)
Primary Immunodeficiency Diseases , Pulmonary Disease, Chronic Obstructive , Respiratory Tract Infections , alpha 1-Antitrypsin Deficiency , Bronchiectasis/diagnosis , Bronchiectasis/etiology , Diagnostic Errors/prevention & control , Female , Genetic Association Studies , Germany/epidemiology , Humans , Male , Middle Aged , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/epidemiology , Primary Immunodeficiency Diseases/physiopathology , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/diagnosis , Pulmonary Emphysema/etiology , Recurrence , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/immunology , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin Deficiency/diagnosis , alpha 1-Antitrypsin Deficiency/epidemiology , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/immunology
9.
Chest ; 158(5): 1919-1922, 2020 11.
Article in English | MEDLINE | ID: mdl-32417149
10.
Int J Chron Obstruct Pulmon Dis ; 14: 1879-1893, 2019.
Article in English | MEDLINE | ID: mdl-31686800

ABSTRACT

Background: Airway inflammation may drive the progression of chronic obstructive pulmonary disease (COPD) associated with alpha-1 antitrypsin deficiency (AATD), but the relationship between airway microbiota and inflammation has not been investigated. Methods: We studied 21 non-treated AATD (AATD-noT) patients, 20 AATD-COPD patients under augmentation therapy (AATD-AT), 20 cigarette smoke-associated COPD patients, 20 control healthy smokers (CS) and 21 non-smokers (CON) with normal lung function. We quantified sputum inflammatory cells and inflammatory markers (IL-27, CCL3, CCL5, CXCL8, LTB4, MPO) by ELISA, total bacterial load (16S) and pathogenic bacteria by qRT-PCR. Results: AATD-AT patients were younger but had similar spirometric and DLCO values compared to cigarette smoke-associated COPD, despite a lower burden of smoking history. Compared to cigarette smoke-associated COPD, AATD-noT and AATD-AT patients had lower sputum neutrophil levels (p=0.0446, p=0.0135), total bacterial load (16S) (p=0.0081, p=0.0223), M. catarrhalis (p=0.0115, p=0.0127) and S. pneumoniae (p=0.0013, p=0.0001). Sputum IL-27 was significantly elevated in CS and cigarette smoke-associated COPD. AATD-AT, but not AATD-noT patients, had IL-27 sputum levels (pg/ml) significantly lower than COPD (p=0.0297) and these positively correlated with FEV1% predicted values (r=0.578, p=0.0307). Conclusions: Compared to cigarette smoke-associated COPD, AATD-AT (COPD) patients have a distinct airway inflammatory and microbiological profile. The decreased sputum bacterial load and IL-27 levels in AATD-AT patients suggests that augmentation therapy play a role in these changes.


Subject(s)
Bacteria/isolation & purification , Inflammation Mediators/analysis , Lung/immunology , Lung/microbiology , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , alpha 1-Antitrypsin Deficiency/complications , Aged , Bacteria/genetics , Bacteria/pathogenicity , Bacterial Load , Case-Control Studies , Female , Host-Pathogen Interactions , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Risk Factors , Sputum/immunology , Sputum/microbiology , alpha 1-Antitrypsin Deficiency/diagnosis , alpha 1-Antitrypsin Deficiency/immunology , alpha 1-Antitrypsin Deficiency/microbiology
11.
Biologicals ; 62: 77-84, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31522909

ABSTRACT

Multiple analytical and preclinical studies were performed to compare the biochemical characteristics, pharmacokinetics (PK), safety and neoantigenicity of a new 5% liquid formulation of Alpha-1 Proteinase Inhibitor (Liquid A1PI, Prolastin®-C Liquid) with the lyophilized version (Lyophilized A1PI, Prolastin®-C). Liquid A1PI and Lyophilized A1PI had similar average mass (~52 kDa), and both forms exhibited glycoform patterns consistent with the known banding pattern of A1PI (dominated by the M6 and M4 bands, including deconvoluted masses). Both Liquid A1PI and Lyophilized A1PI yielded average percent purity values ranging from 96% to 99% and had active content ranging from 53  mg/mL to 59  mg/mL. The PK profile of Liquid A1PI was similar to Lyophilized A1PI. Safety assessments in rabbits showed good tolerability and no test article-related changes in mortality, clinical signs, clinical pathology, body weight, food consumption, or urinalysis parameters. Following immunodepletion of antibodies that recognize Lyophilized A1PI, there were no significant differences in the anti-drug titers among animals immunized with Lyophilized A1PI and Liquid A1PI (p > 0.05), indicating that no antibodies to neoantigens were generated. Liquid A1PI and Lyophilized A1PI have similar profiles with respect to biochemical characteristics, PK, safety and neoantigenicity.


Subject(s)
alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin , Animals , Antibodies/blood , Antibodies/immunology , Freeze Drying , Humans , Rabbits , alpha 1-Antitrypsin/adverse effects , alpha 1-Antitrypsin/immunology , alpha 1-Antitrypsin/pharmacokinetics , alpha 1-Antitrypsin/pharmacology , alpha 1-Antitrypsin Deficiency/blood , alpha 1-Antitrypsin Deficiency/immunology
12.
Sci Rep ; 9(1): 13088, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511551

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.


Subject(s)
Exome Sequencing , Genetic Predisposition to Disease/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Molecular Sequence Annotation
13.
Front Immunol ; 10: 1417, 2019.
Article in English | MEDLINE | ID: mdl-31293581

ABSTRACT

Rationale: The association between non-tuberculous mycobacterial lung disease and alpha-1-antitrypsin (AAT) deficiency is likely due, in part, to underlying emphysema or bronchiectasis. But there is increasing evidence that AAT itself enhances host immunity against microbial pathogens and thus deficiency could compromise host protection. Objectives: The goal of this project is to determine if AAT could augment macrophage activity against non-tuberculous mycobacteria. Methods: We compared the ability of monocyte-derived macrophages cultured in autologous plasma that were obtained immediately before and soon after AAT infusion-given to individuals with AAT deficiency-to control an ex vivo Mycobacterium intracellulare infection. Measurements and Main Results: We found that compared to pre-AAT infused monocyte-derived macrophages plus plasma, macrophages, and contemporaneous plasma obtained after a session of AAT infusion were significantly better able to control M. intracellulare infection; the reduced bacterial burden was linked with greater phagosome-lysosome fusion and increased autophagosome formation/maturation, the latter due to AAT inhibition of both M. intracellulare-induced nuclear factor-kappa B activation and A20 expression. While there was a modest increase in apoptosis in the M. intracellulare-infected post-AAT infused macrophages and plasma, inhibiting caspase-3 in THP-1 cells, monocyte-derived macrophages, and alveolar macrophages unexpectedly reduced the M. intracellulare burden, indicating that apoptosis impairs macrophage control of M. intracellulare and that the host protective effects of AAT occurred despite inducing apoptosis. Conclusion: AAT augments macrophage control of M. intracellulare infection through enhancing phagosome-lysosome fusion and autophagy.


Subject(s)
Macrophages, Alveolar/immunology , Mycobacterium avium Complex/immunology , Mycobacterium avium-intracellulare Infection/immunology , alpha 1-Antitrypsin Deficiency/immunology , alpha 1-Antitrypsin/immunology , Autophagy/immunology , Bronchiectasis/etiology , Emphysema/etiology , Humans , Lung Diseases/immunology , Lung Diseases/microbiology , Macrophage Activation/immunology , Phagosomes/immunology , Transcription Factor RelA/metabolism , alpha 1-Antitrypsin Deficiency/pathology
14.
J Immunol ; 202(8): 2240-2253, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30796179

ABSTRACT

Alpha-1 antitrypsin (AAT) is an acute phase protein that possesses immune-regulatory and anti-inflammatory functions independent of antiprotease activity. AAT deficiency (AATD) is associated with early-onset emphysema and chronic obstructive pulmonary disease. Of interest are the AATD nonsense mutations (termed null or Q0), the majority of which arise from premature termination codons in the mRNA coding region. We have recently demonstrated that plasma from an AATD patient homozygous for the Null Bolton allele (Q0bolton ) contains AAT protein of truncated size. Although the potential to alleviate the phenotypic consequences of AATD by increasing levels of truncated protein holds therapeutic promise, protein functionality is key. The goal of this study was to evaluate the structural features and anti-inflammatory capacity of Q0bolton-AAT. A low-abundance, truncated AAT protein was confirmed in plasma of a Q0bolton-AATD patient and was secreted by patient-derived induced pluripotent stem cell-hepatic cells. Functional assays confirmed the ability of purified Q0bolton-AAT protein to bind neutrophil elastase and to inhibit protease activity. Q0bolton-AAT bound IL-8 and leukotriene B4, comparable to healthy control M-AAT, and significantly decreased leukotriene B4-induced neutrophil adhesion (p = 0.04). Through a mechanism involving increased mRNA stability (p = 0.007), ataluren treatment of HEK-293 significantly increased Q0bolton-AAT mRNA expression (p = 0.03) and Q0bolton-AAT truncated protein secretion (p = 0.04). Results support the rationale for treatment with pharmacological agents that augment levels of functional Q0bolton-AAT protein, thus offering a potential therapeutic option for AATD patients with rare mutations of similar theratype.


Subject(s)
Alleles , Codon, Nonsense , alpha 1-Antitrypsin Deficiency , alpha 1-Antitrypsin , Adult , Female , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Liver/immunology , Liver/metabolism , Male , alpha 1-Antitrypsin/blood , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/immunology , alpha 1-Antitrypsin Deficiency/blood , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/immunology
15.
Respiration ; 95(2): 114-121, 2018.
Article in English | MEDLINE | ID: mdl-29253843

ABSTRACT

BACKGROUND: α1-Antitrypsin deficiency (AATD) predisposes to chronic obstructive pulmonary disease (COPD). In COPD unrelated to AATD, the role of a higher blood eosinophil count in disease and subsequent personalization of therapy has recently received much attention. We sought to investigate this concept in patients with AATD-associated COPD. OBJECTIVES: The study aims to evaluate eosinophilia status against outcomes including mortality and FEV1 decline in patients with AATD. METHODS: All patients with PiSZ and PiZZ genotypes were identified from the UK AATD registry. The participants were substratified according to inhaled corticosteroid (ICS) use. Blood eosinophil counts were assessed from baseline and annually during follow-up (range 1-18 years). Eosinophilia was defined as a level >0.2 × 109/L, and classified by the frequency of such counts into "always," "intermittent," or "never present." Univariate and multivariate analyses were conducted. RESULTS: In total, 646 participants were included, 53.9% of whom demonstrated intermittent and 7.4% persistent eosinophilia. Survival did not differ according to eosinophilic group (p > 0.05). Those with persistent eosinophilia showed a slower FEV1 decline (p < 0.001). There was no clear association with exacerbation frequency. Patients on ICS at baseline were more likely to be eosinophilic (p = 0.002) and having a lower FEV1 (p < 0.001) and greater pack-year exposure (16.5 vs. 7.8 pack-years, p < 0.001). When the multivariate analyses of FEV1 decline were stratified for baseline ICS use, the association of persistent eosinophilia with slower decline persisted in those on ICS. CONCLUSIONS: Blood eosinophil levels persistently >0.2 × 109/L may be an indication for ICS use in PiZZ AATD in order to reduce FEV1 decline.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Eosinophilia , Forced Expiratory Volume/drug effects , Pulmonary Disease, Chronic Obstructive/immunology , alpha 1-Antitrypsin Deficiency/immunology , Administration, Inhalation , Adult , Aged , Female , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/drug therapy , Registries , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin Deficiency/drug therapy
16.
Proc Natl Acad Sci U S A ; 114(7): 1655-1659, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137880

ABSTRACT

Adeno-associated virus (AAV)-mediated gene therapy is currently being pursued as a treatment for the monogenic disorder α-1-antitrypsin (AAT) deficiency. Results from phase I and II studies have shown relatively stable and dose-dependent increases in transgene-derived wild-type AAT after local intramuscular vector administration. In this report we describe the appearance of transgene-specific T-cell responses in two subjects that were part of the phase II trial. The patient with the more robust T-cell response, which was associated with a reduction in transgene expression, was characterized more thoroughly in this study. We learned that the AAT-specific T cells in this patient were cytolytic in phenotype, mapped to a peptide in the endogenous mutant AAT protein that contained a common polymorphism not incorporated into the transgene, and were restricted by a rare HLA class I C alleles present only in this patient. These human studies illustrate the genetic influence of the endogenous gene and HLA haplotype on the outcome of gene therapy.


Subject(s)
Genetic Therapy/methods , Peptides/immunology , T-Lymphocytes/immunology , alpha 1-Antitrypsin Deficiency/therapy , alpha 1-Antitrypsin/immunology , Adult , Aged , Alleles , Amino Acid Sequence , Dependovirus/genetics , Female , HLA Antigens/genetics , HLA Antigens/immunology , Humans , K562 Cells , Male , Middle Aged , Peptides/genetics , Peptides/metabolism , Polymorphism, Genetic , T-Lymphocytes/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/immunology
17.
Ann Am Thorac Soc ; 13 Suppl 4: S297-304, 2016 08.
Article in English | MEDLINE | ID: mdl-27564664

ABSTRACT

Alpha-1 antitrypsin deficiency (AATD) is characterized by low levels of circulating alpha-1 antitrypsin and an increased risk for emphysema, liver disease, and panniculitis. The reduced levels of alpha-1 antitrypsin in AATD predispose the lung to unopposed proteolytic activity, predominantly from neutrophil-derived proteases, chiefly neutrophil elastase. This leads to emphysema. The mechanisms subtending the liver disease are less well understood, but are probably due to a "gain-of function" inflammatory process in the liver, stoked by intracellular retention of aberrantly folded alpha-1 antitrypsin. The panniculitis associated with AATD is most likely due to unopposed proteolytic activity in the skin. Although AATD has been traditionally viewed as a condition arising from a protease-antiprotease imbalance in the lung, it is increasingly recognized that AATD is an inflammatory disorder, both in the lung and in the extrapulmonary manifestations associated with the condition. This inflammation is predominantly neutrophil driven, and there are several alpha-1 antitrypsin-related mechanisms involved in potentiating this neutrophilic response. The rationale for AAT augmentation therapy in AATD is classically based on restoring the antiprotease balance in the lung, but its beneficial effects may also be exerted systemically, further exposing the pathogenesis of AATD-related disease and indicating a potential usage for alpha-1 antitrypsin in other inflammatory conditions.


Subject(s)
Lung/immunology , Neutrophils/immunology , alpha 1-Antitrypsin Deficiency/immunology , alpha 1-Antitrypsin/immunology , Humans , Leukocyte Elastase/immunology , Liver Diseases/etiology , Liver Diseases/immunology , Panniculitis/etiology , Panniculitis/immunology , Pulmonary Emphysema/etiology , Pulmonary Emphysema/immunology , Tumor Necrosis Factor-alpha/immunology , alpha 1-Antitrypsin Deficiency/complications
18.
Ann Am Thorac Soc ; 13 Suppl 4: S305-10, 2016 08.
Article in English | MEDLINE | ID: mdl-27564665

ABSTRACT

From the discovery that alpha-1 antitrypsin (AAT) was an effective inhibitor of neutrophil elastase originated the classic paradigm of protease/antiprotease imbalance, linking lung destruction to the unopposed effect of proteases in patients with the deficiency. Notwithstanding its importance as an antiprotease, it has become evident that alpha-1 antitrypsin has important antiinflammatory and immune-regulatory activities, which may be critically involved in lung destruction. We review here recent evidence showing that, indeed, an important adaptive immune reaction is present in lungs with AAT deficiency, similar to the one seen in severe chronic obstructive pulmonary disease with normal AAT. On the basis of recent evidence from epidemiological, clinical, and pathogenetic studies, it is likely time to move on from the original protease/antiprotease hypothesis for the production of emphysema toward a more complex paradigm, involving the antiinflammatory and immune modulating functions of AAT.


Subject(s)
Adaptive Immunity/immunology , Peptide Hydrolases/metabolism , Protease Inhibitors/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , alpha 1-Antitrypsin Deficiency/immunology , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/metabolism
19.
Respiration ; 91(5): 380-5, 2016.
Article in English | MEDLINE | ID: mdl-27164860

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.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Lung/immunology , Pulmonary Emphysema/immunology , Smoking/immunology , alpha 1-Antitrypsin Deficiency/immunology , alpha 1-Antitrypsin/immunology , Humans , Lung/enzymology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Emphysema/enzymology , alpha 1-Antitrypsin Deficiency/enzymology
20.
Ann Am Thorac Soc ; 13 Suppl 2: S123-9, 2016 04.
Article in English | MEDLINE | ID: mdl-27115946

ABSTRACT

The neutrophil is a powerful cellular defender of the vulnerable interface between the environment and pulmonary tissues. This cell's potent weapons are carefully calibrated in the healthy state to maximize effectiveness in fighting pathogens while minimizing tissue damage and allowing for repair of what damage does occur. The three related chronic airway disorders of cystic fibrosis, non-cystic fibrosis bronchiectasis, and alpha-1 antitrypsin deficiency all demonstrate significant derangements of this homeostatic system that result in their respective pathologies. An important shared feature among them is the inefficient resolution of chronic inflammation that serves as a central means for neutrophil-driven lung damage resulting in disease progression. Examining the commonalities and divergences between these diseases in the light of their immunopathology is informative and may help guide us toward future therapeutics designed to modulate the neutrophil's interplay with the pulmonary environment.


Subject(s)
Bronchiectasis/immunology , Cystic Fibrosis/immunology , Lung/immunology , Neutrophils/immunology , alpha 1-Antitrypsin Deficiency/immunology , Disease Progression , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...