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
|
Sun S, Wang C, Hu J, Zhao P, Wang X, Balch WE. Spatial covariance reveals isothiocyanate natural products adjust redox stress to restore function in alpha-1-antitrypsin deficiency. Cell Rep Med 2025; 6:101917. [PMID: 39809267 PMCID: PMC11866504 DOI: 10.1016/j.xcrm.2024.101917] [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: 08/24/2023] [Revised: 06/09/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
Alpha-1 antitrypsin (AAT) deficiency (AATD) is a monogenic disease caused by misfolding of AAT variants resulting in gain-of-toxic aggregation in the liver and loss of monomer activity in the lung leading to chronic obstructive pulmonary disease (COPD). Using high-throughput screening, we discovered a bioactive natural product, phenethyl isothiocyanate (PEITC), highly enriched in cruciferous vegetables, including watercress and broccoli, which improves the level of monomer secretion and neutrophil elastase (NE) inhibitory activity of AAT-Z through the endoplasmic reticulum (ER) redox sensor protein disulfide isomerase (PDI) A4 (PDIA4). The intracellular polymer burden of AAT-Z can be managed by combination treatment of PEITC and an autophagy activator. Using Gaussian process (GP)-based spatial covariance (SCV) (GP-SCV) machine learning to map on a residue-by-residue basis at atomic resolution all variants in the worldwide AATD clinical population, we reveal a global rescue of monomer secretion and NE inhibitory activity for most variants triggering disease. We present a proof of concept that GP-SCV mapping of restoration of AAT variant function serves as a standard model to discover natural products such as the anti-oxidant PEITC that could potentially impact the redox/inflammatory environment of the ER to provide a nutraceutical approach to help minimize disease in AATD patients.
Collapse
Affiliation(s)
- Shuhong Sun
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, Nanjing 211166, China.
| | - Chao Wang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Junyan Hu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Pei Zhao
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Xi Wang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - William E Balch
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Balderacchi AM, Bignotti M, Ottaviani S, Denardo A, Barzon V, Ben Khlifa E, Vailati G, Piloni D, Benini F, Corda L, Corsico AG, Ferrarotti I, Fra A. Quantification of circulating alpha-1-antitrypsin polymers associated with different SERPINA1 genotypes. Clin Chem Lab Med 2024; 62:1980-1990. [PMID: 38407261 DOI: 10.1515/cclm-2023-1348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVES Alpha-1-antitrypsin deficiency is a genetic disorder caused by mutations in the SERPINA1 gene encoding alpha-1-antitrypsin (AAT), the major serine protease inhibitor in plasma. Reduced AAT levels are associated with elevated risk of developing emphysema mainly due to uncontrolled activity of neutrophil elastase in the lungs. The prevalent Z-AAT mutant and many rare pathogenic AAT variants also predispose to liver disease due to their accumulation as polymeric chains in hepatocytes. Part of these polymers are secreted into the bloodstream and could represent biomarkers of intra-hepatic accumulation. Moreover, being inactive, they further lower lung protection against proteases. Aim of our study is to accurately quantify the percentage of circulating polymers (CP) in a cohort of subjects with different SERPINA1 genotypes. METHODS CP concentration was measured in plasma or Dried Blood Spot (DBS) by a sensitive sandwich ELISA based on capture by the polymer-specific 2C1 monoclonal antibody. RESULTS CP were significantly elevated in patients with the prevalent PI*SZ and PI*ZZ genotypes, with considerable intra-genotype variability. Notably, higher percentage of polymers was observed in association with elevated C-reactive protein. CP levels were also increased in carriers of the Mmalton variant, and of Mprocida, I, Plowell and Mherleen in heterozygosity with Z-AAT. CONCLUSIONS These findings highlight the importance of implementing CP quantification in a clinical laboratory. Indeed, the variable amount of CP in patients with the same genotype may correlate with the variable severity of the associated lung and liver diseases. Moreover, CP can reveal the polymerogenic potential of newly discovered ultrarare AAT variants.
Collapse
Affiliation(s)
- Alice M Balderacchi
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, UOC Pulmonology, 18631Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mattia Bignotti
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 9297University of Brescia, Brescia, Italy
| | - Stefania Ottaviani
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, UOC Pulmonology, 18631Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Denardo
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 9297University of Brescia, Brescia, Italy
| | - Valentina Barzon
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, 19001University of Pavia, Pavia, Italy
| | - Emna Ben Khlifa
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 9297University of Brescia, Brescia, Italy
| | - Guido Vailati
- Referral Centre for Alpha-1 Antitrypsin Deficiency, 18515 Spedali Civili , Brescia, Italy
| | - Davide Piloni
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, UOC Pulmonology, 18631Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federica Benini
- Referral Centre for Alpha-1 Antitrypsin Deficiency, 18515 Spedali Civili , Brescia, Italy
| | - Luciano Corda
- Referral Centre for Alpha-1 Antitrypsin Deficiency, 18515 Spedali Civili , Brescia, Italy
| | - Angelo G Corsico
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, UOC Pulmonology, 18631Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, 19001University of Pavia, Pavia, Italy
| | - Ilaria Ferrarotti
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, UOC Pulmonology, 18631Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, 19001University of Pavia, Pavia, Italy
| | - Annamaria Fra
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 9297University of Brescia, Brescia, Italy
| |
Collapse
|
4
|
Kamuda K, Ronzoni R, Majumdar A, Guan FHX, Irving JA, Lomas DA. A novel pathological mutant reveals the role of torsional flexibility in the serpin breach in adoption of an aggregation-prone intermediate. FEBS J 2024; 291:2937-2954. [PMID: 38523412 PMCID: PMC11753496 DOI: 10.1111/febs.17121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/17/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024]
Abstract
Mutants of alpha-1-antitrypsin cause the protein to self-associate and form ordered aggregates ('polymers') that are retained within hepatocytes, resulting in a predisposition to the development of liver disease. The associated reduction in secretion, and for some mutants, impairment of function, leads to a failure to protect lung tissue against proteases released during the inflammatory response and an increased risk of emphysema. We report here a novel deficiency mutation (Gly192Cys), that we name the Sydney variant, identified in a patient in heterozygosity with the Z allele (Glu342Lys). Cellular analysis revealed that the novel variant was mostly retained as insoluble polymers within the endoplasmic reticulum. The basis for this behaviour was investigated using biophysical and structural techniques. The variant showed a 40% reduction in inhibitory activity and a reduced stability as assessed by thermal unfolding experiments. Polymerisation involves adoption of an aggregation-prone intermediate and paradoxically the energy barrier for transition to this state was increased by 16% for the Gly192Cys variant with respect to the wild-type protein. However, with activation to the intermediate state, polymerisation occurred at a 3.8-fold faster rate overall. X-ray crystallography provided two crystal structures of the Gly192Cys variant, revealing perturbation within the 'breach' region with Cys192 in two different orientations: in one structure it faces towards the hydrophobic core while in the second it is solvent-exposed. This orientational heterogeneity was confirmed by PEGylation. These data show the critical role of the torsional freedom imparted by Gly192 in inhibitory activity and stability against polymerisation.
Collapse
Affiliation(s)
- Kamila Kamuda
- Division of Medicine, UCL Respiratory, Rayne InstituteUniversity College LondonUK
- Institute of Structural and Molecular Biology, Birkbeck CollegeUniversity College LondonUK
| | - Riccardo Ronzoni
- Division of Medicine, UCL Respiratory, Rayne InstituteUniversity College LondonUK
- Institute of Structural and Molecular Biology, Birkbeck CollegeUniversity College LondonUK
| | - Avik Majumdar
- AW Morrow Gastroenterology and Liver CentreRoyal Prince Alfred HospitalSydneyAustralia
- Victorian Liver Transplant UnitAustin HealthMelbourneAustralia
- The University of MelbourneMelbourneAustralia
| | - Fiona H. X. Guan
- AW Morrow Gastroenterology and Liver CentreRoyal Prince Alfred HospitalSydneyAustralia
| | - James A. Irving
- Division of Medicine, UCL Respiratory, Rayne InstituteUniversity College LondonUK
- Institute of Structural and Molecular Biology, Birkbeck CollegeUniversity College LondonUK
| | - David A. Lomas
- Division of Medicine, UCL Respiratory, Rayne InstituteUniversity College LondonUK
- Institute of Structural and Molecular Biology, Birkbeck CollegeUniversity College LondonUK
| |
Collapse
|
5
|
Denardo A, Ben Khlifa E, Bignotti M, Fra A. Characterization of Novel Alpha-1-Antitrypsin Coding Variants in a Mammalian Cellular Model. Methods Mol Biol 2024; 2750:79-93. [PMID: 38108969 DOI: 10.1007/978-1-0716-3605-3_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Advances in genetic screening technologies have considerably accelerated the discovery of rare alpha-1-antitrypsin (AAT) variants. Expression in cellular models is an effective approach to evaluate the pathogenic potential of these new AAT variants, whose clinical significance would otherwise remain uncertain. Here we provide a detailed description of established methods for in vitro characterization of AAT coding variants expressed in HEK293T/17 cells. The protocols include determination of secretion efficiency, the tendency to form polymeric chains and the anti-elastase inhibitory activity.
Collapse
Affiliation(s)
- Andrea Denardo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emna Ben Khlifa
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Bignotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
6
|
Ottaviani S, Bartoli G, Carroll TP, Gangemi F, Balderacchi AM, Barzon V, Corino A, Piloni D, McElvaney NG, Corsico AG, Irving JA, Fra A, Ferrarotti I. Comprehensive Clinical Diagnostic Pipelines Reveal New Variants in Alpha-1 Antitrypsin Deficiency. Am J Respir Cell Mol Biol 2023; 69:355-366. [PMID: 37071847 DOI: 10.1165/rcmb.2022-0470oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/18/2023] [Indexed: 04/20/2023] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is an underdiagnosed disorder associated with mutations in the SERPINA1 gene encoding alpha-1 antitrypsin (AAT). Severe AATD can manifest as pulmonary emphysema and progressive liver disease. Besides the most common pathogenic variants S (E264V) and Z (E342K), many rarer genetic variants of AAT have been found in patients and in the general population. Here we report a panel of new SERPINA1 variants, including 4 null and 16 missense alleles, identified among a cohort of individuals with suspected AATD whose phenotypic follow-up showed inconclusive or atypical results. Because the pathogenic significance of the missense variants was unclear purely on the basis of clinical data, the integration of computational, biochemical, and cellular studies was used to define the associated risk of disease. Established pathogenicity predictors and structural analysis identified a panel of candidate damaging mutations that were characterized by expression in mammalian cell models. Polymer formation, intracellular accumulation, and secretory efficiency were evaluated experimentally. Our results identified two AAT mutants with a Z-like polymerogenic severe deficiency profile (Smilano and Mcampolongo) and three milder variants (Xsarezzo, Pdublin, and Ctiberias). Overall, the experimentally determined behavior of the variants was in agreement with the pathogenicity scores of the REVEL (an ensemble method for predicting the pathogenicity of rare missense variants) predictor, supporting the utility of this bioinformatic tool in the initial assessment of newly identified amino acid substitutions of AAT. Our study, in addition to describing 20 new SERPINA1 variants, provides a model for a multidisciplinary approach to classification of rare AAT variants and their clinical impact on individuals with rare AATD genotypes.
Collapse
Affiliation(s)
- Stefania Ottaviani
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Giulia Bartoli
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Tomás P Carroll
- α-1 Foundation Ireland, Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Fabrizio Gangemi
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alice M Balderacchi
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Valentina Barzon
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, University of Pavia, Pavia, Italy
| | - Alessandra Corino
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Davide Piloni
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Noel G McElvaney
- α-1 Foundation Ireland, Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Angelo G Corsico
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, University of Pavia, Pavia, Italy
- European Reference Network on Rare Respiratory Diseases (ERN-LUNG); and
| | - James A Irving
- University College London Respiratory, Rayne Institute and the Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Annamaria Fra
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ilaria Ferrarotti
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Unità Operativa Complessa Pneumologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, Pulmonology Unit, University of Pavia, Pavia, Italy
- European Reference Network on Rare Respiratory Diseases (ERN-LUNG); and
| |
Collapse
|
7
|
Roboti P, O'Keefe S, Duah KB, Shi WQ, High S. Ipomoeassin-F disrupts multiple aspects of secretory protein biogenesis. Sci Rep 2021; 11:11562. [PMID: 34079010 PMCID: PMC8173012 DOI: 10.1038/s41598-021-91107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
The Sec61 complex translocates nascent polypeptides into and across the membrane of the endoplasmic reticulum (ER), providing access to the secretory pathway. In this study, we show that Ipomoeassin-F (Ipom-F), a selective inhibitor of protein entry into the ER lumen, blocks the in vitro translocation of certain secretory proteins and ER lumenal folding factors whilst barely affecting others such as albumin. The effects of Ipom-F on protein secretion from HepG2 cells are twofold: reduced ER translocation combined, in some cases, with defective ER lumenal folding. This latter issue is most likely a consequence of Ipom-F preventing the cell from replenishing its ER lumenal chaperones. Ipom-F treatment results in two cellular stress responses: firstly, an upregulation of stress-inducible cytosolic chaperones, Hsp70 and Hsp90; secondly, an atypical unfolded protein response (UPR) linked to the Ipom-F-mediated perturbation of ER function. Hence, although levels of spliced XBP1 and CHOP mRNA and ATF4 protein increase with Ipom-F, the accompanying increase in the levels of ER lumenal BiP and GRP94 seen with tunicamycin are not observed. In short, although Ipom-F reduces the biosynthetic load of newly synthesised secretory proteins entering the ER lumen, its effects on the UPR preclude the cell restoring ER homeostasis.
Collapse
Affiliation(s)
- Peristera Roboti
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| | - Sarah O'Keefe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Kwabena B Duah
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Stephen High
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
8
|
Ronzoni R, Ferrarotti I, D’Acunto E, Balderacchi AM, Ottaviani S, Lomas DA, Irving JA, Miranda E, Fra A. The Importance of N186 in the Alpha-1-Antitrypsin Shutter Region Is Revealed by the Novel Bologna Deficiency Variant. Int J Mol Sci 2021; 22:5668. [PMID: 34073489 PMCID: PMC8198886 DOI: 10.3390/ijms22115668] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Alpha-1-antitrypsin (AAT) deficiency causes pulmonary disease due to decreased levels of circulating AAT and consequently unbalanced protease activity in the lungs. Deposition of specific AAT variants, such as the common Z AAT, within hepatocytes may also result in liver disease. These deposits are comprised of ordered polymers of AAT formed by an inter-molecular domain swap. The discovery and characterization of rare variants of AAT and other serpins have historically played a crucial role in the dissection of the structural mechanisms leading to AAT polymer formation. Here, we report a severely deficient shutter region variant, Bologna AAT (N186Y), which was identified in five unrelated subjects with different geographical origins. We characterized the new variant by expression in cellular models in comparison with known polymerogenic AAT variants. Bologna AAT showed secretion deficiency and intracellular accumulation as detergent-insoluble polymers. Extracellular polymers were detected in both the culture media of cells expressing Bologna AAT and in the plasma of a patient homozygous for this variant. Structural modelling revealed that the mutation disrupts the hydrogen bonding network in the AAT shutter region. These data support a crucial coordinating role for asparagine 186 and the importance of this network in promoting formation of the native structure.
Collapse
Affiliation(s)
- Riccardo Ronzoni
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London WC1E 6JF, UK; (D.A.L.); (J.A.I.)
| | - Ilaria Ferrarotti
- Pneumology Unit, Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy; (I.F.); (A.M.B.); (S.O.)
| | - Emanuela D’Acunto
- Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, 00185 Rome, Italy; (E.D.); (E.M.)
| | - Alice M. Balderacchi
- Pneumology Unit, Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy; (I.F.); (A.M.B.); (S.O.)
| | - Stefania Ottaviani
- Pneumology Unit, Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy; (I.F.); (A.M.B.); (S.O.)
| | - David A. Lomas
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London WC1E 6JF, UK; (D.A.L.); (J.A.I.)
| | - James A. Irving
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London WC1E 6JF, UK; (D.A.L.); (J.A.I.)
| | - Elena Miranda
- Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, 00185 Rome, Italy; (E.D.); (E.M.)
- Italian Pasteur Institute—Cenci Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
9
|
Karatas E, Raymond AA, Leon C, Dupuy JW, Di-Tommaso S, Senant N, Collardeau-Frachon S, Ruiz M, Lachaux A, Saltel F, Bouchecareilh M. Hepatocyte proteomes reveal the role of protein disulfide isomerase 4 in alpha 1-antitrypsin deficiency. JHEP Rep 2021; 3:100297. [PMID: 34151245 PMCID: PMC8192868 DOI: 10.1016/j.jhepr.2021.100297] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/25/2022] Open
Abstract
Background & Aims A single point mutation in the Z-variant of alpha 1-antitrypsin (Z-AAT) alone can lead to both a protein folding and trafficking defect, preventing its exit from the endoplasmic reticulum (ER), and the formation of aggregates that are retained as inclusions within the ER of hepatocytes. These defects result in a systemic AAT deficiency (AATD) that causes lung disease, whereas the ER-retained aggregates can induce severe liver injury in patients with ZZ-AATD. Unfortunately, therapeutic approaches are still limited and liver transplantation represents the only curative treatment option. To overcome this limitation, a better understanding of the molecular basis of ER aggregate formation could provide new strategies for therapeutic intervention. Methods Our functional and omics approaches here based on human hepatocytes from patients with ZZ-AATD have enabled the identification and characterisation of the role of the protein disulfide isomerase (PDI) A4/ERP72 in features of AATD-mediated liver disease. Results We report that 4 members of the PDI family (PDIA4, PDIA3, P4HB, and TXNDC5) are specifically upregulated in ZZ-AATD liver samples from adult patients. Furthermore, we show that only PDIA4 knockdown or alteration of its activity by cysteamine treatment can promote Z-AAT secretion and lead to a marked decrease in Z aggregates. Finally, detailed analysis of the Z-AAT interactome shows that PDIA4 silencing provides a more conducive environment for folding of the Z mutant, accompanied by reduction of Z-AAT-mediated oxidative stress, a feature of AATD-mediated liver disease. Conclusions PDIA4 is involved in AATD-mediated liver disease and thus represents a therapeutic target for inhibition by drugs such as cysteamine. PDI inhibition therefore represents a potential therapeutic approach for treatment of AATD. Lay summary Protein disulfide isomerase (PDI) family members, and particularly PDIA4, are upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. PDI inhibition upon cysteamine treatment leads to improvements in features of AATD and hence represents a therapeutic approach for treatment of AATD-mediated liver disease. PDIA4 is upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. Knockdown of PDIA4 by siRNA or inhibition upon cysteamine treatment leads to improvements in features of AATD. RNA interference against PDIA4 or cysteamine represent approaches for treatment of AATD-mediated liver disease.
Collapse
Key Words
- AAT, alpha 1-antitrypsin
- AATD, alpha 1-antitrypsin deficiency
- Alpha 1-antitrypsin deficiency
- CF, cystic fibrosis
- CFTR, cystic fibrosis transmembrane conductance regulator
- Cysteamine
- ER, endoplasmic reticulum
- FFPE, formalin-fixed paraffin-embedded
- FKBP10, FK506-binding protein (FKBP) isoform 10
- HCC, hepatocellular carcinoma
- IHC, immunohistochemistry
- IP, immunoprecipitation
- Liver damage
- NHK, null Hong Kong variant of AAT
- P4HB, prolyl 4-hydroxylase subunit beta/PDIA1
- PDI, protein disulfide isomerase
- PDIA3, protein disulfide isomerase family A member 3/ERP57
- PDIA4
- PDIA4, protein disulfide isomerase family A member 4/ERP70/ERP72
- PDIi, PDI inhibitors
- Protein disulfide isomerase
- ROS, reactive oxygen species
- SURF4, proteins Surfeit 4
- Scr, scramble
- TRX, thioredoxin
- TXNDC5, thioredoxin domain containing 5/PDIA15
- Treatment
- WT, wild-type
- Z-AAT, alpha 1-antitrypsin Z variant
- ZZ, homozygosis for the Z mutant allele
- siRNA, small RNA interference
- ΔF508-CFTR, most common mutation of CFTR, which deletes phenylalanine508
Collapse
Affiliation(s)
- Esra Karatas
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | - Anne-Aurélie Raymond
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Céline Leon
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | | | - Sylvaine Di-Tommaso
- Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Nathalie Senant
- Plateforme d'histopathologie, TBM-Core US 005, Bordeaux, France
| | - Sophie Collardeau-Frachon
- Department of Pathology, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Mathias Ruiz
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Alain Lachaux
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Saltel
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | | |
Collapse
|
10
|
Ronzoni R, Heyer‐Chauhan N, Fra A, Pearce AC, Rüdiger M, Miranda E, Irving JA, Lomas DA. The molecular species responsible for α 1 -antitrypsin deficiency are suppressed by a small molecule chaperone. FEBS J 2021; 288:2222-2237. [PMID: 33058391 PMCID: PMC8436759 DOI: 10.1111/febs.15597] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/28/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
The formation of ordered Z (Glu342Lys) α1 -antitrypsin polymers in hepatocytes is central to liver disease in α1 -antitrypsin deficiency. In vitro experiments have identified an intermediate conformational state (M*) that precedes polymer formation, but this has yet to be identified in vivo. Moreover, the mechanism of polymer formation and their fate in cells have been incompletely characterised. We have used cell models of disease in conjunction with conformation-selective monoclonal antibodies and a small molecule inhibitor of polymerisation to define the dynamics of polymer formation, accumulation and secretion. Pulse-chase experiments demonstrate that Z α1 -antitrypsin accumulates as short-chain polymers that partition with soluble cellular components and are partially secreted by cells. These precede the formation of larger, insoluble polymers with a longer half-life (10.9 ± 1.7 h and 20.9 ± 7.4 h for soluble and insoluble polymers, respectively). The M* intermediate (or a by-product thereof) was identified in the cells by a conformation-specific monoclonal antibody. This was completely abrogated by treatment with the small molecule, which also blocked the formation of intracellular polymers. These data allow us to conclude that the M* conformation is central to polymerisation of Z α1 -antitrypsin in vivo; preventing its accumulation represents a tractable approach for pharmacological treatment of this condition; polymers are partially secreted; and polymers exist as two distinct populations in cells whose different dynamics have likely consequences for the aetiology of the disease.
Collapse
Affiliation(s)
| | | | - Annamaria Fra
- Department of Molecular and Translational MedicineUniversity of BresciaItaly
| | | | | | - Elena Miranda
- Department of Biology and Biotechnologies‘Charles Darwin’ and Pasteur Institute – Cenci‐Bolognetti FoundationSapienza University of RomeItaly
| | - James A. Irving
- UCL RespiratoryDivision of MedicineUniversity College LondonUK
| | - David A. Lomas
- UCL RespiratoryDivision of MedicineUniversity College LondonUK
| |
Collapse
|
11
|
Hamada M, Bhakta V, Andres SN, Sheffield WP. Stepwise Reversion of Multiply Mutated Recombinant Antitrypsin Reveals a Selective Inhibitor of Coagulation Factor XIa as Active as the M358R Variant. Front Cardiovasc Med 2021; 8:647405. [PMID: 33816577 PMCID: PMC8017132 DOI: 10.3389/fcvm.2021.647405] [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: 12/29/2020] [Accepted: 02/16/2021] [Indexed: 11/13/2022] Open
Abstract
Alpha-1 antitrypsin (AAT, also known as alpha-1 proteinase inhibitor or SERPINA1) is the most abundant member of the serpin superfamily found in human plasma. The naturally occurring variant AAT M358R, altered at the P1 position of the critical reactive center loop (RCL), is re-directed away from inhibition of AAT's chief natural target, neutrophil elastase, and toward accelerated inhibition of thrombin (FIIa), kallikrein (Kal), and other proteases such as factor XIa (FXIa). FXIa is an emerging target for the development of antithrombotic agents, since patients with FXI deficiency are protected from thromboembolic disease and do not exhibit a strong bleeding tendency. Previously, we used phage display, bacterial lysate screening, and combinatorial mutagenesis to identify AAT-RC, an engineered AAT M358R with additional changes between RCL positions P7-P3', CLEVEPR-STE [with changes bolded and the P1-P1' (R358-S359) reactive center shown as R-S]. AAT-RC was 279- and 16-fold more selective for FXIa/IIa or FXIa/Kal than AAT M358R; the increased selectivity came at a cost of a 2.3-fold decrease in the rate of FXIa inhibition and a 3.3-fold increase in the stoichiometry of inhibition (SI). Here, we asked which alterations in AAT-RC were most important for the observed increases in selectivity for FXIa inhibition. We back-mutated AAT-RC to AAT-RC-1 (P7-P3' FLEVEPRSTE), AAT-RC-2 (P7-P3' FLEAEPRSTE), and AAT RC-3 (P7-P3' FLEAIPR-STE). Proteins were expressed as cleavable, hexahistidine-tagged glutathione sulfotransferase fusion proteins in E. coli and purified by proteolytic elution from glutathione agarose, with polishing on nickel chelate agarose. Selectivity for FXIa over Kal of AAT-RC-1, −2, and −3 was 14, 21, and 2.3, respectively. AAT-RC-2 inhibited FXIa 31% more rapidly than AAT M358R, with the same SI, and enhanced selectivity for FXIa over Kal, FXa, FXIIa, activated protein C, and FIIa of 25-, 130-, 420-, 440-, and 470-fold, respectively. Structural modeling of the AAT-RC-2/FXIa encounter complex suggested that both E (Glu) substitutions at P3 and P3' may promote FXIa binding via hydrogen bonding to K192 in FXIa. AAT-RC-2 is the most selective and active AAT variant reported to date for FXIa inhibition and will be tested in animal models of thrombosis and bleeding.
Collapse
Affiliation(s)
- Mostafa Hamada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Varsha Bhakta
- Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
| | - Sara N Andres
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - William P Sheffield
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
| |
Collapse
|
12
|
Oxidative Stress and Endoplasmic Reticulum Stress in Rare Respiratory Diseases. J Clin Med 2021; 10:jcm10061268. [PMID: 33803835 PMCID: PMC8003245 DOI: 10.3390/jcm10061268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies have shown that some rare respiratory diseases, such as alpha-1 antitrypsin deficiency (AATD), idiopathic pulmonary fibrosis (IPF), cystic fibrosis (CF), and primary ciliary dyskinesia (PCD) present oxidative stress (OS) and endoplasmic reticulum (ER) stress. Their involvement in these pathologies and the use of antioxidants as therapeutic agents to minimize the effects of OS are discussed in this review.
Collapse
|
13
|
Gansemer ER, McCommis KS, Martino M, King-McAlpin AQ, Potthoff MJ, Finck BN, Taylor EB, Rutkowski DT. NADPH and Glutathione Redox Link TCA Cycle Activity to Endoplasmic Reticulum Homeostasis. iScience 2020; 23:101116. [PMID: 32417402 PMCID: PMC7254477 DOI: 10.1016/j.isci.2020.101116] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/25/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023] Open
Abstract
Many metabolic diseases disrupt endoplasmic reticulum (ER) homeostasis, but little is known about how metabolic activity is communicated to the ER. Here, we show in hepatocytes and other metabolically active cells that decreasing the availability of substrate for the tricarboxylic acid (TCA) cycle diminished NADPH production, elevated glutathione oxidation, led to altered oxidative maturation of ER client proteins, and attenuated ER stress. This attenuation was prevented when glutathione oxidation was disfavored. ER stress was also alleviated by inhibiting either TCA-dependent NADPH production or Glutathione Reductase. Conversely, stimulating TCA activity increased NADPH production, glutathione reduction, and ER stress. Validating these findings, deletion of the Mitochondrial Pyruvate Carrier-which is known to decrease TCA cycle activity and protect the liver from steatohepatitis-also diminished NADPH, elevated glutathione oxidation, and alleviated ER stress. Together, our results demonstrate a novel pathway by which mitochondrial metabolic activity is communicated to the ER through the relay of redox metabolites.
Collapse
Affiliation(s)
- Erica R Gansemer
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kyle S McCommis
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine in Saint Louis, St. Louis, MO 63110, USA
| | - Michael Martino
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine in Saint Louis, St. Louis, MO 63110, USA
| | - Abdul Qaadir King-McAlpin
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Obesity Research Initiative, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Brian N Finck
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine in Saint Louis, St. Louis, MO 63110, USA
| | - Eric B Taylor
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Obesity Research Initiative, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - D Thomas Rutkowski
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
14
|
Adams BM, Ke H, Gierasch LM, Gershenson A, Hebert DN. Proper secretion of the serpin antithrombin relies strictly on thiol-dependent quality control. J Biol Chem 2019; 294:18992-19011. [PMID: 31662433 DOI: 10.1074/jbc.ra119.010450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/24/2019] [Indexed: 01/04/2023] Open
Abstract
The protein quality control machinery of the endoplasmic reticulum (ERQC) ensures that client proteins are properly folded. ERQC substrates may be recognized as nonnative by the presence of exposed hydrophobic surfaces, free thiols, or processed N-glycans. How these features dictate which ERQC pathways engage a given substrate is poorly understood. Here, using metabolic labeling, immunoprecipitations, various biochemical assays, and the human serpin antithrombin III (ATIII) as a model, we explored the role of ERQC systems in mammalian cells. Although ATIII has N-glycans and a hydrophobic core, we found that its quality control depended solely on free thiol content. Mutagenesis of all six Cys residues in ATIII to Ala resulted in its efficient secretion even though the product was not natively folded. ATIII variants with free thiols were retained in the endoplasmic reticulum but not degraded. These results provide insight into the hierarchy of ERQC systems and reveal a fundamental vulnerability of ERQC in a case of reliance on the thiol-dependent quality control pathway.
Collapse
Affiliation(s)
- Benjamin M Adams
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Haiping Ke
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lila M Gierasch
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003.,Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| | - Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Daniel N Hebert
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003 .,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
15
|
Recent Developments in mRNA-Based Protein Supplementation Therapy to Target Lung Diseases. Mol Ther 2019; 27:803-823. [PMID: 30905577 DOI: 10.1016/j.ymthe.2019.02.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022] Open
Abstract
Protein supplementation therapy using in vitro-transcribed (IVT) mRNA for genetic diseases contains huge potential as a new class of therapy. From the early ages of synthetic mRNA discovery, a great number of studies showed the versatile use of IVT mRNA as a novel approach to supplement faulty or absent protein and also as a vaccine. Many modifications have been made to produce high expressions of mRNA causing less immunogenicity and more stability. Recent advancements in the in vivo lung delivery of mRNA complexed with various carriers encouraged the whole mRNA community to tackle various genetic lung diseases. This review gives a comprehensive overview of cells associated with various lung diseases and recent advancements in mRNA-based protein replacement therapy. This review also covers a brief summary of developments in mRNA modifications and nanocarriers toward clinical translation.
Collapse
|
16
|
Laffranchi M, Berardelli R, Ronzoni R, Lomas DA, Fra A. Heteropolymerization of α-1-antitrypsin mutants in cell models mimicking heterozygosity. Hum Mol Genet 2019. [PMID: 29538751 DOI: 10.1093/hmg/ddy090] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The most common genotype associated with severe α-1-antitrypsin deficiency (AATD) is the Z homozygote. The Z variant (Glu342Lys) of α-1-antitrypsin (AAT) undergoes a conformational change and is retained within the endoplasmic reticulum (ER) of hepatocytes leading to the formation of ordered polymeric chains and inclusion bodies. Accumulation of mutated protein predisposes to cirrhosis whilst plasma AAT deficiency leads to emphysema. Increased risk of liver and lung disease has also been reported in heterozygous subjects who carry Z in association with the milder S allele (Glu264Val) or even with wild-type M. However, it is unknown whether Z AAT can co-polymerize with other AAT variants in vivo. We co-expressed two AAT variants, each modified by a different tag, in cell models that replicate AAT deficiency. We used pull-down assays to investigate interactions between co-expressed variants and showed that Z AAT forms heteropolymers with S and with the rare Mmalton (Phe52del) and Mwurzburg (Pro369Ser) mutants, and to a lesser extent with the wild-type protein. Heteropolymers were recognized by the 2C1 mAb that binds to Z polymers in vivo. There was increased intracellular accumulation of AAT variants when co-expressed with Z AAT, suggesting a dominant negative effect of the Z allele. The molecular interactions between S and Z AAT were confirmed by confocal microscopy showing their colocalization within dilated ER cisternae and by positivity in Proximity Ligation Assays. These results provide the first evidence of intracellular co-polymerization of AAT mutants and contribute to understanding the risk of liver disease in SZ and MZ heterozygotes.
Collapse
Affiliation(s)
- Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Romina Berardelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Riccardo Ronzoni
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.,UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - David A Lomas
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
17
|
Laffranchi M, Elliston ELK, Gangemi F, Berardelli R, Lomas DA, Irving JA, Fra A. Characterisation of a type II functionally-deficient variant of alpha-1-antitrypsin discovered in the general population. PLoS One 2019; 14:e0206955. [PMID: 30633749 PMCID: PMC6329500 DOI: 10.1371/journal.pone.0206955] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/02/2019] [Indexed: 11/23/2022] Open
Abstract
Lung disease in alpha-1-antitrypsin deficiency (AATD) results from dysregulated proteolytic activity, mainly by neutrophil elastase (HNE), in the lung parenchyma. This is the result of a substantial reduction of circulating alpha-1-antitrypsin (AAT) and the presence in the plasma of inactive polymers of AAT. Moreover, some AAT mutants have reduced intrinsic activity toward HNE, as demonstrated for the common Z mutant, as well as for other rarer variants. Here we report the identification and characterisation of the novel AAT reactive centre loop variant Gly349Arg (p.G373R) present in the ExAC database. This AAT variant is secreted at normal levels in cellular models of AATD but shows a severe reduction in anti-HNE activity. Biochemical and molecular dynamics studies suggest it exhibits unfavourable RCL presentation to cognate proteases and compromised insertion of the RCL into β-sheet A. Identification of a fully dysfunctional AAT mutant that does not show a secretory defect underlines the importance of accurate genotyping of patients with pulmonary AATD manifestations regardless of the presence of normal levels of AAT in the circulation. This subtype of disease is reminiscent of dysfunctional phenotypes in anti-thrombin and C1-inibitor deficiencies so, accordingly, we classify this variant as the first pure functionally-deficient (type II) AATD mutant.
Collapse
Affiliation(s)
- Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emma L. K. Elliston
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Fabrizio Gangemi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Romina Berardelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - David A. Lomas
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - James A. Irving
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
18
|
Tao YX, Conn PM. Pharmacoperones as Novel Therapeutics for Diverse Protein Conformational Diseases. Physiol Rev 2018; 98:697-725. [PMID: 29442594 DOI: 10.1152/physrev.00029.2016] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
After synthesis, proteins are folded into their native conformations aided by molecular chaperones. Dysfunction in folding caused by genetic mutations in numerous genes causes protein conformational diseases. Membrane proteins are more prone to misfolding due to their more intricate folding than soluble proteins. Misfolded proteins are detected by the cellular quality control systems, especially in the endoplasmic reticulum, and proteins may be retained there for eventual degradation by the ubiquitin-proteasome system or through autophagy. Some misfolded proteins aggregate, leading to pathologies in numerous neurological diseases. In vitro, modulating mutant protein folding by altering molecular chaperone expression can ameliorate some misfolding. Some small molecules known as chemical chaperones also correct mutant protein misfolding in vitro and in vivo. However, due to their lack of specificity, their potential as therapeutics is limited. Another class of compounds, known as pharmacological chaperones (pharmacoperones), binds with high specificity to misfolded proteins, either as enzyme substrates or receptor ligands, leading to decreased folding energy barriers and correction of the misfolding. Because many of the misfolded proteins are misrouted but do not have defects in function per se, pharmacoperones have promising potential in advancing to the clinic as therapeutics, since correcting routing may ameliorate the underlying mechanism of disease. This review will comprehensively summarize this exciting area of research, surveying the literature from in vitro studies in cell lines to transgenic animal models and clinical trials in several protein misfolding diseases.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, Alabama ; and Departments of Internal Medicine and Cell Biology, Texas Tech University Health Science Center , Lubbock, Texas
| | - P Michael Conn
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, Alabama ; and Departments of Internal Medicine and Cell Biology, Texas Tech University Health Science Center , Lubbock, Texas
| |
Collapse
|
19
|
Serebryany E, Yu S, Trauger SA, Budnik B, Shakhnovich EI. Dynamic disulfide exchange in a crystallin protein in the human eye lens promotes cataract-associated aggregation. J Biol Chem 2018; 293:17997-18009. [PMID: 30242128 DOI: 10.1074/jbc.ra118.004551] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/14/2018] [Indexed: 12/31/2022] Open
Abstract
Increased light scattering in the eye lens due to aggregation of the long-lived lens proteins, crystallins, is the cause of cataract disease. Several mutations in the gene encoding human γD-crystallin (HγD) cause misfolding and aggregation. Cataract-associated substitutions at Trp42 cause the protein to aggregate in vitro from a partially unfolded intermediate locked by an internal disulfide bridge, and proteomic evidence suggests a similar aggregation precursor is involved in age-onset cataract. Surprisingly, WT HγD can promote aggregation of the W42Q variant while itself remaining soluble. Here, a search for a biochemical mechanism for this interaction has revealed a previously unknown oxidoreductase activity in HγD. Using in vitro oxidation, mutational analysis, cysteine labeling, and MS, we have assigned this activity to a redox-active internal disulfide bond that is dynamically exchanged among HγD molecules. The W42Q variant acts as a disulfide sink, reducing oxidized WT and forming a distinct internal disulfide that kinetically traps the aggregation-prone intermediate. Our findings suggest a redox "hot potato" competition among WT and mutant or modified polypeptides wherein variants with the lowest kinetic stability are trapped in aggregation-prone intermediate states upon accepting disulfides from more stable variants. Such reactions may occur in other long-lived proteins that function in oxidizing environments. In these cases, aggregation may be forestalled by inhibiting disulfide flow toward mutant or damaged polypeptides.
Collapse
Affiliation(s)
- Eugene Serebryany
- From the Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Shuhuai Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu, China
| | | | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts 02138
| | - Eugene I Shakhnovich
- From the Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138.
| |
Collapse
|
20
|
Lamriben L, Oster ME, Tamura T, Tian W, Yang Z, Clausen H, Hebert DN. EDEM1's mannosidase-like domain binds ERAD client proteins in a redox-sensitive manner and possesses catalytic activity. J Biol Chem 2018; 293:13932-13945. [PMID: 30021839 DOI: 10.1074/jbc.ra118.004183] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/26/2018] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum (ER) degradation-enhancing α-mannosidase-like 1 protein (EDEM1) is a protein quality control factor that was initially proposed to recognize N-linked glycans on misfolded proteins through its mannosidase-like domain (MLD). However, recent studies have demonstrated that EDEM1 binds to some misfolded proteins in a glycan-independent manner, suggesting a more complex binding landscape for EDEM1. In this study, we have identified a thiol-dependent substrate interaction between EDEM1 and the α1-antitrypsin ER-associated protein degradation (ERAD) clients Z and NHK, specifically through the single Cys residue on Z/NHK (Cys256), required for binding under stringent detergent conditions. In addition to the thiol-dependent interaction, the presence of weaker protein-protein interactions was confirmed, suggestive of bipartite client-binding properties. About four reactive thiols on EDEM1 were identified and were not directly responsible for the observed redox-sensitive binding by EDEM1. Moreover, a protein construct comprising the EDEM1 MLD had thiol-dependent binding properties along with its active glycan-trimming activities. Lastly, we identified an additional intrinsically disordered region (IDR) located at the C terminus of EDEM1 in addition to its previously identified N-terminal IDR. We also determined that both IDRs are required for binding to the ERAD component ERdj5 as an interaction with ERdj5 was not observed with the MLD alone. Together, our findings indicate that EDEM1 employs different binding modalities to interact with ERAD clients and ER quality control (ERQC) machinery partners and that some of these properties are shared with its homologues EDEM2 and EDEM3.
Collapse
Affiliation(s)
- Lydia Lamriben
- From the Department of Biochemistry and Molecular Biology and.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Michela E Oster
- From the Department of Biochemistry and Molecular Biology and
| | - Taku Tamura
- From the Department of Biochemistry and Molecular Biology and
| | - Weihua Tian
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Daniel N Hebert
- From the Department of Biochemistry and Molecular Biology and .,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 and
| |
Collapse
|
21
|
Torres-Durán M, Lopez-Campos JL, Barrecheguren M, Miravitlles M, Martinez-Delgado B, Castillo S, Escribano A, Baloira A, Navarro-Garcia MM, Pellicer D, Bañuls L, Magallón M, Casas F, Dasí F. Alpha-1 antitrypsin deficiency: outstanding questions and future directions. Orphanet J Rare Dis 2018; 13:114. [PMID: 29996870 PMCID: PMC6042212 DOI: 10.1186/s13023-018-0856-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/26/2018] [Indexed: 12/14/2022] Open
Abstract
Background Alpha-1 antitrypsin deficiency (AATD) is a rare hereditary condition that leads to decreased circulating alpha-1 antitrypsin (AAT) levels, significantly increasing the risk of serious lung and/or liver disease in children and adults, in which some aspects remain unresolved. Methods In this review, we summarise and update current knowledge on alpha-1 antitrypsin deficiency in order to identify and discuss areas of controversy and formulate questions that need further research. Results 1) AATD is a highly underdiagnosed condition. Over 120,000 European individuals are estimated to have severe AATD and more than 90% of them are underdiagnosed. Conclusions 2) Several clinical and etiological aspects of the disease are yet to be resolved. New strategies for early detection and biomarkers for patient outcome prediction are needed to reduce morbidity and mortality in these patients; 3) Augmentation therapy is the only specific approved therapy that has shown clinical efficacy in delaying the progression of emphysema. Regrettably, some countries reject registration and reimbursement for this treatment because of the lack of larger randomised, placebo-controlled trials. 4) Alternative strategies are currently being investigated, including the use of gene therapy or induced pluripotent stem cells, and non-augmentation strategies to prevent AAT polymerisation inside hepatocytes.
Collapse
Affiliation(s)
- María Torres-Durán
- Pulmonary Department, Hospital Álvaro Cunqueiro EOXI, Vigo, Spain.,NeumoVigo I+i Research Group, IIS Galicia Sur, Vigo, Spain
| | - José Luis Lopez-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Sevilla, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Miriam Barrecheguren
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Pneumology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Marc Miravitlles
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Pneumology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Beatriz Martinez-Delgado
- Molecular Genetics Unit, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Silvia Castillo
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - Amparo Escribano
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Paediatrics, Obstetrics and Gynaecology, University of Valencia, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - Adolfo Baloira
- Pneumology Department, Complejo Hospitalario Universitario de Pontevedra, Pontevedra, Spain
| | - María Mercedes Navarro-Garcia
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - Daniel Pellicer
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - Lucía Bañuls
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - María Magallón
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain.,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain
| | - Francisco Casas
- Pneumology Department, Hospital Universitario San Cecilio, Granada, Spain
| | - Francisco Dasí
- Fundación Investigación Hospital Clínico Valencia, Instituto de Investigación Sanitaria INCLIVA, c/Menéndez y Pelayo, 4, 46010, Valencia, Spain. .,School of Medicine, Department of Physiology, Research group on Rare Respiratory Diseases (ERR), University of Valencia, Valencia, Spain.
| |
Collapse
|
22
|
Giacopuzzi E, Laffranchi M, Berardelli R, Ravasio V, Ferrarotti I, Gooptu B, Borsani G, Fra A. Real-world clinical applicability of pathogenicity predictors assessed on SERPINA1
mutations in alpha-1-antitrypsin deficiency. Hum Mutat 2018; 39:1203-1213. [DOI: 10.1002/humu.23562] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/20/2018] [Accepted: 06/05/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Edoardo Giacopuzzi
- Division of Biology and Genetics; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Mattia Laffranchi
- Experimental Oncology and Immunology; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Romina Berardelli
- Experimental Oncology and Immunology; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Viola Ravasio
- Division of Biology and Genetics; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Ilaria Ferrarotti
- Centre for Diagnosis of Inherited Alpha-1 Antitrypsin Deficiency; Department of Internal Medicine and Therapeutics; University of Pavia; Pavia Italy
| | - Bibek Gooptu
- Leicester Institute of Structural and Chemical Biology / NIHR Leicester BRC - Respiratory; University of Leicester; Leicester UK
| | - Giuseppe Borsani
- Division of Biology and Genetics; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Annamaria Fra
- Experimental Oncology and Immunology; Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| |
Collapse
|
23
|
Janciauskiene S, Wrenger S, Immenschuh S, Olejnicka B, Greulich T, Welte T, Chorostowska-Wynimko J. The Multifaceted Effects of Alpha1-Antitrypsin on Neutrophil Functions. Front Pharmacol 2018; 9:341. [PMID: 29719508 PMCID: PMC5914301 DOI: 10.3389/fphar.2018.00341] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/23/2018] [Indexed: 01/02/2023] Open
Abstract
Neutrophils are the predominant immune cells in human blood possessing heterogeneity, plasticity and functional diversity. The activation and recruitment of neutrophils into inflamed tissue in response to stimuli are tightly regulated processes. Alpha1-Antitrypsin (AAT), an acute phase protein, is one of the potent regulators of neutrophil activation via both -protease inhibitory and non-inhibitory functions. This review summarizes our current understanding of the effects of AAT on neutrophils, illustrating the interplay between AAT and the key effector functions of neutrophils.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Sabine Wrenger
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Beata Olejnicka
- Department of Medicine, Trelleborg Hospital, Trelleborg, Sweden
| | - Timm Greulich
- Department of Medicine, Pulmonary and Critical Care Medicine, Member of the German Center for Lung Research (DZL), University Hospital of Giessen and Marburg, University of Marburg, Marburg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|
24
|
Caccia S, Suffritti C, Carzaniga T, Berardelli R, Berra S, Martorana V, Fra A, Drouet C, Cicardi M. Intermittent C1-Inhibitor Deficiency Associated with Recessive Inheritance: Functional and Structural Insight. Sci Rep 2018; 8:977. [PMID: 29343682 PMCID: PMC5772639 DOI: 10.1038/s41598-017-16667-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022] Open
Abstract
C1-inhibitor is a serine protease inhibitor (serpin) controlling complement and contact system activation. Gene mutations result in reduced C1-inhibitor functional plasma level causing hereditary angioedema, a life-threatening disorder. Despite a stable defect, the clinical expression of hereditary angioedema is unpredictable, and the molecular mechanism underlying this variability remains undisclosed. Here we report functional and structural studies on the Arg378Cys C1-inhibitor mutant found in a patient presenting reduced C1-inhibitor levels, episodically undergoing normalization. Expression studies resulted in a drop in mutant C1-innhibitor secretion compared to wild-type. Notwithstanding, the purified proteins had similar features. Thermal denaturation experiments showed a comparable denaturation profile, but the mutant thermal stability decays when tested in conditions reproducing intracellular crowding.Our findings suggest that once correctly folded, the Arg378Cys C1-inhibitor is secreted as an active, although quite unstable, monomer. However, it could bear a folding defect, occasionally promoting protein oligomerization and interfering with the secretion process, thus accounting for its plasma level variability. This defect is exacerbated by the nature of the mutation since the acquired cysteine leads to the formation of non-functional homodimers through inter-molecular disulphide bonding. All the proposed phenomena could be modulated by specific environmental conditions, rendering this mutant exceptionally vulnerable to mild stress.
Collapse
Affiliation(s)
- Sonia Caccia
- "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, via GB Grassi 74, 20157, Milan, Italy.
| | - Chiara Suffritti
- "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, via GB Grassi 74, 20157, Milan, Italy
| | - Thomas Carzaniga
- "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, via GB Grassi 74, 20157, Milan, Italy
| | - Romina Berardelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvia Berra
- "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, via GB Grassi 74, 20157, Milan, Italy
| | - Vincenzo Martorana
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Christian Drouet
- GREPI EA7408, Universite Grenoble Alpes, and CREAK, CHU Grenoble, Grenoble, France
| | - Marco Cicardi
- "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, via GB Grassi 74, 20157, Milan, Italy
- Luigi Sacco Hospital, Milan, Italy
| |
Collapse
|
25
|
Abstract
Our current knowledge about the cellular mechanisms underlying serpin-related disorders, the serpinopathies, is predominantly based on studies in cell culture models of disease, particularly for alpha-1 antitrypsin (AAT, SERPINA1) deficiency causing emphysema and the familial encephalopathy with neuroserpin (NS, SERPINI1) inclusion bodies (FENIB). FENIB, a neurodegenerative dementia, is caused by polymerization of NS (Miranda and Lomas, Cell Mol Life Sci 63:709-722, 2006; Roussel BD et al., Epileptic Disor 18:103-110, 2016), while AAT deficiency presents as a result of several divergent mutations in the AAT gene that cause lack of protein synthesis or complete intracellular degradation (null variants) or polymer formation (polymerogenic variants) (Lomas et al., J Hepatol 65:413-424, 2016; Greene et al., Nat Rev Dis Primers 2:16051, 2016; Ferrarotti et al. Orphanet J Rare D 9:172, 2014). Both diseases have been extensively modeled in cell culture systems by expressing mutant variants in a variety of ways. Here we describe the methodologies we follow in our cell model systems used to examine serpin disorders.
Collapse
Affiliation(s)
- Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emanuela D'Acunto
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy.
- Italian Pasteur Institute - Cenci-Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
26
|
Miranda E, Ferrarotti I, Berardelli R, Laffranchi M, Cerea M, Gangemi F, Haq I, Ottaviani S, Lomas DA, Irving JA, Fra A. The pathological Trento variant of alpha-1-antitrypsin (E75V) shows nonclassical behaviour during polymerization. FEBS J 2017; 284:2110-2126. [PMID: 28504839 PMCID: PMC5518210 DOI: 10.1111/febs.14111] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/26/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022]
Abstract
Severe alpha‐1‐antitrypsin deficiency (AATD) is most frequently associated with the alpha‐1‐antitrypsin (AAT) Z variant (E342K). ZZ homozygotes exhibit accumulation of AAT as polymers in the endoplasmic reticulum of hepatocytes. This protein deposition can lead to liver disease, with the resulting low circulating levels of AAT predisposing to early‐onset emphysema due to dysregulation of elastinolytic activity in the lungs. An increasing number of rare AAT alleles have been identified in patients with severe AATD, typically in combination with the Z allele. Here we report a new mutation (E75V) in a patient with severe plasma deficiency, which we designate Trento. In contrast to the Z mutant, Trento AAT was secreted efficiently when expressed in cellular models but showed compromised conformational stability. Polyacrylamide gel electrophoresis (PAGE) and ELISA‐based analyses of the secreted protein revealed the presence of oligomeric species with electrophoretic and immunorecognition profiles different from those of Z and S (E264V) AAT polymers, including reduced recognition by conformational monoclonal antibodies 2C1 and 4B12. This altered recognition was not due to direct effects on the epitope of the 2C1 monoclonal antibody which we localized between helices E and F. Structural analyses indicate the likely basis for polymer formation is the loss of a highly conserved stabilizing interaction between helix C and the posthelix I loop. These results highlight this region as important for maintaining native state stability and, when compromised, results in the formation of pathological polymers that are different from those produced by Z and S AAT.
Collapse
Affiliation(s)
- Elena Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Ilaria Ferrarotti
- Department of Internal Medicine and Therapeutics, Pneumology Unit, University of Pavia, Italy
| | - Romina Berardelli
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Marta Cerea
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Fabrizio Gangemi
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Imran Haq
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, UK
| | - Stefania Ottaviani
- Center for Diagnosis of Inherited Alpha 1-Antitrypsin Deficiency, Pneumology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - David A Lomas
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, UK
| | - James A Irving
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, UK
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| |
Collapse
|
27
|
Abstract
Thiol groups can undergo numerous modifications, making cysteine a unique molecular switch. Cysteine plays structural and regulatory roles as part of proteins or glutathione, contributing to maintain redox homeostasis and regulate signaling within and amongst cells. Not surprisingly therefore, cysteines are associated with many hereditary and acquired diseases. Mutations in the primary protein sequence (gain or loss of a cysteine) are most frequent in membrane and secretory proteins, correlating with the key roles of disulfide bonds. On the contrary, in the cytosol and nucleus, aberrant post-translational oxidative modifications of thiol groups, reflecting redox changes in the surrounding environment, are a more frequent cause of dysregulation of protein function. This essay highlights the regulatory functions performed by protein cysteine residues and provides a framework for understanding how mutation and/or (in)activation of this key amino acid can cause disease.
Collapse
Affiliation(s)
- Annamaria Fra
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy
| | - Edgar D Yoboue
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele UniversityMilan, Italy.,Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific InstituteMilan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele UniversityMilan, Italy.,Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific InstituteMilan, Italy
| |
Collapse
|