1
|
Graham J, Zhang Y, He L, Gonzalez-Fernandez T. CRISPR-GEM: A Novel Machine Learning Model for CRISPR Genetic Target Discovery and Evaluation. ACS Synth Biol 2024; 13:3413-3429. [PMID: 39375864 PMCID: PMC11494708 DOI: 10.1021/acssynbio.4c00473] [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: 07/09/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
CRISPR gene editing strategies are shaping cell therapies through precise and tunable control over gene expression. However, limitations in safely delivering high quantities of CRISPR machinery demand careful target gene selection to achieve reliable therapeutic effects. Informed target gene selection requires a thorough understanding of the involvement of target genes in gene regulatory networks (GRNs) and thus their impact on cell phenotype. Effective decoding of these complex networks has been achieved using machine learning models, but current techniques are limited to single cell types and focus mainly on transcription factors, limiting their applicability to CRISPR strategies. To address this, we present CRISPR-GEM, a multilayer perceptron (MLP) based synthetic GRN constructed to accurately predict the downstream effects of CRISPR gene editing. First, input and output nodes are identified as differentially expressed genes between defined experimental and target cell/tissue types, respectively. Then, MLP training learns regulatory relationships in a black-box approach allowing accurate prediction of output gene expression using only input gene expression. Finally, CRISPR-mimetic perturbations are made to each input gene individually, and the resulting model predictions are compared to those for the target group to score and assess each input gene as a CRISPR candidate. The top scoring genes provided by CRISPR-GEM therefore best modulate experimental group GRNs to motivate transcriptomic shifts toward a target group phenotype. This machine learning model is the first of its kind for predicting optimal CRISPR target genes and serves as a powerful tool for enhanced CRISPR strategies across a range of cell therapies.
Collapse
Affiliation(s)
- Joshua
P. Graham
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yu Zhang
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Lifang He
- Department
of Computer Science and Engineering, Lehigh
University, Bethlehem, Pennsylvania 18015, United States
| | | |
Collapse
|
2
|
Mazzuca C, Vitiello L, Travaglini S, Maurizi F, Finamore P, Santangelo S, Rigon A, Vadacca M, Angeletti S, Scarlata S. Immunological and homeostatic pathways of alpha -1 antitrypsin: a new therapeutic potential. Front Immunol 2024; 15:1443297. [PMID: 39224588 PMCID: PMC11366583 DOI: 10.3389/fimmu.2024.1443297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
α -1 antitrypsin (A1AT) is a 52 kDa acute-phase glycoprotein belonging to the serine protease inhibitor superfamily (SERPIN). It is primarily synthesized by hepatocytes and to a lesser extent by monocytes, macrophages, intestinal epithelial cells, and bronchial epithelial cells. A1AT is encoded by SERPINA1 locus, also known as PI locus, highly polymorphic with at least 100 allelic variants described and responsible for different A1AT serum levels and function. A1AT inhibits a variety of serine proteinases, but its main target is represented by Neutrophil Elastase (NE). However, recent attention has been directed towards its immune-regulatory and homeostatic activities. A1AT exerts immune-regulatory effects on different cell types involved in innate and adaptive immunity. Additionally, it plays a role in metal and lipid metabolism, contributing to homeostasis. An adequate comprehension of these mechanisms could support the use of A1AT augmentation therapy in many disorders characterized by a chronic immune response. The aim of this review is to provide an up-to-date understanding of the molecular mechanisms and regulatory pathways responsible for immune-regulatory and homeostatic activities of A1AT. This knowledge aims to support the use of A1AT in therapeutic applications. Furthermore, the review summarizes the current state of knowledge regarding the application of A1AT in clinical and laboratory settings human and animal models.
Collapse
Affiliation(s)
- Carmen Mazzuca
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
- Pediatric Allergology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Laura Vitiello
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele University, Rome, Italy
| | - Silvia Travaglini
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Fatima Maurizi
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Panaiotis Finamore
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Simona Santangelo
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Amelia Rigon
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Marta Vadacca
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Simone Scarlata
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| |
Collapse
|
3
|
Graham JP, Zhang Y, He L, Gonzalez-Fernandez T. CRISPR-GEM: A Novel Machine Learning Model for CRISPR Genetic Target Discovery and Evaluation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601587. [PMID: 39005295 PMCID: PMC11244939 DOI: 10.1101/2024.07.01.601587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
CRISPR gene editing strategies are shaping cell therapies through precise and tunable control over gene expression. However, achieving reliable therapeutic effects with improved safety and efficacy requires informed target gene selection. This depends on a thorough understanding of the involvement of target genes in gene regulatory networks (GRNs) that regulate cell phenotype and function. Machine learning models have been previously used for GRN reconstruction using RNA-seq data, but current techniques are limited to single cell types and focus mainly on transcription factors. This restriction overlooks many potential CRISPR target genes, such as those encoding extracellular matrix components, growth factors, and signaling molecules, thus limiting the applicability of these models for CRISPR strategies. To address these limitations, we have developed CRISPR-GEM, a multi-layer perceptron (MLP)-based synthetic GRN constructed to accurately predict the downstream effects of CRISPR gene editing. First, input and output nodes are identified as differentially expressed genes between defined experimental and target cell/tissue types respectively. Then, MLP training learns regulatory relationships in a black-box approach allowing accurate prediction of output gene expression using only input gene expression. Finally, CRISPR-mimetic perturbations are made to each input gene individually and the resulting model predictions are compared to those for the target group to score and assess each input gene as a CRISPR candidate. The top scoring genes provided by CRISPR-GEM therefore best modulate experimental group GRNs to motivate transcriptomic shifts towards a target group phenotype. This machine learning model is the first of its kind for predicting optimal CRISPR target genes and serves as a powerful tool for enhanced CRISPR strategies across a range of cell therapies.
Collapse
Affiliation(s)
- Josh P Graham
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Yu Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA, USA
| | - Lifang He
- Department of Computer Science and Engineering, Lehigh University, Bethlehem, PA, USA
| | | |
Collapse
|
4
|
Human Alpha 1 Antitrypsin Suppresses NF-κB Activity and Extends Lifespan in Adult Drosophila. Biomolecules 2022; 12:biom12101347. [PMID: 36291555 PMCID: PMC9599126 DOI: 10.3390/biom12101347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Human alpha 1 antitrypsin (hAAT) is a multifunctional protein that has been shown to have anti-inflammatory and cellular protective properties. While previous studies demonstrated the antiaging potential of hAAT, the mechanism(s) underlying the antiaging effect remain elusive. In this study, we performed a detailed analysis of transcriptomic data that indicated that NF-κB-targeted genes and NF-κB-regulated pathways were selectively inhibited by hAAT treatment. We further showed that the first detectable impact of hAAT treatment was the inhibition of the nuclear activity of NF-κB. Subsequently, hAAT treatment suppressed the mRNA levels of NF-κB-targeted genes, as well as NF-κB itself (P65 and P50), in human senescent cells. Using Drosophila models, we further examined the impact of hAAT on locomotor activity and endurance. Finally, using an adult-specific promotor, we demonstrated that overexpression of hAAT in the late stage of life significantly extended the lifespan of transgenic flies. These results extend the current understanding of the anti-inflammatory function of hAAT.
Collapse
|
5
|
Sun R, Xu Z, Zhu C, Chen T, Muñoz LE, Dai L, Zhao Y. Alpha-1 antitrypsin in autoimmune diseases: Roles and therapeutic prospects. Int Immunopharmacol 2022; 110:109001. [PMID: 35803133 DOI: 10.1016/j.intimp.2022.109001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023]
Abstract
Alpha-1 antitrypsin (A1AT) is a protease inhibitor in the serum. Its primary function is to inhibit the activity of a series of proteases, including proteinase 3, neutrophil elastase, metalloproteases, and cysteine-aspartate proteases. In addition, A1AT also has anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-viral, and anti-bacterial activities and plays essential roles in the regulation of tissue repair and lymphocyte differentiation and activation. The overactivation of the immune system characterizes the pathogenesis of autoimmune diseases. A1AT treatment shows beneficial effects on patients and animal models with autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. This review summarizes the functions and therapeutic prospects of A1AT in autoimmune diseases.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiqiang Xu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxi Zhu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lunzhi Dai
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Wilkinson DJ, Falconer AMD, Wright HL, Lin H, Yamamoto K, Cheung K, Charlton SH, Arques MDC, Janciauskiene S, Refaie R, Rankin KS, Young DA, Rowan AD. Matrix metalloproteinase-13 is fully activated by neutrophil elastase and inactivates its serpin inhibitor, alpha-1 antitrypsin: Implications for osteoarthritis. FEBS J 2022; 289:121-139. [PMID: 34270864 DOI: 10.1111/febs.16127] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 06/23/2021] [Accepted: 07/15/2021] [Indexed: 01/15/2023]
Abstract
Matrix metalloproteinase-13 (MMP-13) is a uniquely important collagenase that promotes the irreversible destruction of cartilage collagen in osteoarthritis (OA). Collagenase activation is a key control point for cartilage breakdown to occur, yet our understanding of the proteinases involved in this process is limited. Neutrophil elastase (NE) is a well-described proteoglycan-degrading enzyme which is historically associated with inflammatory arthritis, but more recent evidence suggests a potential role in OA. In this study, we investigated the effect of neutrophil elastase on OA cartilage collagen destruction and collagenase activation. Neutrophil elastase induced significant collagen destruction from human OA cartilage ex vivo, in an MMP-dependent manner. In vitro, neutrophil elastase directly and robustly activated pro-MMP-13, and N-terminal sequencing identified cleavage close to the cysteine switch at 72 MKKPR, ultimately resulting in the fully active form with the neo-N terminus of 85 YNVFP. Mole-per-mole, activation was more potent than by MMP-3, a classical collagenase activator. Elastase was detectable in human OA synovial fluid and OA synovia which displayed histologically graded evidence of synovitis. Bioinformatic analyses demonstrated that, compared with other tissues, control cartilage exhibited remarkably high transcript levels of the major elastase inhibitor, (AAT) alpha-1 antitrypsin (gene name SERPINA1), but these were reduced in OA. AAT was located predominantly in superficial cartilage zones, and staining enhanced in regions of cartilage damage. Finally, active MMP-13 specifically inactivated AAT by removal of the serine proteinase cleavage/inhibition site. Taken together, this study identifies elastase as a novel activator of pro-MMP-13 that has relevance for cartilage collagen destruction in OA patients with synovitis.
Collapse
Affiliation(s)
- David J Wilkinson
- Institute of Life Course and Medical Sciences, University of Liverpool, UK
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Adrian M D Falconer
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Helen L Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Hua Lin
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Kathleen Cheung
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Sarah H Charlton
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | | | - Sabina Janciauskiene
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Department of Respiratory Medicine, Deutsches Zentrum für Lungenforschung, Hannover Medical School, Germany
| | - Ramsay Refaie
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Kenneth S Rankin
- Translational and Clinical Research Institute, Newcastle Centre for Cancer, UK
| | - David A Young
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| | - Andrew D Rowan
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, UK
| |
Collapse
|
7
|
Lior Y, Shtriker E, Kahremany S, Lewis EC, Gruzman A. Development of anti-inflammatory peptidomimetics based on the structure of human alpha1-antitrypsin. Eur J Med Chem 2021; 228:113969. [PMID: 34763945 DOI: 10.1016/j.ejmech.2021.113969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/10/2023]
Abstract
Human α1-antitrypsin (hAAT) has two distinguishing functions: anti-protease activity and regulation of the immune system. In the present study we hypothesized that those two protein functions are mediated by different structural domains on the hAAT surface. Indeed, such biologically active immunoregulatory sites (not associated with canonical anti-protease activity) on the surface of hAAT were identified by in silico methods. Several peptides were derived from those immunoregulatory sites. Four peptides exhibited impressive biological effects in pharmacological concentration ranges. Peptidomimetic (14) was developed, based on the structure of the most druggable and active peptide. The compound exhibited a potent anti-inflammatory activity in vitro and in vivo. Such a compound could be used as a basis for developing novel anti-inflammatory drug candidates and as a research tool for better understanding hAAT functions.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Efrat Shtriker
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Shirin Kahremany
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Skin Research Institute, The Dead Sea and Arava Science Center, 86910, Masada, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Arie Gruzman
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| |
Collapse
|
8
|
Liu LP, Gholam MF, Elshikha AS, Kawakibi T, Elmoujahid N, Moussa HH, Song S, Alli AA. Transgenic Mice Overexpressing Human Alpha-1 Antitrypsin Exhibit Low Blood Pressure and Altered Epithelial Transport Mechanisms in the Inactive and Active Cycles. Front Physiol 2021; 12:710313. [PMID: 34630137 PMCID: PMC8493122 DOI: 10.3389/fphys.2021.710313] [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: 05/15/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Abstract
Human alpha-1 antitrypsin (hAAT) is a versatile protease inhibitor, but little is known about its targets in the aldosterone-sensitive distal nephron and its role in electrolyte balance and blood pressure control. We analyzed urinary electrolytes, osmolality, and blood pressure from hAAT transgenic (hAAT-Tg) mice and C57B/6 wild-type control mice maintained on either a normal salt or high salt diet. Urinary sodium, potassium, and chloride concentrations as well as urinary osmolality were lower in hAAT-Tg mice maintained on a high salt diet during both the active and inactive cycles. hAAT-Tg mice showed a lower systolic blood pressure compared to C57B6 mice when maintained on a normal salt diet but this was not observed when they were maintained on a high salt diet. Cathepsin B protein activity was less in hAAT-Tg mice compared to wild-type controls. Protein expression of the alpha subunit of the sodium epithelial channel (ENaC) alpha was also reduced in the hAAT-Tg mice. Natriuretic peptide receptor C (NPRC) protein expression in membrane fractions of the kidney cortex was reduced while circulating levels of atrial natriuretic peptide (ANP) were greater in hAAT-Tg mice compared to wild-type controls. This study characterizes the electrolyte and blood pressure phenotype of hAAT-Tg mice during the inactive and active cycles and investigates the mechanism by which ENaC activation is inhibited in part by a mechanism involving decreased cathepsin B activity and increased ANP levels in the systemic circulation.
Collapse
Affiliation(s)
- Lauren P Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mohammed F Gholam
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ahmed Samir Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tamim Kawakibi
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Nasseem Elmoujahid
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hassan H Moussa
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sihong Song
- Department of Pharmaceutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
9
|
Kaneva MK. Neutrophil elastase and its inhibitors-overlooked players in osteoarthritis. FEBS J 2021; 289:113-116. [PMID: 34580987 DOI: 10.1111/febs.16194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
Cartilage homeostasis is maintained by a delicate balance between anabolism and catabolism. In osteoarthritis, pathological biomechanics or injury triggers cartilage breakdown, nonresolving synovial inflammation, and bone changes, causing reduced joint mobility and incapacitating pain. Undoubtedly, the most important cartilage degrading collagenase during osteoarthritis, matrix metalloproteinase (MMP)-13, is activated by an unlikely player: neutrophil elastase. Although primarily associated with inflammatory arthritis, neutrophil elastase is present in the osteoarthritic joint, and through activating MMP-13, spurs a cascade of events leading not just to the aberrant destruction of the cartilage itself, but to the proteolysis of its own inhibitor, alpha-1-antitrypsin, as described in the new study by Wilkinson et al. Endowed with potent chondrogenic and cartilage-protective properties, the loss of alpha-1-antitrypsin from cartilage will have major consequences for osteoarthritis progression, and strategies to prevent its loss, or replace it, might provide an innovative treatment opportunity that should not be ignored. Comment on: https://doi.org/10.1111/febs.16127.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, UK.,Centre for Inflammation and Therapeutic Innovation (CiTI), Queen Mary University of London, UK
| |
Collapse
|
10
|
Yamamoto K, Wilkinson D, Bou-Gharios G. Targeting Dysregulation of Metalloproteinase Activity in Osteoarthritis. Calcif Tissue Int 2021; 109:277-290. [PMID: 32772139 PMCID: PMC8403128 DOI: 10.1007/s00223-020-00739-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
Metalloproteinases were first identified as collagen cleaving enzymes and are now appreciated to play important roles in a wide variety of biological processes. The aberrant activity and dysregulation of the metalloproteinase family are linked to numerous diseases including cardiovascular and pulmonary diseases, chronic wounds, cancer, fibrosis and arthritis. Osteoarthritis (OA) is the most prevalent age-related joint disorder that causes pain and disability, but there are no disease-modifying drugs available. The hallmark of OA is loss of articular cartilage and elevated activities of matrix-degrading metalloproteinases are responsible. These enzymes do not exist in isolation and their activity is tightly regulated by a number of processes, such as transcription, proteolytic activation, interaction with their inhibitors, cell surface and extracellular matrix molecules, and endocytic clearance from the extracellular milieu. Here, we describe the functions and roles of metalloproteinase family in OA pathogenesis. We highlight recent studies that have illustrated novel mechanisms regulating their extracellular activity and impairment of such regulations that lead to the development of OA. We also discuss how to stop or slow down the degenerative processes by targeting aberrant metalloproteinase activity, which may in future become therapeutic interventions for the disease.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - David Wilkinson
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
11
|
Kaneva MK, Muley MM, Krustev E, Reid AR, Souza PR, Dell'Accio F, McDougall JJ, Perretti M. Alpha-1-antitrypsin reduces inflammation and exerts chondroprotection in arthritis. FASEB J 2021; 35:e21472. [PMID: 33788977 DOI: 10.1096/fj.202001801r] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
While new treatments have been developed to control joint disease in rheumatoid arthritis, they are partially effective and do not promote structural repair of cartilage. Following an initial identification of α-1-Antitrypsin (AAT) during the resolution phase of acute inflammation, we report here the properties of this protein in the context of cartilage protection, joint inflammation, and associated pain behavior. Intra-articular and systemic administration of AAT reversed joint inflammation, nociception, and cartilage degradation in the KBxN serum and neutrophil elastase models of arthritis. Ex vivo analyses of arthritic joints revealed that AAT promoted transcription of col2a1, acan, and sox9 and downregulated mmp13 and adamts5 gene expression. In vitro studies using human chondrocytes revealed that SERPINA1 transfection and rAAT protein promoted chondrogenic differentiation through activation of PKA-dependent CREB signaling and inhibition of Wnt/β-catenin pathways. Thus, AAT is endowed with anti-inflammatory, analgesic, and chondroprotective properties that are partially inter-related. We propose that AAT could be developed for new therapeutic strategies to reduce arthritic pain and repair damaged cartilage.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK
| | - Milind M Muley
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Eugene Krustev
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Allison R Reid
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Patricia R Souza
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK.,Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Mauro Perretti
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK.,Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| |
Collapse
|
12
|
Serpins in cartilage and osteoarthritis: what do we know? Biochem Soc Trans 2021; 49:1013-1026. [PMID: 33843993 PMCID: PMC8106492 DOI: 10.1042/bst20201231] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/17/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Serpins (serine proteinase inhibitors) are an ancient superfamily of structurally similar proteins, the majority of which use an elegant suicide inhibition mechanism to target serine proteinases. Despite likely evolving from a single common ancestor, the 36 human serpins have established roles regulating diverse biological processes, such as blood coagulation, embryonic development and extracellular matrix (ECM) turnover. Genetic mutations in serpin genes underpin a host of monogenic disorders — collectively termed the ‘serpinopathies’ — but serpin dysregulation has also been shown to drive pathological mechanisms in many common diseases. Osteoarthritis is a degenerative joint disorder, characterised by the progressive destruction of articular cartilage. This breakdown of the cartilage is driven by the metalloproteinases, and it has long been established that an imbalance of metalloproteinases to their inhibitors is of critical importance. More recently, a role for serine proteinases in cartilage destruction is emerging; including the activation of latent matrix metalloproteinases and cell-surface receptors, or direct proteolysis of the ECM. Serpins likely regulate these processes, as well as having roles beyond serine proteinase inhibition. Indeed, serpins are routinely observed to be highly modulated in osteoarthritic tissues and fluids by ‘omic analysis, but despite this, they are largely ignored. Confusing nomenclature and an underappreciation for the role of serine proteinases in osteoarthritis (OA) being the likely causes. In this narrative review, serpin structure, biochemistry and nomenclature are introduced, and for the first time, their putative importance in maintaining joint tissues — as well as their dysregulation in OA — are explored.
Collapse
|
13
|
Martini F, De Mattei M, Contini C, Tognon MG. Potential Use of Alpha-1 Anti-trypsin in the Covid-19 Treatment. Front Cell Dev Biol 2020; 8:577528. [PMID: 33195215 PMCID: PMC7644540 DOI: 10.3389/fcell.2020.577528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Fernanda Martini
- Laboraotories of Cell Biology and Molecular Genetics, University of Ferrara, Ferrara, Italy
| | - Monica De Mattei
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Carlo Contini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro G. Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
14
|
Distinct anti-inflammatory properties of alpha1-antitrypsin and corticosteroids reveal unique underlying mechanisms of action. Cell Immunol 2020; 356:104177. [DOI: 10.1016/j.cellimm.2020.104177] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/31/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
|
15
|
Black RM, Wang Y, Struglics A, Lorenzo P, Tillgren V, Rydén M, Grodzinsky AJ, Önnerfjord P. Proteomic analysis reveals dexamethasone rescues matrix breakdown but not anabolic dysregulation in a cartilage injury model. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2. [PMID: 34322675 DOI: 10.1016/j.ocarto.2020.100099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objectives In this exploratory study, we used discovery proteomics to follow the release of proteins from bovine knee articular cartilage in response to mechanical injury and cytokine treatment. We also studied the effect of the glucocorticoid Dexamethasone (Dex) on these responses. Design Bovine cartilage explants were treated with either cytokines alone (10 ng/ml TNFα, 20 ng/ml IL-6, 100 ng/ml sIL-6R), a single compressive mechanical injury, cytokines and injury, or no treatment, and cultured in serum-free DMEM supplemented with 1% ITS for 22 days. All samples were incubated with or without addition of 100 nM Dex. Mass spectrometry and western blot analyses were performed on medium samples for the identification and quantification of released proteins. Results We identified 500 unique proteins present in all three biological replicates. Many proteins involved in the catabolic response of cartilage degradation had increased release after inflammatory stress. Dex rescued many of these catabolic effects. The release of some proteins involved in anabolic and chondroprotective processes was inconsistent, indicating differential effects on processes that may protect cartilage from injury. Dex restored only a small fraction of these to the control state, while others had their effects exacerbated by Dex exposure. Conclusions We identified proteins that were released upon cytokine treatment which could be potential biomarkers of the inflammatory contribution to cartilage degradation. We also demonstrated the imperfect rescue of Dex on the effects of cartilage degradation, with many catabolic factors being reduced, while other anabolic or chondroprotective processes were not.
Collapse
Affiliation(s)
- Rebecca Mae Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - André Struglics
- Department of Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pilar Lorenzo
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Viveka Tillgren
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Martin Rydén
- Department of Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Patrik Önnerfjord
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
16
|
Cortes-Lopez R, Barjaktarevic I. Alpha-1 Antitrypsin Deficiency: a Rare Disease? Curr Allergy Asthma Rep 2020; 20:51. [PMID: 32572624 DOI: 10.1007/s11882-020-00942-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Commonly categorized as a rare disease, alpha-1 antitrypsin deficiency (AATD) is neither rare, when compared to many other genetic disorders, nor an actual disease, but rather a predisposition toward a wide variety of diseases. It is one of the most common genetic disorders which can lead to a spectrum of clinical manifestations, ranging from no symptoms to progressively debilitating systemic disease, most commonly affecting the lung and liver. It is therefore imperative for clinicians to recognize and be familiar with the spectrum of presentations, methods of diagnosis, and clinical management of AATD. It is also imperative for scientists to recognize the potential for progress in the management of this disorder. RECENT FINDINGS This review focuses on the current state of knowledge of AATD, including the wide range of presentations, diagnosis, and clinical management. In addition to the clinical implications of severe AATD, we discuss the relevance of heterozygous state with mild or moderate AATD in the development of both lung and liver disease. While our understanding of the multiple roles of alpha-1 antitrypsin (AAT) is on the rise, with appreciation of its immunomodulatory, anti-infective, and anti-inflammatory properties, this knowledge has yet to impact our ability to predict outcomes. We discuss nuances of augmentation therapy and review novel therapeutic approaches currently under investigation. With the expanding knowledge about the complexities of AAT function and its clinical relevance, and with the increasing ability to diagnose early and intervene on AATD, it should be our goal to change the perception of AATD as a correctable inherited disorder rather than a fatal disease.
Collapse
Affiliation(s)
- Roxana Cortes-Lopez
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS, Los Angeles, CA, 90095, USA
| | - Igor Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS, Los Angeles, CA, 90095, USA.
| |
Collapse
|
17
|
Lior Y, Jasevitch M, Ochayon DE, Zaretsky M, Lewis EC, Aharoni A. Application of directed evolution and back-to-consensus algorithms to human alpha1-antitrypsin leads to diminished anti-protease activity and augmented anti-inflammatory activities. Cell Immunol 2020; 355:104135. [PMID: 32703529 DOI: 10.1016/j.cellimm.2020.104135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
Primarily known as an elastase inhibitor, human alpha1-antitrypsin also exerts anti-inflammatory and immunomodulatory effects, both in vitro and in vivo. While the anti-protease mechanism of alpha1-antitrypsin is attributed to a particular protein domain coined the reactive center loop, anti-inflammatory and immunomodulatory loci within the molecule remain to be identified. In the present study, directed evolution and back-to-consensus algorithms were applied to human alpha1-antitrypsin. Six unique functional candidate sites were identified on the surface of the molecule; in manipulating these sites by point mutations, a recombinant mutant form of alpha1-antitrypsin was produced, depicting a requirement for sites outside the reactive center loop as essential for protease inhibition, and displaying enhanced anti-inflammatory activities. Taken together, outcomes of the present study establish a potential use for directed evolution in advancing our understanding of site-specific protein functions, offering a platform for development of context- and disease-specific alpha1-antitrypsin-based therapeutics.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Maria Jasevitch
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - David E Ochayon
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Mariana Zaretsky
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Amir Aharoni
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| |
Collapse
|
18
|
Alpha-1-Antitrypsin Ameliorates Pristane Induced Diffuse Alveolar Hemorrhage in Mice. J Clin Med 2019; 8:jcm8091341. [PMID: 31470606 PMCID: PMC6780888 DOI: 10.3390/jcm8091341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/21/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
Diffuse alveolar hemorrhage (DAH) is a fatal complication in patients with lupus. DAH can be induced in B6 mice by an intraperitoneal injection of pristane. Since human alpha-1-antitrypsin (hAAT) is an anti-inflammatory and immuno-regulatory protein, we investigated the protective effect of hAAT against pristane-induced DAH in B6 mice and hAAT transgenic (hAAT-Tg) mice. We first showed that hAAT Tg expression lowers TNF-α production in B cells, as well as CD4+ T cells in untreated mice. Conversely, the frequency of regulatory CD4+CD25+ and CD4+CD25-IL-10+ cells was significantly higher in hAAT-Tg than in B6 mice. This confirmed the anti-inflammatory effect of hAAT that was observed even at steady state. One week after a pristane injection, the frequency of peritoneal Ly6Chi inflammatory monocytes and neutrophils in hAAT-Tg mice was significantly lower than that in B6 mice. Importantly, pristane-induced DAH was completely prevented in hAAT-Tg mice and this was associated with a modulation of anti- to pro-inflammatory myeloid cell ratio/balance. We also showed that treatment with hAAT decreased the severity of DAH in B6 mice. These results showed for the first time that hAAT has a therapeutic potential for the treatment of DAH.
Collapse
|
19
|
Ma H, Lu Y, Lowe K, van der Meijden-Erkelens L, Wasserfall C, Atkinson MA, Song S. Regulated hAAT Expression from a Novel rAAV Vector and Its Application in the Prevention of Type 1 Diabetes. J Clin Med 2019; 8:jcm8091321. [PMID: 31466263 PMCID: PMC6780368 DOI: 10.3390/jcm8091321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 12/26/2022] Open
Abstract
We, and others, have previously achieved high and sustained levels of transgene expression from viral vectors, such as recombinant adeno-associated virus (rAAV). However, regulatable transgene expression may be preferred in gene therapy for diseases, such as type 1 diabetes (T1D) and rheumatoid arthritis (RA), in which the timing and dosing of the therapeutic gene product play critical roles. In the present study, we generated a positive feedback regulation system for human alpha 1 antitrypsin (hAAT) expression in the rAAV vector. We performed quantitative kinetics studies in vitro and in vivo demonstrating that this vector system can mediate high levels of inducible transgene expression. Transgene induction could be tailored to occur rapidly or gradually, depending on the dose of the inducing drug, doxycycline (Dox). Conversely, after withdrawal of Dox, the silencing of transgene expression occurred slowly over the course of several weeks. Importantly, rAAV delivery of inducible hAAT significantly prevented T1D development in non-obese diabetic (NOD) mice. These results indicate that this Dox-inducible vector system may facilitate the fine-tuning of transgene expression, particularly for hAAT treatment of human autoimmune diseases, including T1D.
Collapse
Affiliation(s)
- Hongxia Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA
| | - Yuanqing Lu
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA
| | - Keith Lowe
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA
| | | | - Clive Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sihong Song
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
20
|
Reeves EP, Dunlea DM, McQuillan K, O'Dwyer CA, Carroll TP, Saldova R, Akepati PR, Wormald MR, McElvaney OJ, Shutchaidat V, Henry M, Meleady P, Keenan J, Liberti DC, Kotton DN, Rudd PM, Wilson AA, McElvaney NG. Circulating Truncated Alpha-1 Antitrypsin Glycoprotein in Patient Plasma Retains Anti-Inflammatory Capacity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:2240-2253. [PMID: 30796179 PMCID: PMC6452030 DOI: 10.4049/jimmunol.1801045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/30/2019] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland;
| | - Danielle M Dunlea
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Karen McQuillan
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Ciara A O'Dwyer
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Tomás P Carroll
- Alpha-1 Foundation Ireland, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Radka Saldova
- GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Dublin, Ireland
| | - Prithvi Reddy Akepati
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118
| | - Mark R Wormald
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford OX1 3QU, United Kingdom; and
| | - Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Vipatsorn Shutchaidat
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Joanne Keenan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Derek C Liberti
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118
| | - Pauline M Rudd
- Alpha-1 Foundation Ireland, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Andrew A Wilson
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
21
|
Elshikha AS, Yuan Y, Lu Y, Chen MJ, Abboud G, Akbar MA, Plate H, Wolney H, Hoffmann T, Tagari E, Zeumer L, Morel L, Song S. Alpha 1 Antitrypsin Gene Therapy Extends the Lifespan of Lupus-Prone Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:131-142. [PMID: 30547047 PMCID: PMC6258868 DOI: 10.1016/j.omtm.2018.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/13/2018] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by high levels of pathogenic autoantibodies and tissue damage. Multiple studies showed that dendritic cell (DC) activation plays a critical role in SLE pathogenesis. Human alpha 1 antitrypsin (hAAT) is a serine proteinase inhibitor with potent anti-inflammatory and cytoprotective properties. In this study, we first examined the effects of hAAT on the functions of DCs from lupus-prone mice, and we showed that hAAT treatment efficiently inhibited CpG- (TLR9 agonist) induced activation of bone marrow-derived conventional and plasmacytoid DCs as well as the production of pro-inflammatory cytokines. The hAAT treatment also attenuated DC help for B cell proliferation and immunoglobulin M (IgM) production. We next tested the protective effect of hAAT protein and gene therapy using recombinant adeno-associated virus 8 (rAAV8-CB-hAAT) in a spontaneous lupus mouse model, and we showed that both treatments decreased autoantibody levels. Importantly, rAAV8-CB-hAAT did not induce an immune response to its transgene product (hAAT), but it showed more pronounced therapeutic effects in reducing urine protein levels and extending the lifespan of these mice. These results indicate that AAT has therapeutic potential in the treatment of SLE in humans.
Collapse
Affiliation(s)
- Ahmed Samir Elshikha
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Sharkia, Egypt
| | - Ye Yuan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Yuanqing Lu
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Mong-Jen Chen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mohammad Ahsanul Akbar
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Henrike Plate
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Hedwig Wolney
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Tanja Hoffmann
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Eleni Tagari
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Leilani Zeumer
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
22
|
Song S. Alpha-1 Antitrypsin Therapy for Autoimmune Disorders. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:289-301. [PMID: 30723786 DOI: 10.15326/jcopdf.5.4.2018.0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Autoimmune diseases are conditions caused by an over reactive immune system that attacks self-tissues and organs. Although the pathogenesis of autoimmune disease is complex and multi-factorial, inflammation is commonly involved. Therefore, anti-inflammatory therapies hold potential for the treatment of autoimmune diseases. However, long-term control of inflammation is challenging and most of the currently used drugs have side effects. Alpha-1 antitrypsin (AAT) is an anti-inflammatory protein with a well-known safety profile. The therapeutic potential of AAT has been tested in several autoimmune disease models. The first study using a recombinant adeno-associated viral (rAAV) vector showed that AAT gene transfer prevented the development of type 1 diabetes (T1D) in the non-obese diabetic (NOD) mouse model. Subsequent studies showed that treatment with AAT protein prevented and reversed type 1 diabetes. The beneficial effects of AAT treatment have also been observed in other autoimmune disease models such as rheumatoid arthritis and systemic lupus erythematosus. This paper reviews the therapeutic application of AAT and discusses possible mechanisms of action in various autoimmune diseases.
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville
| |
Collapse
|
23
|
Baranovski BM, Schuster R, Nisim O, Brami I, Lior Y, Lewis EC. Alpha-1 Antitrypsin Substitution for Extrapulmonary Conditions in Alpha-1 Antitrypsin Deficient Patients. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:267-276. [PMID: 30723784 DOI: 10.15326/jcopdf.5.4.2017.0161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a genetic disorder which most commonly manifests as pulmonary emphysema. Accordingly, alpha-1 antitrypsin (AAT) augmentation therapy aims to reduce the progression of emphysema, as achieved by life-long weekly slow-drip infusions of plasma-derived affinity-purified human AAT. However, not all AATD patients will receive this therapy, due to either lack of medical coverage or low patient compliance. To circumvent these limitations, attempts are being made to develop lung-directed therapies, including inhaled AAT and locally-delivered AAT gene therapy. Lung transplantation is also an ultimate therapy option. Although less common, AATD patients also present with disease manifestations that extend beyond the lung, including vasculitis, diabetes and panniculitis, and appear to experience longer and more frequent hospitalization times and more frequent pneumonia bouts. In the past decade, new mechanism-based clinical indications for AAT therapy have surfaced, depicting a safe, anti-inflammatory, immunomodulatory and tissue-protective agent. Introduced to non-AATD individuals, AAT appears to provide relief from steroid-refractory graft-versus-host disease, from bacterial infections in cystic fibrosis and from autoimmune diabetes; preclinical studies show benefit also in multiple sclerosis, ulcerative colitis, rheumatoid arthritis, acute myocardial infarction and stroke, as well as ischemia-reperfusion injury and aberrant wound healing processes. While the current augmentation therapy is targeted towards treatment of emphysema, it is suggested that AATD patients may benefit from AAT augmentation therapy geared towards extrapulmonary pathologies as well. Thus, development of mechanism-based, context-specific AAT augmentation therapy protocols is encouraged. In the current review, we will discuss extrapulmonary manifestations of AATD and the potential of AAT augmentation therapy for these conditions.
Collapse
Affiliation(s)
- Boris M Baranovski
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Omer Nisim
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ido Brami
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yotam Lior
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
24
|
Gatto M, Luisetto R, Ghirardello A, Cavicchioli L, Codolo G, Biasiolo A, Maggioni G, Saccon F, Beggio M, Cappon A, Venturini R, Pontisso P, Doria A. SERPINB3 Delays Glomerulonephritis and Attenuates the Lupus-Like Disease in Lupus Murine Models by Inducing a More Tolerogenic Immune Phenotype. Front Immunol 2018; 9:2081. [PMID: 30254646 PMCID: PMC6141748 DOI: 10.3389/fimmu.2018.02081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022] Open
Abstract
Objective: To explore the effects of SERPINB3 administration in murine lupus models with a focus on lupus-like nephritis. Methods: 40 NZB/W F1 mice were subdivided into 4 groups and intraperitoneally injected with recombinant SERPINB3 (7.5 μg/0.1 mL or 15 μg/0.1 mL) or PBS (0.1 mL) before (group 1 and 2) or after (group 3 and 4) the development of proteinuria (≥100 mg/dl). Two additional mice groups were provided by including 20 MRL/lpr mice which were prophylactically injected with SERPINB3 (10 mice, group 5) or PBS (10 mice, group 6). Time of occurrence and levels of anti-dsDNA and anti-C1q antibodies, proteinuria and serum creatinine, overall- and proteinuria-free survival were assessed in mice followed up to natural death. Histological analysis was performed in kidneys of both lupus models. The Th17:Treg cell ratio was assessed by flow-cytometry in splenocytes of treated and untreated MRL/lpr mice. Statistical analysis was performed using non parametric tests and Kaplan-Meier curves, when indicated. Results: Autoantibody levels and proteinuria were significantly decreased and time of occurrence significantly delayed in SERPINB3-treated mice vs. controls. In agreement with these findings, proteinuria-free and overall survival were significantly improved in SERPINB3-treated groups vs. controls. Histological analysis demonstrated a lower prevalence of severe tubular lesions in kidneys of group 5 vs. group 6. SERPINB3-treated mice showed an overall trend toward a reduced prevalence of severe lesions in both strains. Th17:Treg ratio was significantly decreased in splenocytes of MRL/lpr mice treated with SERPINB3, compared to untreated control mice. Conclusions: SERPINB3 significantly improves disease course and delays the onset of severe glomerulonephritis in lupus-prone mice, possibly inducing a more tolerogenic immune phenotype.
Collapse
Affiliation(s)
- Mariele Gatto
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anna Ghirardello
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Laura Cavicchioli
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Gaia Codolo
- Department of Biology, University of Padova, Padova, Italy
| | - Alessandra Biasiolo
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Maggioni
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesca Saccon
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marianna Beggio
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Cappon
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberta Venturini
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
| | - Patrizia Pontisso
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
25
|
Gene Delivery of Alpha-1-Antitrypsin Using Recombinant Adeno-Associated Virus (rAAV). Methods Mol Biol 2018. [PMID: 30194601 DOI: 10.1007/978-1-4939-8645-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The challenge for alpha-1-antitrypsin (AAT also known as SERPINA1) gene therapy is to achieve long term and high levels of AAT production. Recombinant adeno-associated virus (rAAV) vector has several advantages for AAT gene delivery including no viral genes in the vector, no requirement of integration for long-term transgene expression, low immunogenicity, and wide tropism. AAV-mediated AAT gene therapy has been developed and tested in animal models for AAT deficiency, type 1 diabetes, rheumatoid arthritis, and osteoporosis. AAV-mediated AAT gene therapy has also been tested in clinical studies and has shown promising results. Here we describe the methods of rAAV-AAT vector construction and production as well as AAT gene delivery through (1) liver-directed, (2) muscle-directed, and (3) mesenchymal stem cell (MSC)-mediated routes. We will also describe methods for the evaluation of AAT expression for each delivery approach.
Collapse
|
26
|
Kim M, Cai Q, Oh Y. Therapeutic potential of alpha-1 antitrypsin in human disease. Ann Pediatr Endocrinol Metab 2018; 23:131-135. [PMID: 30286568 PMCID: PMC6177666 DOI: 10.6065/apem.2018.23.3.131] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
Alpha-1 antitrypsin (AAT), an alpha globulin glycoprotein, is a member of the serine protease inhibitor (serpin) superfamily. The clinical significance of AAT is highlighted by AAT deficiency. Genetic deficiency of AAT can present as several neutrophilic diseases associated with emphysema, liver cirrhosis, panniculitis, and systemic vasculitis. Recently, animal and human studies have shown that AAT can control inflammatory, immunological, and tissue-protective responses. In addition, AAT treatment can prevent overt hyperglycemia, increase insulin secretion, and reduce cytokine-mediated apoptosis of pancreatic β-cells in diabetes. These multifunctional roles of AAT draw attention to the glycoprotein's therapeutic potential for many inflammatory and autoimmune diseases beyond AAT deficiency. As underlying mechanisms, recent studies have suggested the importance of serine protease inhibitory activity of AAT in obesity-associated insulin resistance, chronic obstructive pulmonary disease, and cystic fibrosis. In this review, we explore the multiple functions of AAT, in particular, the anti-inflammatory and serine protease inhibitory functions, and AAT's therapeutic potential in a variety of human diseases through published literature.
Collapse
Affiliation(s)
- Minsun Kim
- Department of Pediatrics, Chonbuk National University Medical School, Jeonju, Korea,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Qing Cai
- Department of Pathology, School of Medicine Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Youngman Oh
- Department of Pathology, School of Medicine Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA,Address for correspondence: Youngman Oh, PhD Department of Pathology, School of Medicine Medical College of Virginia Campus, Virginia Commonwealth University, 1101 East Marshall St., P.O. Box 980662, Richmond, VA 23298-0662, USA Tel: +1-804-827-1324 Fax: +1-804-828-9749 E-mail:
| |
Collapse
|
27
|
Zhang Z, Li P, Lin D, Wang D, Zhang Y. Proteome analysis of the potential serum biomarkers for chronic benzene poisoning. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:157-164. [PMID: 29729575 DOI: 10.1016/j.etap.2018.04.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 06/08/2023]
Abstract
The aim of our study is to seek novel specific biomarkers which could provide clues to the mechanism of chronic benzene poisoning (CBP) and might also be used as specific markers for early detection and diagnosis. In this study, a comparative serological proteome analysis between normal controls and CBP patients at three different levels of poisoning were performed via a 2D-DIGE and MALDI-TOF-MS. As the result a total of 10 proteins were found significantly altered between the normal and the mild, moderate and severe poisoning. The identified differentially expressed proteins were classified according to their molecular functions, biological processes, and protein classes, and three important serum proteins among them, apolipoproteinA-1, alpha-1-antitrypsin and complement C3, were further confirmed by immune turbidimetric analysis for their significant up-regulation in the CBP patients. Our findings suggest that these differential proteins may help elucidate the mechanism of CBP and provide potential biomarkers for diagnosis.
Collapse
Affiliation(s)
- Zhimin Zhang
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518001, Guangdong, China
| | - Peimao Li
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518001, Guangdong, China
| | - Dafeng Lin
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518001, Guangdong, China
| | - Dianpeng Wang
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518001, Guangdong, China
| | - Yanfang Zhang
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518001, Guangdong, China.
| |
Collapse
|
28
|
Mirea AM, Tack CJ, Chavakis T, Joosten LAB, Toonen EJM. IL-1 Family Cytokine Pathways Underlying NAFLD: Towards New Treatment Strategies. Trends Mol Med 2018; 24:458-471. [PMID: 29665983 PMCID: PMC5939989 DOI: 10.1016/j.molmed.2018.03.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide. Pathways responsible for the activation of IL-1 family cytokines are key in the development of NAFLD but underlying mechanisms are not fully understood. Many studies have focused on the inflammasome-caspase-1 pathway and have shown that this pathway is an important inducer of inflammation in NAFLD. However, this pathway is not solely responsible for the activation of proinflammatory cytokines. Also, neutrophil serine proteases (NSPs) are capable of activating cytokines and recent studies reported that these proteases also contribute to NAFLD. These studies provided, for the first time, evidence that this inflammasome-independent pathway is involved in NAFLD. In our opinion, these new insights open up new approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik J M Toonen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; R&D Department, Hycult Biotech, Uden, The Netherlands.
| |
Collapse
|
29
|
Lior Y, Zaretsky M, Ochayon DE, Lotysh D, Baranovski BM, Schuster R, Guttman O, Aharoni A, Lewis EC. Point Mutation of a Non-Elastase-Binding Site in Human α1-Antitrypsin Alters Its Anti-Inflammatory Properties. Front Immunol 2018; 9:759. [PMID: 29780379 PMCID: PMC5946014 DOI: 10.3389/fimmu.2018.00759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/27/2018] [Indexed: 12/20/2022] Open
Abstract
Introduction Human α1-antitrypsin (hAAT) is a 394-amino acid long anti-inflammatory, neutrophil elastase inhibitor, which binds elastase via a sequence-specific molecular protrusion (reactive center loop, RCL; positions 357-366). hAAT formulations that lack protease inhibition were shown to maintain their anti-inflammatory activities, suggesting that some attributes of the molecule may reside in extra-RCL segments. Here, we compare the protease-inhibitory and anti-inflammatory profiles of an extra-RCL mutation (cys232pro) and two intra-RCL mutations (pro357cys, pro357ala), to naïve [wild-type (WT)] recombinant hAAT, in vitro, and in vivo. Methods His-tag recombinant point-mutated hAAT constructs were expressed in HEK-293F cells. Purified proteins were evaluated for elastase inhibition, and their anti-inflammatory activities were assessed using several cell-types: RAW264.7 cells, mouse bone marrow-derived macrophages, and primary peritoneal macrophages. The pharmacokinetics of the recombinant variants and their effect on LPS-induced peritonitis were determined in vivo. Results Compared to WT and to RCL-mutated hAAT variants, cys232pro exhibited superior anti-inflammatory activities, as well as a longer circulating half-life, despite all three mutated forms of hAAT lacking anti-elastase activity. TNFα expression and its proteolytic membranal shedding were differently affected by the variants; specifically, cys232pro and pro357cys altered supernatant and serum TNFα dynamics without suppressing transcription or shedding. Conclusion Our data suggest that the anti-inflammatory profile of hAAT extends beyond direct RCL regions. Such regions might be relevant for the elaboration of hAAT formulations, as well as hAAT-based drugs, with enhanced anti-inflammatory attributes.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Mariana Zaretsky
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| | - David E Ochayon
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Diana Lotysh
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| | - Boris M Baranovski
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Ofer Guttman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Amir Aharoni
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
30
|
Wilkinson DJ, Arques MDC, Huesa C, Rowan AD. Serine proteinases in the turnover of the cartilage extracellular matrix in the joint: implications for therapeutics. Br J Pharmacol 2018; 176:38-51. [PMID: 29473950 DOI: 10.1111/bph.14173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 12/24/2022] Open
Abstract
Cartilage destruction is a key characteristic of arthritic disease, a process now widely established to be mediated by metzincins such as MMPs. Despite showing promise in preclinical trials during the 1990s, MMP inhibitors for the blockade of extracellular matrix turnover in the treatment of cancer and arthritis failed clinically, primarily due to poor selectivity for target MMPs. In recent years, roles for serine proteinases in the proteolytic cascades leading to cartilage destruction have become increasingly apparent, renewing interest in the potential for new therapeutic strategies that utilize pharmacological inhibitors against this class of proteinases. Herein, we describe key serine proteinases with likely importance in arthritic disease and highlight recent advances in this field. LINKED ARTICLES: This article is part of a themed section on Translating the Matrix. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.1/issuetoc.
Collapse
Affiliation(s)
- David J Wilkinson
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Maria Del Carmen Arques
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Carmen Huesa
- Institute of Biomedical and Environmental Health Research, University of the West of Scotland, Paisley, UK
| | - Andrew D Rowan
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
31
|
Yuan Y, DiCiaccio B, Li Y, Elshikha AS, Titov D, Brenner B, Seifer L, Pan H, Karic N, Akbar MA, Lu Y, Song S, Zhou L. Anti-inflammaging effects of human alpha-1 antitrypsin. Aging Cell 2018; 17:e12694. [PMID: 29045001 PMCID: PMC5770780 DOI: 10.1111/acel.12694] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2017] [Indexed: 12/21/2022] Open
Abstract
Inflammaging plays an important role in most age-related diseases. However, the mechanism of inflammaging is largely unknown, and therapeutic control of inflammaging is challenging. Human alpha-1 antitrypsin (hAAT) has immune-regulatory, anti-inflammatory, and cytoprotective properties as demonstrated in several disease models including type 1 diabetes, arthritis, lupus, osteoporosis, and stroke. To test the potential anti-inflammaging effect of hAAT, we generated transgenic Drosophila lines expressing hAAT. Surprisingly, the lifespan of hAAT-expressing lines was significantly longer than that of genetically matched controls. To understand the mechanism underlying the anti-aging effect of hAAT, we monitored the expression of aging-associated genes and found that aging-induced expressions of Relish (NF-ĸB orthologue) and Diptericin were significantly lower in hAAT lines than in control lines. RNA-seq analysis revealed that innate immunity genes regulated by NF-kB were significantly and specifically inhibited in hAAT transgenic Drosophila lines. To confirm this anti-inflammaging effect in human cells, we treated X-ray-induced senescence cells with hAAT and showed that hAAT treatment significantly decreased the expression and maturation of IL-6 and IL-8, two major factors of senescence-associated secretory phenotype. Consistent with results from Drosophila,RNA-seq analysis also showed that hAAT treatment significantly inhibited inflammation related genes and pathways. Together, our results demonstrated that hAAT significantly inhibited inflammaging in both Drosophila and human cell models. As hAAT is a FDA-approved drug with a confirmed safety profile, this novel therapeutic potential may make hAAT a promising candidate to combat aging and aging-related diseases.
Collapse
Affiliation(s)
- Ye Yuan
- Department of PharmaceuticsUniversity of FloridaGainesvilleFLUSA
| | - Benedetto DiCiaccio
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Ying Li
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | | | - Denis Titov
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Brian Brenner
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Lee Seifer
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Hope Pan
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Nurdina Karic
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | | | - Yuanqing Lu
- Department of PharmaceuticsUniversity of FloridaGainesvilleFLUSA
| | - Sihong Song
- Department of PharmaceuticsUniversity of FloridaGainesvilleFLUSA
- University of Florida Genetics InstituteGainesvilleFLUSA
| | - Lei Zhou
- Department of Molecular Genetics & MicrobiologyUniversity of FloridaGainesvilleFLUSA
- University of Florida Genetics InstituteGainesvilleFLUSA
- UF Health Cancer CenterGainesvilleFLUSA
| |
Collapse
|
32
|
Song S, Lu Y, Elshikha AS. In Vivo Analysis of Alpha-1-Antitrypsin Functions in Autoimmune Disease Models. Methods Mol Biol 2018; 1826:143-155. [PMID: 30194599 DOI: 10.1007/978-1-4939-8645-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Alpha-1-antitrypsin (AAT) is a circulating protein, a serpin, with multiple protective functions. Beside the well-known proteinase inhibitory function, which protects the lungs from chronic obstructive pulmonary disease (COPD), many studies have shown that AAT inhibits pro-inflammatory cytokine gene expression and functions. These anti-inflammatory and immune-regulatory properties have led to studies testing the therapeutic effect of AAT in autoimmune disease models. Initially, a study using recombinant adeno-associated viral (rAAV) vector showed that AAT gene therapy prevented type 1 diabetes (T1D) development in a nonobese diabetic (NOD) mouse model. Consequently, several studies confirmed that AAT therapy prevented and reversed T1D. AAT therapy has also been tested and has demonstrated protective effects in a collagen-induced arthritis model and a systemic lupus erythematosus (SLE) mouse model. This chapter describes methods that evaluate AAT functions in autoimmune mouse models.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Dependovirus
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Genetic Therapy
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/pathology
- Lupus Erythematosus, Systemic/therapy
- Mice
- Mice, Inbred NOD
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/pathology
- Pulmonary Disease, Chronic Obstructive/therapy
- Transduction, Genetic
- alpha 1-Antitrypsin/biosynthesis
- alpha 1-Antitrypsin/genetics
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Yuanqing Lu
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ahmed S Elshikha
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Low Levels of IgG Recognizing the α-1-Antitrypsin Peptide and Its Association with Taiwanese Women with Primary Sjögren's Syndrome. Int J Mol Sci 2017; 18:ijms18122750. [PMID: 29258281 PMCID: PMC5751349 DOI: 10.3390/ijms18122750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 01/06/2023] Open
Abstract
The aim of this study was to examine oxidative stress and low level of α-1-antitrypsin (A1AT) in primary Sjögren’s syndrome (pSS), and evaluate the associated autoreactivity against unmodified and their 4-hydroxy-2-nonenal (HNE)-modified peptides with pSS. Two differentially expressed proteins, α-1-acid glycoprotein 1 (A1AG1) and A1AT, exhibited 2-fold differences, and their HNE modifications were identified by depleted-albumin and immunoglobulin G (IgG) serum protein, in-solution digestion, in-gel digestion, and nano-liquid chromatography–tandem mass spectrometry (nano-LC-MS/MS) from pSS patients and age-matched healthy controls (HCs). Furthermore, levels of proteins, confirmation of HNE modifications, HNE-protein adducts and autoreactivity against unmodified and their HNE-modified peptides were further validated. Levels of the HNE-protein adduct and A1AG1 were significantly higher in pSS patients than HCs, but levels of A1AT were significantly lower in pSS patients compared to HCs. Only the HNE modification of A1AT was confirmed. Our study suggests that elevated HNE-protein adduct, oxidative stress, level (odds ratio (OR) 4.877, p = 0.003), lowered A1AT level (OR 3.910, p = 0.010) and a decreased level of anti-A1AT50–63 IgG (OR 3.360, p = 0.010) showed an increased risk in pSS patients compared to HCs, respectively.
Collapse
|
34
|
McCarthy C, Reeves EP, Orr C, Fearon U, Veale DJ, McElvaney NG. Reply. Arthritis Rheumatol 2017; 69:2404-2406. [DOI: 10.1002/art.40309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Cormac McCarthy
- Royal College of Surgeons in Ireland; and Beaumont Hospital; Dublin Ireland
| | - Emer P. Reeves
- Royal College of Surgeons in Ireland; and Beaumont Hospital; Dublin Ireland
| | - Carl Orr
- St. Vincent's University Hospital; Dublin Academic Health Care; and University College Dublin; Dublin Ireland
| | - Ursula Fearon
- St. Vincent's University Hospital; Dublin Academic Health Care; and University College Dublin; Dublin Ireland
| | - Douglas J. Veale
- St. Vincent's University Hospital; Dublin Academic Health Care; and University College Dublin; Dublin Ireland
| | - Noel G. McElvaney
- Royal College of Surgeons in Ireland; and Beaumont Hospital; Dublin Ireland
| |
Collapse
|
35
|
Recombinant Human Alpha-1 Antitrypsin-Fc Fusion Protein Reduces Mouse Myocardial Inflammatory Injury After Ischemia-Reperfusion Independent of Elastase Inhibition. J Cardiovasc Pharmacol 2017; 68:27-32. [PMID: 26945157 DOI: 10.1097/fjc.0000000000000383] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alpha-1-antitrypsin (AAT) is an abundant plasma protein with neutrophil elastase-inhibiting activity, and AAT is available as a plasma-derived therapeutic (pAAT). In experimental myocardial infarction, pAAT reduced acute inflammatory injury because of ischemia-reperfusion. The aim of the present study was to assess the properties of a recombinant protein composed of human AAT fused to the human immunoglobulin (Ig) G1 Fc fragment (rhAAT-Fc) in experimental myocardial infarction. METHODS Ten-week-old CD1 male mice underwent transient occlusion (30 minutes) of the left anterior coronary artery. rhAAT-Fc (2 mg/kg) or pAAT (60 mg/kg) were administered upon reperfusion. We used human plasma-derived Ig (2 mg/kg) or a matching volume of NaCl 0.9% as control solutions. After 24 hours, infarct size and caspase-1 activity were quantified. The left ventricular ejection fraction (LVEF) was measured by echocardiography at 24 hours and 7 days. A variant of rhAAT-Fc lacking elastase inhibition activity, rhAAT-Fc, was also tested. RESULTS The rhAAT-Fc induced a significant reduction in infarct size (P < 0.01 vs. all controls, P > 0.05 vs. pAAT). Caspase-1 activity was reduced to the same degree with rhAAT-Fc and pAAT (-70%; P < 0.05; P > 0.05 rhAAT-Fc vs. pAAT). The effects on infarct size after a single administration were reflected by preservation of LVEF at 24 hours and 7 days (all P < 0.05). rhAAT-Fc without elastase inhibiting activity, rhAAT-Fc, conferred comparable effects on infarct size, caspase-1 activity, and LVEF (P > 0.2 vs. rhAAT-Fc). CONCLUSIONS The pAAT and recombinant human AAT-Fc reduce the acute myocardial inflammatory injury after ischemia-reperfusion in the mouse leading to preservation of viable myocardium and systolic function, independent on the effects on neutrophil elastase.
Collapse
|
36
|
McCarthy C, Orr C, Fee LT, Carroll TP, Dunlea DM, Hunt DJL, Dunne E, O'Connell P, McCarthy G, Kenny D, Fearon U, Veale DJ, Reeves EP, McElvaney NG. Brief Report: Genetic Variation of the α 1 -Antitrypsin Gene Is Associated With Increased Autoantibody Production in Rheumatoid Arthritis. Arthritis Rheumatol 2017; 69:1576-1579. [PMID: 28409899 DOI: 10.1002/art.40127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/11/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To examine the prevalence of α1 -antitrypsin deficiency (AATD) in rheumatoid arthritis (RA), and to determine whether AATD is associated with higher levels of rheumatoid factor (RF), antinuclear antibodies (ANAs), and anti-citrullinated peptide autoantibodies (ACPAs). METHODS RF, ANAs, and ACPAs were measured by standard immunoturbidimetry, immunofluorescence assay, and enzyme-linked immunosorbent assay, respectively. Characterization of AAT phenotypes was performed by isoelectric focusing and immunofixation. The chi-square test with Yates' correction and the Mann-Whitney U test were used to assess the prevalence of alleles associated with AATD in RA and to compare mean antibody titers, respectively. RESULTS Of 246 patients with RA, 24 who were heterozygous for AATD were identified, with no statistically significant difference in the prevalence of AATD between RA patients and the general population (P = 0.39). A positive association between heterozygosity for AATD and the production of ACPAs was observed (P < 0.0001), with increased ACPA titers recorded in the AATD RA cohort compared with the general population (P = 0.01). CONCLUSION AAT heterozygous status in RA is strongly associated with positive ACPAs and may define a distinct subset of patients with increased disease severity.
Collapse
Affiliation(s)
- Cormac McCarthy
- Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| | - Carl Orr
- St. Vincent's University Hospital, Dublin Academic Health Care, and University College Dublin, Dublin, Ireland
| | - Laura T Fee
- Alpha-One Foundation, Royal College of Surgeons in Ireland, and Beaumont Hospital, Dublin, Ireland
| | - Tomás P Carroll
- Alpha-One Foundation, Royal College of Surgeons in Ireland, and Beaumont Hospital, Dublin, Ireland
| | - Danielle M Dunlea
- Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| | - David J L Hunt
- Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| | - Eimear Dunne
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Geraldine McCarthy
- University College Dublin and Mater Misericordiae University Hospital, Dublin, Ireland
| | - Dermot Kenny
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ursula Fearon
- St. Vincent's University Hospital, Dublin Academic Health Care, and University College Dublin, Dublin, Ireland
| | - Douglas J Veale
- St. Vincent's University Hospital, Dublin Academic Health Care, and University College Dublin, Dublin, Ireland
| | - Emer P Reeves
- Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
37
|
Urbaniak B, Nowicki P, Sikorska D, Samborski W, Kokot ZJ. The feature selection approach for evaluation of potential rheumatoid arthritis markers using MALDI-TOF datasets. Anal Biochem 2017; 525:29-37. [DOI: 10.1016/j.ab.2017.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 10/20/2022]
|
38
|
iTRAQ-Based Proteomics of Chronic Renal Failure Rats after FuShengong Decoction Treatment Reveals Haptoglobin and Alpha-1-Antitrypsin as Potential Biomarkers. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:1480514. [PMID: 28536642 PMCID: PMC5425835 DOI: 10.1155/2017/1480514] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/29/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023]
Abstract
Background. Chronic renal failure (CRF) has become a global health problem and bears a huge economic burden. FuShengong Decoction (FSGD) as traditional Chinese medicine has multiple pharmacological effects. Objectives. To understand the underlying molecular mechanism and signaling pathway involved in the FSGD treatment of CRF and screen differentially expressed proteins in rats with CRF treated with FSGD. Methods. Thirty-three male Sprague-Dawley rats were randomly divided into control group, CRF group, and FSGD group. Differentially expressed proteins were screened by iTRAQ coupled with nanoLC-MS/MS, and these identified proteins were later analyzed by GO, KEGG, and STRING. Additionally, haptoglobin (HP) and alpha-1-antitrypsin (AAT) were finally verified by ELISA, Western blot, and real time PCR. Results. A total of 417 proteins were identified. Nineteen differentially expressed proteins were identified in the FSGD group compared with the model group, of which 3 proteins were upregulated and 16 proteins were downregulated. Cluster analysis indicated that inflammatory response was associated with these proteins and complement and coagulation cascade pathways were predominantly involved. The validation methods further confirmed that the levels of HP and AAT were significantly increased. Conclusions. HP and AAT may be the important biomarkers in the pathogenesis of CRF and FSGD therapy.
Collapse
|
39
|
Akbar MA, Nardo D, Chen MJ, Elshikha AS, Ahamed R, Elsayed EM, Bigot C, Holliday LS, Song S. Alpha-1 antitrypsin inhibits RANKL-induced osteoclast formation and functions. Mol Med 2017; 23:57-69. [PMID: 28332697 DOI: 10.2119/molmed.2016.00170] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 03/15/2017] [Indexed: 01/05/2023] Open
Abstract
Osteoporosis is a global public health problem affecting more than 200 million people worldwide. We previously showed that treatment with alpha-1 antitrypsin (AAT), a multifunctional protein with anti-inflammatory properties, mitigated bone loss in an ovariectomized mouse model. However, the underlying mechanisms of the protective effect of AAT on bone tissue are largely unknown. In this study, we investigated the effect of AAT on osteoclast formation and function in vitro. Our results showed that AAT dose-dependently inhibited the formation of RANKL (receptor activator of nuclear factor κB ligand) induced osteoclasts derived from mouse bone marrow macrophages/monocyte (BMM) lineage cells and the murine macrophage cell line, RAW 264.7 cells. In order to elucidate the possible mechanisms underlying this inhibition, we tested the effect of AAT on the gene expression of cell surface molecules, transcription factors, and cytokines associated with osteoclast formation. We showed that AAT inhibited M-CSF (macrophage colony-stimulating factor) induced cell surface RANK expression in osteoclast precursor cells. In addition, AAT inhibited RANKL-induced TNF-α production, cell surface CD9 expression, and dendritic cell-specific transmembrane protein (DC-STAMP) gene expression. Importantly, AAT treatment significantly inhibited osteoclast-associated mineral resorption. Together, these results uncovered new mechanisms for the protective effects of AAT and strongly support the notion that AAT has therapeutic potential for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Mohammad Ahsanul Akbar
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - David Nardo
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Mong-Jen Chen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Ahmed S Elshikha
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Rubina Ahamed
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Eslam M Elsayed
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Egypt
| | - Claire Bigot
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Lexie Shannon Holliday
- Department of Orthodontics, College of Dentistry, University of Florida, Gainesville, Florida, USA.,Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
40
|
Identification of carbamylated alpha 1 anti-trypsin (A1AT) as an antigenic target of anti-CarP antibodies in patients with rheumatoid arthritis. J Autoimmun 2017; 80:77-84. [PMID: 28291659 DOI: 10.1016/j.jaut.2017.02.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 01/14/2023]
Abstract
In 2011 a novel autoantibody system, anti-carbamylated protein (anti-CarP) antibodies, was described in rheumatoid arthritis (RA) patients. Anti-CarP antibody positivity associates with a more severe disease course, is observed years before disease onset, and may predict the development of RA in arthralgia patients. Although many clinical observations have been carried out, information on the antigenic targets of anti-CarP antibodies is limited. Most studies on anti-CarP antibodies utilize an ELISA-based assay with carbamylated fetal calf serum (Ca-FCS) as antigen, a complex mixture of proteins. Therefore, we analysed the molecular identity of proteins within Ca-FCS that are recognized by anti-CarP antibodies. Ca-FCS was fractionated using ion exchange chromatography, selecting one of the fractions for further investigation. Using mass-spectrometry, carbamylated alpha-1-antitrypsin (Ca-A1AT) was identified as a potential antigenic target of anti-CarP antibodies in RA patients. A1AT contains several lysines on the protein surface that can readily be carbamylated. A large proportion of the RA patients harbour antibodies that bind human Ca-A1AT in ELISA, indicating that Ca-A1AT is indeed an autoantigen for anti-CarP antibodies. Next to the Ca-A1AT protein, several homocitrulline-containing peptides of A1AT were recognized by RA sera. Moreover, we identified a carbamylated peptide of A1AT in the synovial fluid of an RA patient using mass spectrometry. We conclude that Ca-A1AT is not only a target of anti-CarP antibodies but is also present in the synovial compartment, suggesting that Ca-A1AT recognized by anti-CarP antibodies in the joint may contribute to synovial inflammation in anti-CarP-positive RA.
Collapse
|
41
|
Kaneva MK, Greco KV, Headland SE, Montero-Melendez T, Mori P, Greenslade K, Pitzalis C, Moore A, Perretti M. Identification of Novel Chondroprotective Mediators in Resolving Inflammatory Exudates. THE JOURNAL OF IMMUNOLOGY 2017; 198:2876-2885. [PMID: 28242648 DOI: 10.4049/jimmunol.1601111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/29/2017] [Indexed: 12/11/2022]
Abstract
We hypothesized that exudates collected at the beginning of the resolution phase of inflammation might be enriched for tissue protective molecules; thus an integrated cellular and molecular approach was applied to identify novel chondroprotective bioactions. Exudates were collected 6 h (inflammatory) and 24 h (resolving) following carrageenan-induced pleurisy in rats. The resolving exudate was subjected to gel filtration chromatography followed by proteomics, identifying 61 proteins. Fractions were added to C28/I2 chondrocytes, grown in micromasses, ions with or without IL-1β or osteoarthritic synovial fluids for 48 h. Three proteins were selected from the proteomic analysis, α1-antitrypsin (AAT), hemopexin (HX), and gelsolin (GSN), and tested against catabolic stimulation for their effects on glycosaminoglycan deposition as assessed by Alcian blue staining, and gene expression of key anabolic proteins by real-time PCR. In an in vivo model of inflammatory arthritis, cartilage integrity was determined histologically 48 h after intra-articular injection of AAT or GSN. The resolving exudate displayed protective activities on chondrocytes, using multiple readouts: these effects were retained in low m.w. fractions of the exudate (46.7% increase in glycosaminoglycan deposition; ∼20% upregulation of COL2A1 and aggrecan mRNA expression), which reversed the effect of IL-1β. Exogenous administration of HX, GSN, or AAT abrogated the effects of IL-1β and osteoarthritic synovial fluids on anabolic gene expression and increased glycosaminoglycan deposition. Intra-articular injection of AAT or GSN protected cartilage integrity in mice with inflammatory arthritis. In summary, the strategy for identification of novel chondroprotective activities in resolving exudates identified HX, GSN and AAT as potential leads for new drug discovery programs.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Karin V Greco
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Sarah E Headland
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Trinidad Montero-Melendez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | | | | | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | | | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| |
Collapse
|
42
|
Maicas N, van der Vlag J, Bublitz J, Florquin S, Bakker-van Bebber M, Dinarello CA, Verweij V, Masereeuw R, Joosten LA, Hilbrands LB. Human Alpha-1-Antitrypsin (hAAT) therapy reduces renal dysfunction and acute tubular necrosis in a murine model of bilateral kidney ischemia-reperfusion injury. PLoS One 2017; 12:e0168981. [PMID: 28235038 PMCID: PMC5325207 DOI: 10.1371/journal.pone.0168981] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/11/2016] [Indexed: 11/18/2022] Open
Abstract
Several lines of evidence have demonstrated the anti-inflammatory and cytoprotective effects of alpha-1-antitrypsin (AAT), the major serum serine protease inhibitor. The aim of the present study was to investigate the effects of human AAT (hAAT) monotherapy during the early and recovery phase of ischemia-induced acute kidney injury. Mild renal ischemia-reperfusion (I/R) injury was induced in male C57Bl/6 mice by bilateral clamping of the renal artery and vein for 20 min. hAAT (80 mg/kg, Prolastin®) was administered daily intraperitoneally (i.p.) from day -1 until day 7 after surgery. Control animals received the same amount of human serum albumin (hAlb). Plasma, urine and kidneys were collected at 2h, 1, 2, 3, 8 and 15 days after reperfusion for histological and biochemical analysis. hAAT partially preserved renal function and tubular integrity after induction of bilateral kidney I/R injury, which was accompanied with reduced renal influx of macrophages and a significant decrease of neutrophil gelatinase-associated lipocalin (NGAL) protein levels in urine and plasma. During the recovery phase, hAAT significantly decreased kidney injury molecule-1 (KIM-1) protein levels in urine but showed no significant effect on renal fibrosis. Although the observed effect size of hAAT administration was limited and therefore the clinical relevance of our findings should be evaluated carefully, these data support the potential of this natural protein to ameliorate ischemic and inflammatory conditions.
Collapse
Affiliation(s)
- Nuria Maicas
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Janin Bublitz
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandrine Florquin
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medicine, University of Colorado Health Sciences Center Denver, Colorado, United States of America
| | - Vivienne Verweij
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Roos Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - Leo A Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
43
|
Aggarwal N, Korenbaum E, Mahadeva R, Immenschuh S, Grau V, Dinarello CA, Welte T, Janciauskiene S. α-Linoleic acid enhances the capacity of α-1 antitrypsin to inhibit lipopolysaccharide induced IL-1β in human blood neutrophils. Mol Med 2016; 22:680-693. [PMID: 27452044 DOI: 10.2119/molmed.2016.00119] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/05/2016] [Indexed: 11/06/2022] Open
Abstract
Alpha1-antitrypsin (A1AT, SERPINA1), a major circulating inhibitor of neutrophil elastase (NE) and proteinase-3 (PR3), has been proposed to reduce the processing and release of IL-1β. Since the anti-inflammatory properties of A1AT are influenced by the presence of polyunsaturated fatty acids, we compared effects of fatty acid-free (A1AT-0) and α-linoleic acid bound (A1AT-LA) forms of A1AT on lipopolysaccharide (LPS)-induced synthesis of IL-1β precursor and the release of IL-1β from human blood neutrophils. The presence of A1AT-LA or A1AT-0 significantly reduced LPS induced release of mature IL-1β. However, only A1AT-LA reduced both steady state mRNA levels of IL-1β and the secretion of mature IL-1β. In LPS-stimulated neutrophils, mRNA levels of TLR2/4, NFKBIA, P2RX7, NLRP3, and CASP1 decreased significantly in the presence of A1AT-LA but not A1AT-0. A1AT-0 and A1AT-LA did not inhibit the direct enzymatic activity of caspase-1, but we observed complexes of either form of A1AT with NE and PR3. Consistent with the effect on TLR and IL-1β gene expression, only A1AT-LA inhibited LPS-induced gene expression of NE and PR3. Increased gene expression of PPAR-γ was observed in A1AT-LA treated neutrophils without of LPS stimulation, and the selective PPAR-γ antagonist (GW9662) prevented the reduction in IL-1β by A1AT-LA. We conclude from our data, that the ability of A1AT to reduce TLR and IL-1β gene expression depends on its association with LA. Moreover, the anti-inflammatory properties of A1AT-LA are likely to be mediated by the activation of PPAR-γ.
Collapse
Affiliation(s)
- Nupur Aggarwal
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Elena Korenbaum
- Institute of Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Ravi Mahadeva
- Cambridge NIHR Biomedical Research Centre, Department of Respiratory Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Veronika Grau
- Department of General and Thoracic Surgery, Laboratory of Experimental Surgery, Justus-Liebig-University Giessen, Giessen, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045.,Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tobias Welte
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| |
Collapse
|
44
|
Akbar MA, Cao JJ, Lu Y, Nardo D, Chen MJ, Elshikha AS, Ahamed R, Brantly M, Holliday LS, Song S. Alpha-1 Antitrypsin Gene Therapy Ameliorates Bone Loss in Ovariectomy-Induced Osteoporosis Mouse Model. Hum Gene Ther 2016; 27:679-86. [DOI: 10.1089/hum.2016.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
| | - Jay J. Cao
- USDA, ARS Grand Fork Human Nutrition Research Center, Grand Fork, North Dakota
| | - Yuanqing Lu
- Department of Medicine, University of Florida, Gainesville, Florida
| | - David Nardo
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Mong-Jen Chen
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Ahmed S. Elshikha
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Rubina Ahamed
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Mark Brantly
- Department of Medicine, University of Florida, Gainesville, Florida
| | | | - Sihong Song
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| |
Collapse
|
45
|
Alpha 1 Antitrypsin Inhibits Dendritic Cell Activation and Attenuates Nephritis in a Mouse Model of Lupus. PLoS One 2016; 11:e0156583. [PMID: 27232337 PMCID: PMC4883758 DOI: 10.1371/journal.pone.0156583] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/17/2016] [Indexed: 01/11/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with a worldwide distribution and considerable mortality and morbidity. Although the pathogenesis of this disease remains elusive, over-reactive dendritic cells (DCs) play a critical role in the disease development. It has been shown that human alpha-1 antitrypsin (hAAT) has protective effects in type 1 diabetes and rheumatoid arthritis mouse models. In the present study, we tested the effect of AAT on DC differentiation and functions, as well as its protective effect in a lupus-prone mouse model. We showed that hAAT treatment significantly inhibited LPS (TLR4 agonist) and CpG (TLR9 agonist) -induced bone-marrow (BM)-derived conventional and plasmacytoid DC (cDC and pDC) activation and reduced the production of inflammatory cytokines including IFN-I, TNF-α and IL-1β. In MRL/lpr mice, hAAT treatment significantly reduced BM-derived DC differentiation, serum autoantibody levels, and importantly attenuated renal pathology. Our results for the first time demonstrate that hAAT inhibits DC activation and function, and it also attenuates autoimmunity and renal damage in the MRL/lpr lupus model. These results imply that hAAT has a therapeutic potential for the treatment of SLE in humans.
Collapse
|
46
|
Toonen EJM, Mirea AM, Tack CJ, Stienstra R, Ballak DB, van Diepen JA, Hijmans A, Chavakis T, Dokter WH, Pham CTN, Netea MG, Dinarello CA, Joosten LAB. Activation of proteinase 3 contributes to Non-alcoholic Fatty Liver Disease (NAFLD) and insulin resistance. Mol Med 2016; 22:molmed.2016.00033. [PMID: 27261776 DOI: 10.2119/molmed.2016.00033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 05/16/2016] [Indexed: 01/29/2023] Open
Abstract
Activation of inflammatory pathways is known to accompany development of obesity-induced non-alcoholic fatty liver disease (NAFLD), insulin resistance and type 2 diabetes. In addition to caspase-1, the neutrophil serine proteases proteinase 3, neutrophil elastase and cathepsin G are able to process the inactive pro-inflammatory mediators IL-1β and IL-18 to their bioactive forms, thereby regulating inflammatory responses. In the present study, we investigated whether proteinase 3 is involved in obesity-induced development of insulin resistance and NAFLD. We investigated the development of NAFLD and insulin resistance in mice deficient for neutrophil elastase/proteinase 3 and neutrophil elastase/cathepsin G and in wild-type mice treated with the neutrophil serine proteinase inhibitor human alpha-1 antitrypsin. Expression profiling of metabolically relevant tissues obtained from insulin resistant mice showed that expression of proteinase 3 was specifically upregulated in the liver, whereas neutrophil elastase, cathepsin G and caspase-1 were not. Neutrophil elastase/proteinase 3 deficient mice showed strongly reduced levels of lipids in the liver after fed a high fat diet. Moreover, these mice were resistant to high fat diet-induced weight gain, inflammation and insulin resistance. Injection of proteinase 3 exacerbated insulin resistance in caspase-1(-/-) mice, indicating that proteinase 3 acts independently of caspase-1. Treatment with alpha-1 antitrypsin during the last 10 days of a 16 week high fat diet reduced hepatic lipid content and decreased fasting glucose levels. We conclude that proteinase 3 is involved in NAFLD and insulin resistance and that inhibition of proteinase 3 may have therapeutic potential.
Collapse
Affiliation(s)
- Erik J M Toonen
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands.,Nutrition, Metabolism and Genomics Group, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Dov B Ballak
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Janna A van Diepen
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Anneke Hijmans
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, University Clinic Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Wim H Dokter
- Synthon Research Laboratories, Nijmegen, The Netherlands
| | - Christine T N Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
47
|
Baraldo S, Balestro E, Bazzan E, Tiné ME, Biondini D, Turato G, Cosio MG, Saetta M. Alpha-1 Antitrypsin Deficiency Today: New Insights in the Immunological Pathways. Respiration 2016; 91:380-5. [PMID: 27164860 DOI: 10.1159/000445692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/17/2016] [Indexed: 11/19/2022] Open
Abstract
More than 50 years ago, the observation that absence of the α1 band from protein electrophoresis is associated with severe emphysema established the link between α1-antitrypsin deficiency (AATD) and lung damage. From this discovery, the classic paradigm of protease/antiprotease imbalance was derived, linking lung destruction in patients with AATD to the unopposed effect of proteases. By extension, this paradigm was also applied to patients with 'common' chronic obstructive pulmonary disease, in whom large increases in smoke-induced proteases could overwhelm the antiprotease capability of AAT. However, it has become increasingly evident that AAT has important anti-inflammatory and immunoregulatory activities which, beside its antiprotease function, may be critically involved in lung destruction. From this perspective, we will consider recent evidence, based on epidemiological, clinical and immunopathological studies, suggesting that it is time to move on from the original protease/antiprotease paradigm toward a more complex view of the condition, which embraces its immunomodulating functions. Of importance, the potent immunoregulatory, tolerogenic role of AAT may support its therapeutic use in a number of diseases other than AATD, particularly in immune-related disorders.
Collapse
Affiliation(s)
- Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova and Padova City Hospital, Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Blanco I, Lipsker D, Lara B, Janciauskiene S. Neutrophilic panniculitis associated with alpha-1-antitrypsin deficiency: an update. Br J Dermatol 2016; 174:753-62. [DOI: 10.1111/bjd.14309] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2015] [Indexed: 12/13/2022]
Affiliation(s)
- I. Blanco
- Board of Directors of the Alpha1-Antitrypsin Deficiency Spanish Registry; Lung Foundation Breathe; Spanish Society of Pneumology (SEPAR), Provenza; 108 Bajo 08029 Barcelona Spain
| | - D. Lipsker
- Faculté de Médecine; Université de Strasbourg et Clinique Dermatologique; Hôpitaux Universitaires de Strasbourg; 1 Place de l'Hôpital 67091 Strasbourg CEDEX France
| | - B. Lara
- Respiratory Medicine Department; Royal Exeter and Devon Hospital; Exeter U.K
| | - S. Janciauskiene
- Department of Respiratory Medicine; Hannover Medical School; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH); Member of the German Center for Lung Research (DZL); 30626 Hanover Germany
| |
Collapse
|
49
|
Wanner A, Sandhaus RA. Alpha-1 Antitrypsin as a Therapeutic Agent for Conditions not Associated with Alpha-1 Antitrypsin Deficiency. ALPHA-1 ANTITRYPSIN 2016. [PMCID: PMC7121596 DOI: 10.1007/978-3-319-23449-6_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Alpha-1 antitrypsin is a positive acute phase reactant whose serum level rises in response to inflammatory stress, presumably to balance pro-inflammatory processes. In addition to its serine protease inhibitory action, alpha-1 antitrypsin exhibits broader anti-inflammatory and immunomodulatory activity, and increasing its serum concentration by the administration of exogenous alpha-1 antitrypsin to above-normal levels potentially could be therapeutic in conditions other than alpha-1 antitrypsin deficiency. In vitro observations, studies in animal models and in some instances early human trials suggest that intravenous or inhaled alpha-1 antitrypsin has beneficial effects in type 1 diabetes, viral infections, graft-versus-host disease, cystic fibrosis, and alpha-1 antitrypsin-replete chronic obstructive pulmonary disease among others. While the results of pivotal clinical trials have not been reported to date, new indications for alpha-1 antitrypsin therapy are likely to emerge in the future based on currently available scientific data.
Collapse
Affiliation(s)
- Adam Wanner
- University of Miami Miller School of Medicine, Miami, Florida USA
| | | |
Collapse
|
50
|
Hurley K, Reeves EP, Carroll TP, McElvaney NG. Tumor necrosis factor-α driven inflammation in alpha-1 antitrypsin deficiency: a new model of pathogenesis and treatment. Expert Rev Respir Med 2015; 10:207-22. [PMID: 26634397 DOI: 10.1586/17476348.2016.1127759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Alpha-1 antitrypsin (AAT) deficiency (AATD) has traditionally been thought of as a genetic disorder characterized by lung destruction and early emphysema in a low AAT, and high neutrophil elastase (NE) environment in the lungs of affected individuals. Recently, a growing body of evidence has emerged to support the hypothesis that tumor necrosis factor alpha (TNF-α) is essential in the pathogenesis of both genetic AATD and non-genetic chronic obstructive pulmonary disease (COPD). Reports have highlighted the importance of TNF-α driven immune cell dysfunction in the development of lung disease in AATD. The authors discuss the role of AAT as a key modulator of TNF-α signaling firstly in the setting of AATD and secondly in other conditions where AAT augmentation therapy has potential utility as a novel therapy.
Collapse
Affiliation(s)
- Killian Hurley
- a Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland Education and Research Centre , Beaumont Hospital , Dublin , Ireland
| | - Emer P Reeves
- a Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland Education and Research Centre , Beaumont Hospital , Dublin , Ireland
| | - Tomás P Carroll
- a Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland Education and Research Centre , Beaumont Hospital , Dublin , Ireland
| | - Noel G McElvaney
- a Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland Education and Research Centre , Beaumont Hospital , Dublin , Ireland
| |
Collapse
|