1
|
Liou TG, Argel N, Asfour F, Brown PS, Chatfield BA, Cox DR, Daines CL, Durham D, Francis JA, Glover B, Helms M, Heynekamp T, Hoidal JR, Jensen JL, Kartsonaki C, Keogh R, Kopecky CM, Lechtzin N, Li Y, Lysinger J, Molina O, Nakamura C, Packer KA, Paine R, Poch KR, Quittner AL, Radford P, Redway AJ, Sagel SD, Szczesniak RD, Sprandel S, Taylor-Cousar JL, Vroom JB, Yoshikawa R, Clancy JP, Elborn JS, Olivier KN, Adler FR. Airway inflammation accelerates pulmonary exacerbations in cystic fibrosis. iScience 2024; 27:108835. [PMID: 38384849 PMCID: PMC10879674 DOI: 10.1016/j.isci.2024.108835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/02/2023] [Accepted: 01/03/2024] [Indexed: 02/23/2024] Open
Abstract
Airway inflammation underlies cystic fibrosis (CF) pulmonary exacerbations. In a prospective multicenter study of randomly selected, clinically stable adolescents and adults, we assessed relationships between 24 inflammation-associated molecules and the future occurrence of CF pulmonary exacerbation using proportional hazards models. We explored relationships for potential confounding or mediation by clinical factors and assessed sensitivities to treatments including CF transmembrane regulator (CFTR) protein synthesis modulators. Results from 114 participants, including seven on ivacaftor or lumacaftor-ivacaftor, representative of the US CF population during the study period, identified 10 biomarkers associated with future exacerbations mediated by percent predicted forced expiratory volume in 1 s. The findings were not sensitive to anti-inflammatory, antibiotic, and CFTR modulator treatments. The analyses suggest that combination treatments addressing RAGE-axis inflammation, protease-mediated injury, and oxidative stress might prevent pulmonary exacerbations. Our work may apply to other airway inflammatory diseases such as bronchiectasis and the acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Theodore G Liou
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
- Primary Children's Cystic Fibrosis Center, Division of Pediatric Pulmonology, Department of Pediatrics, University of Utah, 81 North Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Natalia Argel
- Cystic Fibrosis Center, Phoenix Children's Hospital, 1919 East Thomas Road, Phoenix, AZ 85016, USA
| | - Fadi Asfour
- Primary Children's Cystic Fibrosis Center, Division of Pediatric Pulmonology, Department of Pediatrics, University of Utah, 81 North Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Perry S Brown
- St. Luke's Cystic Fibrosis Center of Idaho, 610 W. Hays Street, Boise, ID 83702, USA
| | - Barbara A Chatfield
- Primary Children's Cystic Fibrosis Center, Division of Pediatric Pulmonology, Department of Pediatrics, University of Utah, 81 North Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - David R Cox
- Nuffield College, 1 New Rd, Oxford OX1 1NF, UK
| | - Cori L Daines
- Division of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Arizona Health Sciences, University of Arizona, 1501 N. Campbell Avenue, Room 3301, PO Box 245073, Tucson, AZ 85724, USA
| | | | - Jessica A Francis
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Barbara Glover
- Cystic Fibrosis Center, 3006 S. Maryland Pkwy, Suite #315, Las Vegas, NV 89109, USA
| | - My Helms
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Theresa Heynekamp
- Adult Cystic Fibrosis Program, Division of Pulmonary, Critical Care and Sleep Medicine, DoIM MSC10-5550, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - John R Hoidal
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Judy L Jensen
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit and Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ruth Keogh
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Carol M Kopecky
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Noah Lechtzin
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD 21205, USA
| | - Yanping Li
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Jerimiah Lysinger
- Montana Cystic Fibrosis Center, Billings Clinic, 2800 10th Avenue N, Billings, MT 59101, USA
| | - Osmara Molina
- Division of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Arizona Health Sciences, University of Arizona, 1501 N. Campbell Avenue, Room 3301, PO Box 245073, Tucson, AZ 85724, USA
| | - Craig Nakamura
- Cystic Fibrosis Center, 3006 S. Maryland Pkwy, Suite #315, Las Vegas, NV 89109, USA
| | - Kristyn A Packer
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Robert Paine
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Katie R Poch
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | | | - Peggy Radford
- Cystic Fibrosis Center, Phoenix Children's Hospital, 1919 East Thomas Road, Phoenix, AZ 85016, USA
| | - Abby J Redway
- Adult Cystic Fibrosis Program, Division of Pulmonary, Critical Care and Sleep Medicine, DoIM MSC10-5550, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Scott D Sagel
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Rhonda D Szczesniak
- Division of Biostatistics & Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shawna Sprandel
- Montana Cystic Fibrosis Center, Billings Clinic, 2800 10th Avenue N, Billings, MT 59101, USA
| | - Jennifer L Taylor-Cousar
- Division of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
- Division of Pulmonology, Department of Pediatrics, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Jane B Vroom
- Adult Cystic Fibrosis Center, Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, 26 North Mario Capecchi Drive, Salt Lake City, UT 84132, USA
- Primary Children's Cystic Fibrosis Center, Division of Pediatric Pulmonology, Department of Pediatrics, University of Utah, 81 North Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Ryan Yoshikawa
- Cystic Fibrosis Center, 3006 S. Maryland Pkwy, Suite #315, Las Vegas, NV 89109, USA
| | - John P Clancy
- Former: Division of Pulmonary Medicine, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - J Stuart Elborn
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Health Sciences Building, Lisburn Rd, Belfast BT9 7AE, UK
| | - Kenneth N Olivier
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive MSC1454, Building 10-CRC, Room 1408A, Bethesda, MD 20892, USA
| | - Frederick R Adler
- Department of Mathematics, 155 South 1400 East, University of Utah, Salt Lake City, UT 84112, USA
- School of Biological Sciences, 257 South 1400 East, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
2
|
Varjú I, Tóth E, Farkas ÁZ, Farkas VJ, Komorowicz E, Feller T, Kiss B, Kellermayer MZ, Szabó L, Wacha A, Bóta A, Longstaff C, Kolev K. Citrullinated fibrinogen forms densely packed clots with decreased permeability. J Thromb Haemost 2022; 20:2862-2872. [PMID: 36083779 PMCID: PMC9828116 DOI: 10.1111/jth.15875] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/17/2022] [Accepted: 09/07/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Fibrin, the main scaffold of thrombi, is susceptible to citrullination by PAD (peptidyl arginine deiminase) 4, secreted from neutrophils during the formation of neutrophil extracellular traps. Citrullinated fibrinogen (citFg) has been detected in human plasma as well as in murine venous thrombi, and it decreases the lysability and mechanical resistance of fibrin clots. OBJECTIVE To investigate the effect of fibrinogen citrullination on the structure of fibrin clots. METHODS Fibrinogen was citrullinated with PAD4 and clotted with thrombin. Scanning electron microscopy (SEM) and atomic force microscopy (AFM) were used to measure fiber thickness, fiber height/width ratio, and fiber persistence length in clots containing citFg. Fiber density was measured with laser scanning microscopy (LSM) and permeability measurements were carried out to estimate the porosity of the clots. The intra-fiber structure of fibrin was analyzed with small-angle X-ray scattering (SAXS). RESULTS SEM images revealed a decrease in the median fiber diameter that correlated with the fraction of citFg in the clot, while the fiber width/length ratio remained unchanged according to AFM. With SAXS we observed that citrullination resulted in the formation of denser clots in line with increased fiber density shown by LSM. The permeability constant of citrullinated fibrin decreased more than 3-fold indicating significantly decreased porosity. SAXS also showed largely preserved periodicity in the longitudinal assembly of fibrin monomers. CONCLUSION The current observations of thin fibers combined with dense packing and low porosity in the presence of citFg can provide a structural framework for the mechanical fragility and lytic resistance of citrullinated fibrin.
Collapse
Affiliation(s)
- Imre Varjú
- Program in Cellular and Molecular MedicineBoston Children's HospitalBostonMassachusettsUSA
- Department of PediatricsHarvard Medical SchoolBostonMassachusettsUSA
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
- Department of Sociomedical Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Erzsébet Tóth
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Ádám Z. Farkas
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Veronika J. Farkas
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Erzsébet Komorowicz
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Tímea Feller
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Balázs Kiss
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | | | - László Szabó
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
- Department of Functional and Structural Materials, Institute of Materials and Environmental Chemistry, Research Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - András Wacha
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - Attila Bóta
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - Colin Longstaff
- National Institute for Biological Standards and ControlSouth MimmsUK
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| |
Collapse
|
3
|
Patterson EK, Fraser DD, Cepinskas G. Neutrophil serine proteases in vitro: How much and why? J Leukoc Biol 2022; 112:585-586. [PMID: 36073346 DOI: 10.1002/jlb.3lt0722-360r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Eric K Patterson
- Centre for Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Douglas D Fraser
- Centre for Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada.,Department of Pediatrics, Western University, London, Ontario, Canada.,Department of Physiology & Pharmacology, Western University, London, Ontario, Canada.,Department of Clinical Neurological Sciences, Western University, London, Ontario, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Gediminas Cepinskas
- Centre for Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
4
|
Cruz MA, Bohinc D, Andraska EA, Alvikas J, Raghunathan S, Masters NA, van Kleef ND, Bane KL, Hart K, Medrow K, Sun M, Liu H, Haldeman S, Banerjee A, Lessieur EM, Hageman K, Gandhi A, de la Fuente M, Nieman MT, Kern TS, Maas C, de Maat S, Neeves KB, Neal MD, Sen Gupta A, Stavrou EX. Nanomedicine platform for targeting activated neutrophils and neutrophil-platelet complexes using an α 1-antitrypsin-derived peptide motif. NATURE NANOTECHNOLOGY 2022; 17:1004-1014. [PMID: 35851383 PMCID: PMC9909445 DOI: 10.1038/s41565-022-01161-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2022] [Indexed: 05/30/2023]
Abstract
Targeted drug delivery to disease-associated activated neutrophils can provide novel therapeutic opportunities while avoiding systemic effects on immune functions. We created a nanomedicine platform that uniquely utilizes an α1-antitrypsin-derived peptide to confer binding specificity to neutrophil elastase on activated neutrophils. Surface decoration with this peptide enabled specific anchorage of nanoparticles to activated neutrophils and platelet-neutrophil aggregates, in vitro and in vivo. Nanoparticle delivery of a model drug, hydroxychloroquine, demonstrated significant reduction of neutrophil activities in vitro and a therapeutic effect on murine venous thrombosis in vivo. This innovative approach of cell-specific and activation-state-specific targeting can be applied to several neutrophil-driven pathologies.
Collapse
Affiliation(s)
- Michelle A Cruz
- Department of Pathology, Immunology Training Program, CWRU School of Medicine, Cleveland, OH, USA
| | - Dillon Bohinc
- Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA
| | - Elizabeth A Andraska
- Department of Surgery, Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jurgis Alvikas
- Department of Surgery, Pittsburgh Trauma Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Shruti Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Nicole A Masters
- Department of Civil and Environmental Engineering, Colorado School of Mines, Golden, CO, USA
| | - Nadine D van Kleef
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Kara L Bane
- Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA
| | - Kathryn Hart
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Kathryn Medrow
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Michael Sun
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Haitao Liu
- Department of Ophthalmology, Children's Hospital of University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shannon Haldeman
- Department of Surgery, Pittsburgh Trauma Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ankush Banerjee
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Emma M Lessieur
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
| | - Kara Hageman
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA
| | - Agharnan Gandhi
- Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA
| | | | - Marvin T Nieman
- Department of Pharmacology, CWRU School of Medicine, Cleveland, OH, USA
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
- Veterans Administration Medical Center Research Service, Long Beach, CA, USA
| | - Coen Maas
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Steven de Maat
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Keith B Neeves
- Department of Bioengineering and Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant Hemophilia and Thrombosis Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Neal
- Department of Surgery, Pittsburgh Trauma Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Anirban Sen Gupta
- Department of Pathology, Immunology Training Program, CWRU School of Medicine, Cleveland, OH, USA.
- Department of Biomedical Engineering, Case Western Reserve University (CWRU), Cleveland, OH, USA.
- Department of Pharmacology, CWRU School of Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Evi X Stavrou
- Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
- Department of Medicine, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH, USA.
| |
Collapse
|
5
|
Lopes BRP, da Silva GS, de Lima Menezes G, de Oliveira J, Watanabe ASA, Porto BN, da Silva RA, Toledo KA. Serine proteases in neutrophil extracellular traps exhibit anti-Respiratory Syncytial Virus activity. Int Immunopharmacol 2022; 106:108573. [PMID: 35183035 DOI: 10.1016/j.intimp.2022.108573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 11/24/2022]
Abstract
Human respiratory syncytial virus (hRSV) is an infectious agent in infants and young children which there are no vaccines or drugs for treatment. Neutrophils are recruited for airway, where they are stimulated by hRSV to release large amounts of neutrophil extracellular traps (NETs). NETs are compound by DNA and proteins, including microbicidal enzymes. They constitute a large part of the mucus accumulated in the lung of patients, compromising their breathing capacity. In contrast, NETs can capture/inactivate hRSV, but the molecules responsible for this effect are unknown. OBJECTIVES We selected microbicidal NET enzymes (elastase, myeloperoxidase, cathepsin-G, and proteinase-3) to assess their anti-hRSV role. METHODS AND RESULTS Through in vitro assays using HEp-2 cells, we observed that elastase, proteinase-3, and cathepsin-G, but not myeloperoxidase, showed virucidal effects even at non-cytotoxic concentrations. Elastase and proteinase-3, but not cathepsin-G, cleaved viral F-protein, which is responsible for viral adhesion and fusion with the target cells. Molecular docking analysis indicated the interaction of these macromolecules in the antigenic regions of F-protein through the active regions of the enzymes. CONCLUSIONS Serine proteases from NETs interact and inactive hRSV. These results contribute to the understanding the role of NETs in hRSV infection and to designing treatment strategies for the inflammatory process during respiratory infections.
Collapse
Affiliation(s)
- Bruno Rafael Pereira Lopes
- São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences, São José do Rio Preto - SP, Brazil
| | - Gabriel Soares da Silva
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, Brazil
| | - Gabriela de Lima Menezes
- Biosystems Collaborative Nucleus, Institute of Exact Sciences, Federal University of Jatai, Jatai-GO, Brazil
| | - Juliana de Oliveira
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, Brazil; Graduate Program in Applied and Computational Mathematics - PGMAC - State University of Londrina, Londrina-PR, Brazil
| | - Aripuanã Sakurada Aranha Watanabe
- Virology Laboratory, Center for Microbiology Studies, Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Bárbara Nery Porto
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Roosevelt Alves da Silva
- Biosystems Collaborative Nucleus, Institute of Exact Sciences, Federal University of Jatai, Jatai-GO, Brazil
| | - Karina Alves Toledo
- São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences, São José do Rio Preto - SP, Brazil; São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, Brazil.
| |
Collapse
|
6
|
Patterson EK, Gillio-Meina C, Martin CM, Fraser DD, Van Nynatten LR, Slessarev M, Cepinskas G. Proteinase 3 contributes to endothelial dysfunction in an experimental model of sepsis. Exp Biol Med (Maywood) 2021; 246:2338-2345. [PMID: 34292081 DOI: 10.1177/15353702211029284] [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] [Indexed: 11/15/2022] Open
Abstract
In sepsis-induced inflammation, polymorphonuclear neutrophils (PMNs) contribute to vascular dysfunction. The serine proteases proteinase 3 (PR3) and human leukocyte elastase (HLE) are abundant in PMNs and are released upon degranulation. While HLE's role in inflammation-induced endothelial dysfunction is well studied, PR3's role is largely uninvestigated. We hypothesized that PR3, similarly to HLE, contributes to vascular barrier dysfunction in sepsis. Plasma PR3 and HLE concentrations and their leukocyte mRNA levels were measured by ELISA and qPCR, respectively, in sepsis patients and controls. Exogenous PR3 or HLE was applied to human umbilical vein endothelial cells (HUVECs) and HUVEC dysfunction was assessed by FITC-dextran permeability and electrical resistance. Both PR3 and HLE protein and mRNA levels were significantly increased in sepsis patients (P < 0.0001 and P < 0.05, respectively). Additionally, each enzyme independently increased HUVEC monolayer FITC-dextran permeability (P < 0.01), and decreased electrical resistance in a time- and dose-dependent manner (P < 0.001), an effect that could be ameliorated by novel treatment with carbon monoxide-releasing molecule 3 (CORM-3). The serine protease PR3, in addition to HLE, lead to vascular dysfunction and increased endothelial permeability, a hallmark pathological consequence of sepsis-induced inflammation. CORMs may offer a new strategy to reduce serine protease-induced vascular dysfunction.
Collapse
Affiliation(s)
- Eric K Patterson
- Centre for Critical Illness Research, 151158Lawson Health Research Institute, Lawson Health Research Institute, London, N6A 5W9, Canada
| | - Carolina Gillio-Meina
- Children's Health Research Institute and Translational Research Centre, Lawson Health Research Institute, London, N6A 5W9, Canada
| | - Claudio M Martin
- Centre for Critical Illness Research, 151158Lawson Health Research Institute, Lawson Health Research Institute, London, N6A 5W9, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Douglas D Fraser
- Centre for Critical Illness Research, 151158Lawson Health Research Institute, Lawson Health Research Institute, London, N6A 5W9, Canada.,Children's Health Research Institute and Translational Research Centre, Lawson Health Research Institute, London, N6A 5W9, Canada.,Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Logan R Van Nynatten
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Marat Slessarev
- Centre for Critical Illness Research, 151158Lawson Health Research Institute, Lawson Health Research Institute, London, N6A 5W9, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Gediminas Cepinskas
- Centre for Critical Illness Research, 151158Lawson Health Research Institute, Lawson Health Research Institute, London, N6A 5W9, Canada.,Department of Medical Biophysics, Western University, London, N6A 5C1, Canada
| |
Collapse
|
7
|
Patterson EK, Vanin Moreno N, Fraser DD, Cepinskas G, Iida T, Berard RA. A Proteinase 3 Contribution to Juvenile Idiopathic Arthritis-Associated Cartilage Damage. PATHOPHYSIOLOGY 2021; 28:320-327. [PMID: 35366277 PMCID: PMC8830470 DOI: 10.3390/pathophysiology28030021] [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: 05/03/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
A full understanding of the molecular mechanisms implicated in the etiopathogenesis of juvenile idiopathic arthritis (JIA) is lacking. A critical role for leukocyte proteolytic activity (e.g., elastase and cathepsin G) has been proposed. While leukocyte elastase’s (HLE) role has been documented, the potential contribution of proteinase 3 (PR3), a serine protease present in abundance in neutrophils, has not been evaluated. In this study we investigated: (1) PR3 concentrations in the synovial fluid of JIA patients using ELISA and (2) the cartilage degradation potential of PR3 by measuring the hydrolysis of fluorescently labeled collagen II in vitro. In parallel, concentrations and collagen II hydrolysis by HLE were assessed. Additionally, the levels of the co-secreted primary granule protein myeloperoxidase (MPO) were assessed in synovial fluid of patients diagnosed with JIA. We report the following levels of analytes in JIA synovial fluid: PR3—114 ± 100 ng/mL (mean ± SD), HLE—1272 ± 1219 ng/mL, and MPO—1129 ± 1659 ng/mL, with a very strong correlation between the PR3 and HLE concentrations (rs = 0.898, p < 1 × 10–6). Importantly, PR3 hydrolyzed fluorescently labeled collagen II as efficiently as HLE. Taken together, these novel findings suggest that PR3 (in addition to HLE) contributes to JIA-associated joint damage.
Collapse
Affiliation(s)
- Eric K. Patterson
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada; (E.K.P.); (N.V.M.); (G.C.); (T.I.)
| | - Nicolas Vanin Moreno
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada; (E.K.P.); (N.V.M.); (G.C.); (T.I.)
| | - Douglas D. Fraser
- Lawson Health Research Institute, Children’s Health Research Institute, London, ON N6A 5W9, Canada;
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Gediminas Cepinskas
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada; (E.K.P.); (N.V.M.); (G.C.); (T.I.)
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada
| | - Takaya Iida
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada; (E.K.P.); (N.V.M.); (G.C.); (T.I.)
| | - Roberta A. Berard
- Lawson Health Research Institute, Children’s Health Research Institute, London, ON N6A 5W9, Canada;
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Division of Rheumatology, London Health Sciences Centre, Children’s Hospital, London, ON N6A 5W9, Canada
- Correspondence:
| |
Collapse
|
8
|
Lee J, Lee D, Lawler S, Kim Y. Role of neutrophil extracellular traps in regulation of lung cancer invasion and metastasis: Structural insights from a computational model. PLoS Comput Biol 2021; 17:e1008257. [PMID: 33596197 PMCID: PMC7920364 DOI: 10.1371/journal.pcbi.1008257] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/01/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related deaths worldwide and is characterized by hijacking immune system for active growth and aggressive metastasis. Neutrophils, which in their original form should establish immune activities to the tumor as a first line of defense, are undermined by tumor cells to promote tumor invasion in several ways. In this study, we investigate the mutual interactions between the tumor cells and the neutrophils that facilitate tumor invasion by developing a mathematical model that involves taxis-reaction-diffusion equations for the critical components in the interaction. These include the densities of tumor and neutrophils, and the concentrations of signaling molecules and structure such as neutrophil extracellular traps (NETs). We apply the mathematical model to a Boyden invasion assay used in the experiments to demonstrate that the tumor-associated neutrophils can enhance tumor cell invasion by secreting the neutrophil elastase. We show that the model can both reproduce the major experimental observation on NET-mediated cancer invasion and make several important predictions to guide future experiments with the goal of the development of new anti-tumor strategies. Moreover, using this model, we investigate the fundamental mechanism of NET-mediated invasion of cancer cells and the impact of internal and external heterogeneity on the migration patterning of tumour cells and their response to different treatment schedules.
Collapse
Affiliation(s)
- Junho Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Sean Lawler
- Department of neurosurgery, Brigham and Women’s Hospital & Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
9
|
Synthesis and evaluation of benzenesulfonic acid derivatives as human neutrophil elastase (hNE) inhibitors. Med Chem Res 2021; 30:387-398. [PMID: 33456292 PMCID: PMC7801566 DOI: 10.1007/s00044-020-02684-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/02/2020] [Indexed: 01/16/2023]
Abstract
Herein we report our investigation concerning the development of Human neutrophil elastase (hNE) inhibitors for the treatment of Acute Respiratory Distress Syndrome (ARDS). Various benzenesulfonic acid derived compounds were synthesized and evaluated as competitive inhibitors of hNE. Biological screening revealed that compound 4f shows moderate inhibitory activity (IC50 = 35.2 μM) against hNE. Compound 4f was also superimposed onto the active center of hNE to understand the binding mode.
Collapse
|
10
|
Alpha-1 Antitrypsin-A Target for MicroRNA-Based Therapeutic Development for Cystic Fibrosis. Int J Mol Sci 2020; 21:ijms21030836. [PMID: 32012925 PMCID: PMC7037267 DOI: 10.3390/ijms21030836] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder arising from mutations to the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Disruption to normal ion homeostasis in the airway results in impaired mucociliary clearance, leaving the lung more vulnerable to recurrent and chronic bacterial infections. The CF lung endures an excess of neutrophilic inflammation, and whilst neutrophil serine proteases are a crucial part of the innate host defence to infection, a surplus of neutrophil elastase (NE) is understood to create a net destructive effect. Alpha-1 antitrypsin (A1AT) is a key antiprotease in the control of NE protease activity but is ineffective in the CF lung due to the huge imbalance of NE levels. Therapeutic strategies to boost levels of protective antiproteases such as A1AT in the lung remain an attractive research strategy to limit the damage from excess protease activity. microRNAs are small non-coding RNA molecules that bind specific cognate sequences to inhibit expression of target mRNAs. The inhibition of miRNAs which target the SERPINA1 (A1AT-encoding gene) mRNA represents a novel therapeutic approach for CF inflammation. This could involve the delivery of antagomirs that bind and sequester the target miRNA, or target site blockers that bind miRNA recognition elements within the target mRNA to prevent miRNA interaction. Therefore, miRNA targeted therapies offer an alternative strategy to drive endogenous A1AT production and thus supplement the antiprotease shield of the CF lung.
Collapse
|
11
|
Networks that stop the flow: A fresh look at fibrin and neutrophil extracellular traps. Thromb Res 2019; 182:1-11. [DOI: 10.1016/j.thromres.2019.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/18/2019] [Accepted: 08/05/2019] [Indexed: 12/23/2022]
|
12
|
Crocetti L, Quinn MT, Schepetkin IA, Giovannoni MP. A patenting perspective on human neutrophil elastase (HNE) inhibitors (2014-2018) and their therapeutic applications. Expert Opin Ther Pat 2019; 29:555-578. [PMID: 31204543 PMCID: PMC9642779 DOI: 10.1080/13543776.2019.1630379] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/07/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Human neutrophil elastase (HNE) is involved in a variety of serious chronic diseases, especially cardiopulmonary pathologies. For this reason, the regulation of HNE activity represents a promising therapeutic approach, which is evident by the development of a number of new and selective HNE inhibitors, both in the academic and pharmaceutical environments. AREAS COVERED The present review analyzes and summarizes the patent literature regarding human neutrophil elastase inhibitors for the treatment of cardiopulmonary diseases over 2014-2018. EXPERT OPINION HNE is an interesting and defined target to treat various inflammatory diseases, including a number of cardiopulmonary pathologies. The research in this field is quite active, and a number of HNE inhibitors are currently in various stages of clinical development. In addition, new opportunities for HNE inhibitor development stem from recent studies demonstrating the involvement of HNE in many other inflammatory pathologies, including rheumatoid arthritis, inflammatory bowel disease, skin diseases, and cancer. Furthermore, the development of dual HNE/proteinase 3 inhibitors is being pursued as an innovative approach for the treatment of neutrophilic inflammatory diseases. Thus, these new developments will likely stimulate new and increased interest in this important therapeutic target and for the development of novel and selective HNE inhibitors.
Collapse
Affiliation(s)
- L Crocetti
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - MT Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - IA Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - MP Giovannoni
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Stockley RA, Halpin DMG, Celli BR, Singh D. Chronic Obstructive Pulmonary Disease Biomarkers and Their Interpretation. Am J Respir Crit Care Med 2019; 199:1195-1204. [DOI: 10.1164/rccm.201810-1860so] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Robert A. Stockley
- Lung Investigation Unit, Medicine, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - David M. G. Halpin
- Department of Respiratory Medicine, Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Bartolome R. Celli
- Pulmonary and Critical Care Department, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Hospital Trust, Manchester, United Kingdom
| |
Collapse
|
14
|
Jasper AE, McIver WJ, Sapey E, Walton GM. Understanding the role of neutrophils in chronic inflammatory airway disease. F1000Res 2019; 8. [PMID: 31069060 PMCID: PMC6489989 DOI: 10.12688/f1000research.18411.1] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2019] [Indexed: 12/28/2022] Open
Abstract
Airway neutrophilia is a common feature of many chronic inflammatory lung diseases and is associated with disease progression, often regardless of the initiating cause. Neutrophils and their products are thought to be key mediators of the inflammatory changes in the airways of patients with chronic obstructive pulmonary disease (COPD) and have been shown to cause many of the pathological features associated with disease, including emphysema and mucus hypersecretion. Patients with COPD also have high rates of bacterial colonisation and recurrent infective exacerbations, suggesting that neutrophil host defence mechanisms are impaired, a concept supported by studies showing alterations to neutrophil migration, degranulation and reactive oxygen species production in cells isolated from patients with COPD. Although the role of neutrophils is best described in COPD, many of the pathological features of this disease are not unique to COPD and also feature in other chronic inflammatory airway diseases, including asthma, cystic fibrosis, alpha-1 anti-trypsin deficiency, and bronchiectasis. There is increasing evidence for immune cell dysfunction contributing to inflammation in many of these diseases, focusing interest on the neutrophil as a key driver of pulmonary inflammation and a potential therapeutic target than spans diseases. This review discusses the evidence for neutrophilic involvement in COPD and also considers their roles in alpha-1 anti-trypsin deficiency, bronchiectasis, asthma, and cystic fibrosis. We provide an in-depth assessment of the role of the neutrophil in each of these conditions, exploring recent advances in understanding, and finally discussing the possibility of common mechanisms across diseases.
Collapse
Affiliation(s)
- Alice E Jasper
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - William J McIver
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Georgia M Walton
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| |
Collapse
|
15
|
Mao M, Hao L, Wang Y, Liu QQ. Transplantation of Endothelial Progenitor Cells Attenuates Lipopolysaccharide-Induced Lung Injury via Inhibiting the Inflammatory Secretion of Neutrophils in Rats. Am J Med Sci 2018; 357:49-56. [PMID: 30611320 DOI: 10.1016/j.amjms.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are special types of stem cells and are a potential novel therapeutic approach in acute lung injury (ALI). Transplantation of EPCs can ameliorate the inflammatory state by reducing adhesion and exudation of inflammatory cells. However, the mechanism underlying the effect of EPCs on inflammatory response modulation remains unclear. The aim of the present study was to investigate the effect of EPCs on the modulation of neutrophils in vitro and in vivo. MATERIALS AND METHODS EPCs were cocultured with neutrophils after lipopolysaccharide stimulation in vitro or transplanted into ALI rats, and neutrophil inflammatory mediators including tumor necrosis factor-α, interleukin-1β, neutrophil elastase, myeloperoxidase and matrix metalloproteinases-9 were detected by enzyme-linked immunosorbent assay, an myeloperoxidase detection kits, reverse transcription-polymerase chain reaction and western blotting. RESULTS The results showed that EPCs significantly downregulated the expression of inflammatory mediators when cocultured with neutrophils in vitro or in vivo. CONCLUSIONS These findings demonstrated that EPCs contributed to lung injury in ALI rats by downregulating neutrophil inflammatory mediators.
Collapse
Affiliation(s)
- Mei Mao
- Department of Geriatrics, No 958 Hospital of PLA, Chongqing, China.
| | - Lei Hao
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Yi Wang
- Department of Respiratory Medicine, the Sixth People's Hospital of Ji'nan City Affiliated to Jining Medical College, Jinan, China
| | - Qiu-Qian Liu
- Department of Infection Prevention and Control, No.958 Hospital of PLA, Chongqing, China
| |
Collapse
|
16
|
Attia Z, Rowe JC, Kim E, Varikuti S, Steiner HE, Zaghawa A, Hassan H, Cormet-Boyaka E, Satoskar AR, Boyaka PN. Inhibitors of elastase stimulate murine B lymphocyte differentiation into IgG- and IgA-producing cells. Eur J Immunol 2018; 48:1295-1301. [PMID: 29710424 DOI: 10.1002/eji.201747264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/17/2018] [Accepted: 04/25/2018] [Indexed: 01/11/2023]
Abstract
It is well established that dendritic cells and macrophages play a role in antigen presentation to B and T cells and in shaping B and T cell responses via cytokines they produce. We have previously reported that depletion of neutrophils improves the production of mucosal IgA after sublingual immunization with Bacillus anthracis edema toxin as adjuvant. These past studies also demonstrated that an inverse correlation exists between the number of neutrophils and production of IgA by B cells. Using specific inhibitors of elastase, we addressed whether the elastase activity of neutrophil could be the factor that interferes with production of IgA and possibly other immunoglobulin isotypes. We found that murine splenocytes and mesenteric lymph node cells cultured for 5 days in the presence of neutrophil elastase inhibitors secreted higher levels of IgG and IgA than cells cultured in the absence of inhibitors. The effect of the inhibitors was dose-dependent and was consistent with increased frequency of CD138+ cells expressing IgG or IgA. Finally, neutrophil elastase inhibitors increased transcription of mRNA for AID, IL-10, BAFF and APRIL, factors involved in B cell differentiation. These findings identify inhibitors of elastase as potential adjuvants for increasing production of antibodies.
Collapse
Affiliation(s)
- Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.,Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | - John C Rowe
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Haley E Steiner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Ahmad Zaghawa
- Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | - Hany Hassan
- Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | | | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Kuznetsova I, Arnold T, Aschacher T, Schwager C, Hegedus B, Garay T, Stukova M, Pisareva M, Pleschka S, Bergmann M, Egorov A. Targeting an Oncolytic Influenza A Virus to Tumor Tissue by Elastase. MOLECULAR THERAPY-ONCOLYTICS 2017; 7:37-44. [PMID: 29034314 PMCID: PMC5633860 DOI: 10.1016/j.omto.2017.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/01/2017] [Indexed: 11/24/2022]
Abstract
Oncolytic viruses are currently established as a novel type of immunotherapy. The challenge is to safely target oncolytic viruses to tumors. Previously, we have generated influenza A viruses (IAVs) containing deletions in the viral interferon antagonist. Those deletions have attenuated the virus in normal tissue but allowed replication in tumor cells. IAV entry is mediated by hemagglutinin (HA), which needs to be activated by a serine protease, for example, through trypsin. To further target the IAV to tumors, we have changed the trypsin cleavage site to an elastase cleavage site. We chose this cleavage site because elastase is expressed in the tumor microenvironment. Moreover, the exchange of the cleavage site previously has been shown to attenuate viral growth in lungs. Newly generated elastase-activated influenza viruses (AE viruses) grew to similar titers in tumor cells as the trypsin-activated counterparts (AT viruses). Intratumoral injection of AE viruses into syngeneic B16f1 melanoma-derived tumors in mice reduced tumor growth similar to AT viruses and had a better therapeutic effect in heterologous human PANC-1-derived tumors. Therefore, the introduction of the attenuation marker “elastase cleavage site” in viral HA allows for safe, effective oncolytic virus therapy.
Collapse
Affiliation(s)
- Irina Kuznetsova
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.,Institute for Medical Virology, Justus Liebig University Gießen, School of Medicine, Schubertstraße 81, 35392 Gießen, Germany
| | - Tobias Arnold
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Thomas Aschacher
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Cornelia Schwager
- Avir Green Hills Biotechnology AG, Gersthoferstrasse 29, 1180 Vienna, Austria
| | - Balazs Hegedus
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.,MTA-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Semmelweis University, Üllői út 93, 1091 Budapest, Hungary
| | - Tamas Garay
- MTA-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Semmelweis University, Üllői út 93, 1091 Budapest, Hungary
| | - Marina Stukova
- Research Institute of Influenza, Russian Academy of Medical Sciences, Prof. Popova Str. 15/17, 196376 St. Petersburg, Russia
| | - Maria Pisareva
- Research Institute of Influenza, Russian Academy of Medical Sciences, Prof. Popova Str. 15/17, 196376 St. Petersburg, Russia
| | - Stephan Pleschka
- Institute for Medical Virology, Justus Liebig University Gießen, School of Medicine, Schubertstraße 81, 35392 Gießen, Germany
| | - Michael Bergmann
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.,Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Andrej Egorov
- Research Institute of Influenza, Russian Academy of Medical Sciences, Prof. Popova Str. 15/17, 196376 St. Petersburg, Russia
| |
Collapse
|
18
|
Elevated Leukocyte Azurophilic Enzymes in Human Diabetic Ketoacidosis Plasma Degrade Cerebrovascular Endothelial Junctional Proteins. Crit Care Med 2017; 44:e846-53. [PMID: 27071071 DOI: 10.1097/ccm.0000000000001720] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Diabetic ketoacidosis in children is associated with vasogenic cerebral edema, possibly due to the release of destructive polymorphonuclear neutrophil azurophilic enzymes. Our objectives were to measure plasma azurophilic enzyme levels in children with diabetic ketoacidosis, to correlate plasma azurophilic enzyme levels with diabetic ketoacidosis severity, and to determine whether azurophilic enzymes disrupt the blood-brain barrier in vitro. DESIGN Prospective clinical and laboratory study. SETTING The Children's Hospital, London Health Sciences Centre. SUBJECTS Pediatric type 1 diabetes patients; acute diabetic ketoacidosis or age-/sex-matched insulin-controlled. MEASUREMENTS AND MAIN RESULTS Acute diabetic ketoacidosis in children was associated with elevated polymorphonuclear neutrophils. Plasma azurophilic enzymes were elevated in diabetic ketoacidosis patients, including human leukocyte elastase (p < 0.001), proteinase-3 (p < 0.01), and myeloperoxidase (p < 0.001). A leukocyte origin of human leukocyte elastase and proteinase-3 in diabetic ketoacidosis was confirmed with buffy coat quantitative real-time polymerase chain reaction (p < 0.01). Of the three azurophilic enzymes elevated, only proteinase-3 levels correlated with diabetic ketoacidosis severity (p = 0.002). Recombinant proteinase-3 applied to human brain microvascular endothelial cells degraded both the tight junction protein occludin (p < 0.05) and the adherens junction protein VE-cadherin (p < 0.05). Permeability of human brain microvascular endothelial cell monolayers was increased by recombinant proteinase-3 application (p = 0.010). CONCLUSIONS Our results indicate that diabetic ketoacidosis is associated with systemic polymorphonuclear neutrophil activation and degranulation. Of all the polymorphonuclear neutrophil azurophilic enzymes examined, only proteinase-3 correlated with diabetic ketoacidosis severity and potently degraded the blood-brain barrier in vitro. Proteinase-3 might mediate vasogenic edema during diabetic ketoacidosis, and selective proteinase-3 antagonists may offer future vascular- and neuroprotection.
Collapse
|
19
|
Pechous RD. With Friends Like These: The Complex Role of Neutrophils in the Progression of Severe Pneumonia. Front Cell Infect Microbiol 2017; 7:160. [PMID: 28507954 PMCID: PMC5410563 DOI: 10.3389/fcimb.2017.00160] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/12/2017] [Indexed: 01/12/2023] Open
Abstract
Pneumonia is a leading cause of death from infection in the United States and across the globe. During pulmonary infection, clear resolution of host inflammatory responses occurs in the absence of appreciable lung damage. Neutrophils are the first wave of leukocytes to arrive in the lung upon infection. After activation, neutrophils traffic from the vasculature via transendothelial migration through the lung interstitium and into the alveolar space. Successful pulmonary immunity requires neutrophil-mediated killing of invading pathogens by phagocytosis and release of a myriad of antimicrobial molecules, followed by resolution of inflammation, neutrophil apoptosis, and clearing of dead or dying neutrophils by macrophages. In addition to their antimicrobial role, it is becoming clear that neutrophils are also important modulators of innate and adaptive immune responses, primarily through the release of cytokines and recruitment of additional waves of neutrophils into the airways. Though typically essential to combating severe pneumonia, neutrophil influx into the airways is a double-edged sword: Overzealous neutrophil activation can cause severe tissue damage as a result of the release of toxic agents including proteases, cationic polypeptides, cytokines, and reactive oxygen species (ROS) aimed at killing invading microbes. In extreme cases, the damage caused by neutrophils and other innate immune mediators become the primary source of morbidity and mortality. Here, we review the complex role of neutrophils during severe pneumonia by highlighting specific molecules and processes that contribute to pulmonary immunity, but can also drive progression of severe disease. Depending on the identity of the infectious agent, enhancing or suppressing neutrophil-mediated responses may be key to effectively treating severe and typically lethal pneumonia.
Collapse
Affiliation(s)
- Roger D Pechous
- Department of Microbiology and Immunology, University of Arkansas for Medical SciencesLittle Rock, AR, USA
| |
Collapse
|
20
|
McGuinness AJA, Sapey E. Oxidative Stress in COPD: Sources, Markers, and Potential Mechanisms. J Clin Med 2017; 6:jcm6020021. [PMID: 28212273 PMCID: PMC5332925 DOI: 10.3390/jcm6020021] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/01/2017] [Accepted: 02/05/2017] [Indexed: 12/13/2022] Open
Abstract
Markers of oxidative stress are increased in chronic obstructive pulmonary disease (COPD) and reactive oxygen species (ROS) are able to alter biological molecules, signaling pathways and antioxidant molecule function, many of which have been implicated in the pathogenesis of COPD. However, the involvement of ROS in the development and progression of COPD is not proven. Here, we discuss the sources of ROS, and the defences that have evolved to protect against their harmful effects. We address the role that ROS may have in the development and progression of COPD, as well as current therapeutic attempts at limiting the damage they cause. Evidence has indicated that the function of several key cells appears altered in COPD patients, and expression levels of important oxidant and antioxidant molecules may be abnormal. Therapeutic trials attempting to restore equilibrium to these molecules have not impacted upon all facets of disease and whilst the theory behind ROS influence in COPD appears sound, current models testing relevant pathways to tissue damage are limited. The heterogeneity seen in COPD patients presents a challenge to our understanding, and further research is essential to identify potential targets and stratified COPD patient populations where ROS therapies may be maximally efficacious.
Collapse
Affiliation(s)
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
21
|
Elborn JS, Horsley A, MacGregor G, Bilton D, Grosswald R, Ahuja S, Springman EB. Phase I Studies of Acebilustat: Biomarker Response and Safety in Patients with Cystic Fibrosis. Clin Transl Sci 2016; 10:28-34. [PMID: 27806191 PMCID: PMC5351012 DOI: 10.1111/cts.12428] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022] Open
Abstract
There is a significant unmet need for safe and effective anti‐inflammatory treatment for cystic fibrosis. The aim of this study was to evaluate the safety of acebilustat, a leukotriene A4 hydrolase inhibitor, and its effect on inflammation biomarkers in patients with cystic fibrosis. Seventeen patients with mild to moderate cystic fibrosis were enrolled and randomized into groups receiving placebo or doses of 50 mg or 100 mg acebilustat administered orally, once daily for 15 days. Sputum neutrophil counts were reduced by 65% over baseline values in patients treated with 100 mg acebilustat. A modestly significant 58% reduction vs. placebo in sputum elastase was observed with acebilustat treatment. Favorable trends were observed for reduction of serum C‐reactive protein and sputum neutrophil DNA in acebilustat‐treated patients. No changes in pulmonary function were observed. Acebilustat was safe and well tolerated. The results of this study support further clinical development of acebilustat for treatment of cystic fibrosis.
Collapse
Affiliation(s)
- J S Elborn
- Centre for Infection and Immunity, Dentistry, and Biomedical Sciences, Queens University School of Medicine, Belfast, UK
| | - A Horsley
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - G MacGregor
- West of Scotland CF Centre, Gartnavel General Hospital, Glasgow, UK
| | - D Bilton
- Department of Respiratory Medicine, Royal Brompton Hospital, London, UK
| | | | - S Ahuja
- Celtaxsys, Inc, Atlanta, Georgia, USA
| | | |
Collapse
|
22
|
Boxio R, Wartelle J, Nawrocki-Raby B, Lagrange B, Malleret L, Hirche T, Taggart C, Pacheco Y, Devouassoux G, Bentaher A. Neutrophil elastase cleaves epithelial cadherin in acutely injured lung epithelium. Respir Res 2016; 17:129. [PMID: 27751187 PMCID: PMC5067913 DOI: 10.1186/s12931-016-0449-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/10/2016] [Indexed: 01/05/2023] Open
Abstract
Background In acutely injured lungs, massively recruited polymorphonuclear neutrophils (PMNs) secrete abnormally neutrophil elastase (NE). Active NE creates a localized proteolytic environment where various host molecules are degraded leading to impairment of tissue homeostasis. Among the hallmarks of neutrophil-rich pathologies is a disrupted epithelium characterized by the loss of cell-cell adhesion and integrity. Epithelial-cadherin (E-cad) represents one of the most important intercellular junction proteins. E-cad exhibits various functions including its role in maintenance of tissue integrity. While much interest has focused on the expression and role of E-cad in different physio- and physiopathological states, proteolytic degradation of this structural molecule and ensuing potential consequences on host lung tissue injury are not completely understood. Methods NE capacity to cleave E-cad was determined in cell-free and lung epithelial cell culture systems. The impact of such cleavage on epithelial monolayer integrity was then investigated. Using mice deficient in NE in a clinically relevant experimental model of acute pneumonia, we examined whether degraded E-cad is associated with lung inflammation and injury and whether NE contributes to E-cad cleavage. Finally, we checked for the presence of both degraded E-cad and NE in bronchoalveolar lavage samples obtained from patients with exacerbated COPD, a clinical manifestation characterised by a neutrophilic inflammatory response. Results We show that NE is capable of degrading E-cad in vitro and in cultured cells. NE-mediated degradation of E-cad was accompanied with loss of epithelial monolayer integrity. Our in vivo findings provide evidence that NE contributes to E-cad cleavage that is concomitant with lung inflammation and injury. Importantly, we observed that the presence of degraded E-cad coincided with the detection of NE in diseased human lungs. Conclusions Active NE has the capacity to cleave E-cad and interfere with its cell-cell adhesion function. These data suggest a mechanism by which unchecked NE participates potentially to the pathogenesis of neutrophil-rich lung inflammatory and tissue-destructive diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0449-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rachel Boxio
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France
| | - Julien Wartelle
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France
| | | | - Brice Lagrange
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France
| | - Laurette Malleret
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France
| | - Timothee Hirche
- Department of Pulmonary Medicine, German Clinic for Diagnostics (DKD), Wiesbaden, Germany
| | - Clifford Taggart
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Yves Pacheco
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France
| | - Gilles Devouassoux
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France.,CHU Croix-Rousse, Lyon, France
| | - Abderrazzaq Bentaher
- Inflammation and Immunity of the Respiratory Epithelium Group, Faculté de Médecine Lyon Sud, EA 7426, UCBL 1, Inserm U-1111, Pierre Benite - Lyon Sud, France.
| |
Collapse
|
23
|
Walton GM, Stockley JA, Griffiths D, Sadhra CS, Purvis T, Sapey E. Repurposing Treatments to Enhance Innate Immunity. Can Statins Improve Neutrophil Functions and Clinical Outcomes in COPD? J Clin Med 2016; 5:jcm5100089. [PMID: 27727158 PMCID: PMC5086591 DOI: 10.3390/jcm5100089] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/17/2016] [Accepted: 09/20/2016] [Indexed: 01/08/2023] Open
Abstract
Drug classes used in the treatment of Chronic Obstructive Pulmonary Disease (COPD) have not changed for many years, and none to date have shown disease-modifying activity. Statins are used to help reduce cardiovascular risk, which is high in many patients with COPD. Their use has been associated with improvements in some respiratory manifestations of disease and reduction in all-cause mortality, with greatest reductions seen in patients with the highest inflammatory burden. The mechanism for these effects is poorly understood. Neutrophils are key effector cells in COPD, and correlate with disease severity and inflammation. Recent in vitro studies have shown neutrophil functions are dysregulated in COPD and this is thought to contribute both to the destruction of lung parenchyma and to the poor responses seen in infective exacerbations. In this article, we will discuss the potential utility of statins in COPD, with a particular emphasis on their immune-modulatory effects as well as presenting new data regarding the effects of statins on neutrophil function in vitro.
Collapse
Affiliation(s)
- Georgia M Walton
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK.
| | - James A Stockley
- Lung Function and Sleep, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK.
| | - Diane Griffiths
- Respiratory Research, Research and Development, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK.
| | - Charandeep S Sadhra
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Thomas Purvis
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
24
|
von Nussbaum F, Li VMJ. Neutrophil elastase inhibitors for the treatment of (cardio)pulmonary diseases: Into clinical testing with pre-adaptive pharmacophores. Bioorg Med Chem Lett 2015; 25:4370-81. [PMID: 26358162 DOI: 10.1016/j.bmcl.2015.08.049] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 02/04/2023]
Abstract
Alpha-1 antitrypsin deficiency is linked with an increased risk of suffering from lung emphysema. This discovery from the 1960s led to the development of the protease-antiprotease (im)balance hypothesis: Overshooting protease concentrations, especially high levels of elastase were deemed to have an destructive effect on lung tissue. Consequently, it was postulated that efficient elastase inhibitors could alleviate the situation in patients. However, despite intensive drug discovery efforts, even five decades later, no neutrophil elastase inhibitors are available for a disease-modifying treatment of (cardio)pulmonary diseases such as chronic obstructive pulmonary disease. Here, we critically review the attempts to develop effective human neutrophil elastase inhibitors while strongly focussing on recent developments. On purpose and with perspective distortion we focus on recent developments. One aim of this review is to classify the known HNE inhibitors into several generations, according to their binding modes. In general, there seem to be three major challenges in the development of suitable elastase inhibitors: (1) assuring sufficient potency, (2) securing selectivity, and (3) achieving metabolic stability especially under pathophysiological conditions. Impressive achievements have been made since 2001 with the identification of potent nonreactive, reversible small molecule inhibitors. The most modern inhibitors bind HNE via an induced fit with a frozen bioactive conformation that leads to a significant boost in potency, selectivity, and stability ('pre-adaptive pharmacophores'). These 5th generation inhibitors might succeed in re-establishing the protease-antiprotease balance in patients for the first time.
Collapse
Affiliation(s)
| | - Volkhart M-J Li
- Bayer HealthCare AG, Lead Discovery Wuppertal, 42096 Wuppertal, Germany.
| |
Collapse
|
25
|
Tsai YF, Hwang TL. Neutrophil elastase inhibitors: a patent review and potential applications for inflammatory lung diseases (2010 - 2014). Expert Opin Ther Pat 2015; 25:1145-58. [PMID: 26118988 DOI: 10.1517/13543776.2015.1061998] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The proteolytic activity of neutrophil elastase (NE) not only destroys pathogens but also degrades host matrix tissues by generating a localized protease-antiprotease imbalance. In humans, NE is well known to be involved in various acute and chronic inflammatory diseases, such as chronic obstructive pulmonary disease, emphysema, asthma, acute lung injury, acute respiratory distress syndrome and cystic fibrosis. The regulation of NE activity is thought to represent a promising therapeutic approach, and NE is considered as an important target for the development of novel selective inhibitors to treat these diseases. AREAS COVERED This article summarizes and analyzes patents on NE inhibitors and their therapeutic potential based on a review of patent applications disclosed between 2010 and 2014. EXPERT OPINION According to this review of recent NE inhibitor patents, all of the disclosed inhibitors can be classified into peptide- and non-peptide-based groups. The non-peptide NE inhibitors include heterocyclics, uracil derivatives and deuterium oxide. Among the heterocyclic analogs, derivatives of pyrimidinones, tetrahydropyrrolopyrimidinediones, pyrazinones, benzoxazinones and hypersulfated disaccharides were introduced. The literature has increasingly implicated NE in the pathogenesis of various diseases, of which inflammatory destructive lung diseases remain a major concern. However, only a few agents have been validated for therapeutic use in clinical settings to date.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- a 1 Chang Gung University, Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine , Taoyuan 33302, Taiwan.,b 2 Chang Gung Memorial Hospital, Department of Anesthesiology , Kweishan, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- a 1 Chang Gung University, Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine , Taoyuan 33302, Taiwan.,c 3 Chang Gung University, Healthy Aging Research Center, Chinese Herbal Medicine Research Team , Taoyuan 33302, Taiwan.,d 4 Chang Gung University of Science and Technology, Department of Cosmetic Science and Research Center for Industry of Human Ecology , Taoyuan 33302, Taiwan.,e 5 Chang Gung University, Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine , Taoyuan 33302, Taiwan +88 6 3211 8506 ; +88 6 3211 8506 ;
| |
Collapse
|
26
|
Sinden NJ, Baker MJ, Smith DJ, Kreft JU, Dafforn TR, Stockley RA. α-1-antitrypsin variants and the proteinase/antiproteinase imbalance in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2015; 308:L179-90. [PMID: 25416382 DOI: 10.1152/ajplung.00179.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The excessive activities of the serine proteinases neutrophil elastase and proteinase 3 are associated with tissue damage in chronic obstructive pulmonary disease. Reduced concentrations and/or inhibitory efficiency of the main circulating serine proteinase inhibitor α-1-antitrypsin result from point mutations in its gene. In addition, α-2-macroglobulin competes with α-1-antitrypsin for proteinases, and the α-2-macroglobulin-sequestered enzyme can retain its catalytic activity. We have studied how serine proteinases partition between these inhibitors and the effects of α-1-antitrypsin mutations on this partitioning. Subsequently, we have developed a three-dimensional reaction-diffusion model to describe events occurring in the lung interstitium when serine proteinases diffuse from the neutrophil azurophil granule following degranulation and subsequently bind to either α-1-antitrypsin or α-2-macroglobulin. We found that the proteinases remained uninhibited on the order of 0.1 s after release and diffused on the order of 10 μm into the tissue before becoming sequestered. We have shown that proteinases sequestered to α-2-macroglobulin retain their proteolytic activity and that neutrophil elastase complexes with α-2-macroglobulin are able to degrade elastin. Although neutrophil elastase is implicated in the pathophysiology of emphysema, our results highlight a potentially important role for proteinase 3 because of its greater concentration in azurophil granules, its reduced association rate constant with all α-1-antitrypsin variants studied here, its greater diffusion distance, time spent uninhibited following degranulation, and its greater propensity to partition to α-2-macroglobulin where it retains proteolytic activity.
Collapse
|
27
|
Varjú I, Longstaff C, Szabó L, Farkas ÁZ, Varga-Szabó VJ, Tanka-Salamon A, Machovich R, Kolev K. DNA, histones and neutrophil extracellular traps exert anti-fibrinolytic effects in a plasma environment. Thromb Haemost 2015; 113:1289-98. [PMID: 25789443 DOI: 10.1160/th14-08-0669] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/21/2015] [Indexed: 12/22/2022]
Abstract
In response to various inflammatory stimuli, neutrophils secrete neutrophil extracellular traps (NETs), web-like meshworks of DNA, histones and granular components forming supplementary scaffolds in venous and arterial thrombi. Isolated DNA and histones are known to promote thrombus formation and render fibrin clots more resistant to mechanical forces and tissue-type plasminogen activator (tPA)-induced enzymatic digestion. The present study extends our earlier observations to a physiologically more relevant environment including plasma clots and NET-forming neutrophils. A range of techniques was employed including imaging (scanning electron microscopy (SEM), confocal laser microscopy, and photoscanning of macroscopic lysis fronts), clot permeability measurements, turbidimetric lysis and enzyme inactivation assays. Addition of DNA and histones increased the median fibre diameter of plasma clots formed with 16 nM thrombin from 108 to 121 and 119 nm, respectively, and decreased their permeability constant from 6.4 to 3.1 and 3.7×10(-9) cm(2). Histones effectively protected thrombin from antithrombin-induced inactivation, while DNA inhibited plasminogen activation on the surface of plasma clots and their plasmin-induced resolution by 20 and 40 %, respectively. DNA and histones, as well as NETs secreted by phorbol-myristate-acetate-activated neutrophils, slowed down the tPA-driven lysis of plasma clots and the latter effect could be reversed by the addition of DNase (streptodornase). SEM images taken after complete digestion of fibrin in NET-containing plasma clots evidenced retained NET scaffold that was absent in DNase-treated clots. Our results show that DNA and histones alter the fibrin architecture in plasma clots, while NETs contribute to a decreased lytic susceptibility that can be overcome by DNase.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Krasimir Kolev
- Krasimir Kolev, Semmelweis University, Department of Medical Biochemistry, Tűzoltó utca 37-47., 1094 Budapest, Hungary, Tel.: +36 1 4591500/60035, Fax: +36 1 2670031, E-mail:
| |
Collapse
|
28
|
Unopposed Cathepsin G, Neutrophil Elastase, and Proteinase 3 Cause Severe Lung Damage and Emphysema. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2197-210. [DOI: 10.1016/j.ajpath.2014.04.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 04/02/2014] [Accepted: 04/21/2014] [Indexed: 12/20/2022]
|
29
|
Benabid R, Wartelle J, Malleret L, Guyot N, Gangloff S, Lebargy F, Belaaouaj A. Neutrophil elastase modulates cytokine expression: contribution to host defense against Pseudomonas aeruginosa-induced pneumonia. J Biol Chem 2012; 287:34883-34894. [PMID: 22927440 DOI: 10.1074/jbc.m112.361352] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
There is accumulating evidence that following bacterial infection, the massive recruitment and activation of the phagocytes, neutrophils, is accompanied with the extracellular release of active neutrophil elastase (NE), a potent serine protease. Using NE-deficient mice in a clinically relevant model of Pseudomonas aeruginosa-induced pneumonia, we provide compelling in vivo evidence that the absence of NE was associated with decreased protein and transcript levels of the proinflammatory cytokines TNF-α, MIP-2, and IL-6 in the lungs, coinciding with increased mortality of mutant mice to infection. The implication of NE in the induction of cytokine expression involved at least in part Toll-like receptor 4 (TLR-4). These findings were further confirmed following exposure of cultured macrophages to purified NE. Together, our data suggest strongly for the first time that NE not only plays a direct antibacterial role as it has been previously reported, but released active enzyme can also modulate cytokine expression, which contributes to host protection against P. aeruginosa. In light of our findings, the long held view that considers NE as a prime suspect in P. aeruginosa-associated diseases will need to be carefully reassessed. Also, therapeutic strategies aiming at NE inhibition should take into account the physiologic roles of the enzyme.
Collapse
Affiliation(s)
- Rym Benabid
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - Julien Wartelle
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - Laurette Malleret
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - Nicolas Guyot
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - Sophie Gangloff
- EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - François Lebargy
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France
| | - Azzaq Belaaouaj
- Inserm Avenir Program, CHU de Reims, Maison Blanche, Reims 51092, France; EA 4303, Inflammation and Immunity of the Respiratory Epithelium, CHU de Reims, Maison Blanche, Reims 51092, France.
| |
Collapse
|
30
|
Wohner N, Kovács A, Machovich R, Kolev K. Modulation of the von Willebrand factor-dependent platelet adhesion through alternative proteolytic pathways. Thromb Res 2011; 129:e41-6. [PMID: 22178067 PMCID: PMC3323834 DOI: 10.1016/j.thromres.2011.11.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 11/10/2011] [Accepted: 11/14/2011] [Indexed: 12/31/2022]
Abstract
Introduction Platelet adhesion to collagen under high shear rates depends on the optimal size of the von Willebrand factor (VWF) multimers, which is determined by their limited proteolysis. The present study attempts to identify the role of hemostatic-fibrinolytic enzymes (thrombin, plasmin) and leukocyte-derived proteases (matrix metalloproteinase (MMP)-8, MMP-9, neutrophil elastase) in the cleavage of VWF and to characterize the effect of flow and platelets on this proteolysis and its functional consequences on platelet adhesion. Methods and results According to VWF immunoblots, plasmin, neutrophil elastase and thrombin at concentrations of in vivo relevance resulted in extensive degradation of VWF within several minutes. Platelets protected VWF against this proteolysis under static conditions, whereas perfusion of the proteases at 3350 s-1 shear rate over VWF immobilized on artery cross sections enhanced its degradation and blocked the protective effect of platelets. In parallel with VWF digestion, the examined proteases impaired the VWF-dependent platelet adhesion as reflected in the decreased surface-bound GpIIb/IIIa immunoreactivity following perfusion of collagen-coated surfaces or artery sections with blood and plasmin, neutrophil elastase or thrombin. Within the time frame of minutes no VWF cleavage could be detected under static or flow conditions after exposure to MMP-8 and MMP-9 at concentrations relevant to physiological neutrophil counts. Conclusion Our results indicate a shear- and platelet-dependent role for several proteases in the local modulation of the VWF function.
Collapse
Affiliation(s)
- Nikolett Wohner
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | | | | | | |
Collapse
|
31
|
Noninvasive in vivo quantification of neutrophil elastase activity in acute experimental mouse lung injury. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2011; 2011:581406. [PMID: 21941648 PMCID: PMC3175392 DOI: 10.1155/2011/581406] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/18/2011] [Indexed: 01/20/2023]
Abstract
We developed a neutrophil elastase-specific near-infrared fluorescence imaging agent, which, combined with fluorescence molecular tomographic imaging, allowed us to detect and quantify neutrophil elastase activity in vivo, in real time, and noninvasively in an acute model of lung injury (ALI). Significantly higher fluorescent signal was quantified in mice with LPS/fMLP-induced ALI as compared to healthy controls, correlating with increases in the number of bronchoalveolar lavage cells, neutrophils, and elastase activity. The agent was significantly activated ex vivo in lung sections from ALI but not from control mice, and this activation was ablated by the specific inhibitor sivelestat. Treatment with the specific inhibitor sivelestat significantly reduced lung signal in mice with ALI. These results underscore the unique ability of fluorescence molecular imaging to quantify specific molecular processes in vivo, crucial for understanding the mechanisms underlying disease progression and for assessing and monitoring novel pharmacological interventions.
Collapse
|
32
|
Stevens T, Ekholm K, Gränse M, Lindahl M, Kozma V, Jungar C, Ottosson T, Falk-Håkansson H, Churg A, Wright JL, Lal H, Sanfridson A. AZD9668: pharmacological characterization of a novel oral inhibitor of neutrophil elastase. J Pharmacol Exp Ther 2011; 339:313-20. [PMID: 21791628 DOI: 10.1124/jpet.111.182139] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
N-{[5-(methanesulfonyl)pyridin-2-yl]methyl}-6-methyl-5-(1-methyl-1H-pyrazol-5-yl)-2-oxo-1-[3-(trifluoromethyl)phenyl]-1,2-dihydropyridine-3-carboxamide (AZD9668) is a novel, oral inhibitor of neutrophil elastase (NE), an enzyme implicated in the signs, symptoms, and disease progression in NE-driven respiratory diseases such as bronchiectasis and chronic obstructive pulmonary disease via its role in the inflammatory process, mucus overproduction, and lung tissue damage. In vitro and in vivo experiments were done to evaluate the binding kinetics, potency, and selectivity of AZD9668, its effects in whole-blood and cell-based assays, and its efficacy in models of lung inflammation and damage. In contrast to earlier NE inhibitors, the interaction between AZD9668 and NE was rapidly reversible. AZD9668 was also highly selective for NE over other neutrophil-derived serine proteases. In cell-based assays, AZD9668 inhibited plasma NE activity in zymosan-stimulated whole blood. In isolated human polymorphonuclear cells, AZD9668 inhibited NE activity on the surface of stimulated cells and in the supernatant of primed, stimulated cells. AZD9668 showed good crossover potency to NE from other species. Oral administration of AZD9668 to mice or rats prevented human NE-induced lung injury, measured by lung hemorrhage, and an increase in matrix protein degradation products in bronchoalveolar lavage (BAL) fluid. In an acute smoke model, AZD9668 reduced the inflammatory response to cigarette smoke as indicated by a reduction in BAL neutrophils and interleukin-1β. Finally, AZD9668 prevented airspace enlargement and small airway wall remodeling in guinea pigs in response to chronic tobacco smoke exposure whether dosed therapeutically or prophylactically. In summary, AZD9668 has the potential to reduce lung inflammation and the associated structural and functional changes in human diseases.
Collapse
|
33
|
Tateosian NL, Reiteri RM, Amiano NO, Costa MJ, Villalonga X, Guerrieri D, Maffía PC. Neutrophil elastase treated dendritic cells promote the generation of CD4(+)FOXP3(+) regulatory T cells in vitro. Cell Immunol 2011; 269:128-34. [PMID: 21477798 DOI: 10.1016/j.cellimm.2011.03.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 03/02/2011] [Accepted: 03/15/2011] [Indexed: 12/31/2022]
Abstract
We have previously shown that neutrophilic elastase converts human immature dendritic cells (DCs) into TGF-β secreting cells and reduces its allostimulatory ability. Since TGF-β has been involved in regulatory T cells (Tregs) induction we analyzed whether elastase or neutrophil-derived culture supernatant treated DCs induce CD4(+)FOXP3(+) Tregs in a mixed lymphocyte reaction (MLR). We found that elastase or neutrophil-derived culture supernatant treated DCs increased TGF-β and decreased IL-6 production. Together with this pattern of cytokines, we observed a higher number of CD4(+)FOXP3(+) cells in the MLR cultures induced by elastase or neutrophil-derived culture supernatant treated DCs but not with untreated DCs. The higher number of CD4(+)FOXP3(+) T cell population was not observed when the enzymatic activity of elastase was inhibited with an elastase specific inhibitor and also when a TGF-β1 blocking antibody was added during the MLR culture. The increased number of CD4(+) that express FOXP3 was also seen when CD4(+)CD25(-) purified T cells were cocultured with the TGF-β producing DCs. Furthermore, these FOXP3(+) T cells showed suppressive activity in vitro. These results identify a novel mechanism by which the tolerogenic DCs generated by elastase exposure contribute to the immune regulation and may be relevant in the pathogenesis of several lung diseases where the inflammatory infiltrate contains high numbers of neutrophils and high elastase concentrations.
Collapse
Affiliation(s)
- N L Tateosian
- 3ra Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
IMPORTANCE OF THE FIELD Alpha-1-antitrypsin (A1AT) deficiency is a common genetic condition that predisposes individuals to the development of chronic obstructive pulmonary disease (COPD) as a direct result of damage caused to the lung by proteolytic enzymes released by migrating neutrophils. The lack of A1AT fails to control these enzymes and in the most common genetic deficiency (Pi Z) is due to accumulation of A1AT in the liver as a result of polymer formation. There is no specific treatment for COPD but understanding the pathophysiology of the disease in A1AT deficiency has led to strategies being used or developed to prevent the lung and liver disease. These strategies may have benefits beyond A1AT deficiency. AREAS COVERED IN THIS REVIEW The review covers the history of discovery of the nature and role of A1AT deficiency with particular emphasis on the pathophysiology of the lung disease. Evidence for the role of current therapies is provided together with data of preliminary or experimental strategies that are under development. WHAT THE READER WILL GAIN The reader will gain insight into the role of proteinases in the pathophysiology of COPD with particular reference to A1AT deficiency, which is the only human model of the disease. Current evidence of the efficacy of augmentation is provided together with new ways of readdressing the balance between neutrophil proteinases and natural or synthetic inhibitors or repairing lung damage. TAKE HOME MESSAGE A1AT deficiency is a good model to investigate the role of inflammation and proteolytic enzymes in the pathophysiology of COPD. Augmentation therapy is expensive but restores the deficiency to normal and current evidence suggests this ameliorates progression of the disease. Understanding the mechanisms involved has led to the development of newer strategies to protect the lung and liver from the development of disease but efficacy and safety concerns require careful introduction of these strategies. Although the condition is relatively common in the Northern hemisphere, the ability to deliver conventional Phase III clinical trials with lung physiology as the primary outcome will be limited by the sensitivity of the tests and number of patients required.
Collapse
Affiliation(s)
- Robert A Stockley
- University of Birmingham, Queen Elizabeth Hospital, Department of Medical Sciences, Edgbaston, Birmingham, B15 2TH, UK.
| |
Collapse
|
35
|
Wohner N, Keresztes Z, Sótonyi P, Szabó L, Komorowicz E, Machovich R, Kolev K. Neutrophil granulocyte-dependent proteolysis enhances platelet adhesion to the arterial wall under high-shear flow. J Thromb Haemost 2010; 8:1624-31. [PMID: 20412433 PMCID: PMC2905611 DOI: 10.1111/j.1538-7836.2010.03890.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 04/03/2010] [Indexed: 01/09/2023]
Abstract
SUMMARY BACKGROUND Under high shear stress platelets adhere preferentially to the adventitia layer of the arterial vessel wall in a von Willebrand factor (VWF)-dependent manner. OBJECTIVE The present study was undertaken in an attempt to characterize the structural background of the relative thromboresistance of the media and the impact of neutrophil leukocyte-derived proteases (matrix metalloproteinases, neutrophil elastase) on platelet adhesion in this layer of the arteries. METHODS AND RESULTS Platelet adhesion to cross-sections of the human iliac artery was monitored by indirect immunofluorescent detection of GpIIb/IIIa antigen. Exposure of the vessel wall to activated neutrophils or neutrophil-derived proteases increased platelet adhesion to the media about tenfold over the control level at 3350 s(-1) surface shear rate. In parallel with this enhanced thrombogenicity morphological changes in the media were evidenced by atomic force microscopy (AFM) and scanning electron microscopy (SEM). The fine proteoglycan meshwork seen with Cupromeronic Blue enhancement of the SEM images was removed by the proteolytic treatment and the typical collagen fiber structure was exposed on the AFM images of the media. CONCLUSION Through their proteases activated neutrophils degrade proteoglycans, unmask VWF binding sites and thus abolish the thromboresistance of the media in human arteries.
Collapse
Affiliation(s)
- N Wohner
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
36
|
Piper MG, Massullo PR, Loveland M, Druhan LJ, Kindwall-Keller TL, Ai J, Copelan A, Avalos BR. Neutrophil elastase downmodulates native G-CSFR expression and granulocyte-macrophage colony formation. JOURNAL OF INFLAMMATION-LONDON 2010; 7:5. [PMID: 20205821 PMCID: PMC2824667 DOI: 10.1186/1476-9255-7-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 01/21/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND The granulocyte colony-stimulating factor receptor (G-CSFR) plays a critical role in maintaining homeostatic levels of circulating neutrophils (PMN). The mechanisms modulating G-CSFR surface expression to prevent chronic neutrophilia are poorly understood. Here, we report that neutrophil elastase (NE) proteolytically cleaves the G-CSFR on human PMN and blocks G-CSFR-mediated granulopoiesis in vitro. METHODS Human peripheral blood PMN isolated from healthy donors were incubated with NE. Expression of the G-CSFR was analyzed by flow cytometry and western blot analyses. Detection of G-CSFR cleavage products from the culture supernatants was also performed. Human bone marrow mononuclear cells were also cultured in the presence or absence of NE to determine its effects on the proliferation of granulocyte-macrophage colony forming units (CFU-GM). RESULTS Treatment of PMN with NE induced a time-dependent decrease in G-CSFR expression that correlated with its degradation and the appearance of proteolytic cleavage fragments in conditioned media. Immunoblot analysis confirmed the G-CSFR was cleaved at its amino-terminus. Treatment of progenitor cells with NE prior to culture inhibited the growth of granulocyte-macrophage colony forming units. CONCLUSIONS These findings indicate that in addition to transcriptional controls and ligand-induced internalization, direct proteolytic cleavage of the G-CSFR by NE also downregulates G-CSFR expression and inhibits G-CSFR-mediated granulopoiesis in vitro. Our results suggest that NE negatively regulates granulopoiesis through a novel negative feedback loop.
Collapse
Affiliation(s)
- Melissa G Piper
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, 43210, OH, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Standish AJ, Weiser JN. Human neutrophils kill Streptococcus pneumoniae via serine proteases. THE JOURNAL OF IMMUNOLOGY 2009; 183:2602-9. [PMID: 19620298 DOI: 10.4049/jimmunol.0900688] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neutrophils, or polymorphonuclear leukocytes, comprise a crucial component of innate immunity, controlling bacterial and fungal infection through a combination of both oxidative and nonoxidative mechanisms. Indeed, neutrophils are believed to play an important role in controlling infection caused by the major human pathogen Streptococcus pneumoniae. However, the method by which neutrophils kill the pneumococcus as well as other Gram-positive bacteria, is not fully understood. We investigated human neutrophil killing of the pneumococcus in a complement-dependent opsonophagocytic assay. In contrast to other Gram-positive organisms, inhibition of the NADPH oxidase did not affect killing of S. pneumoniae. Supernatant from degranulated neutrophils killed the pneumococcus, suggesting a role for granular products. When neutrophil granule proteases were inhibited with either a protease mixture, or specific serine protease inhibitors 4-(2-Aminoethyl)benzenesulfonylfluoride and diisopropylfluorophosphate, killing by neutrophils was inhibited in a manner that correlated with increased intracellular survival. All three compounds inhibited intracellular activity of the three major neutrophil serine proteases: elastase, cathepsin G, and proteinase 3. Additionally, purified elastase and cathepsin G were sufficient to kill S. pneumoniae in a serine protease dependent-manner in in vitro assays. Inhibition studies using specific inhibitors of these serine proteases suggested that while each serine protease is sufficient to kill the pneumococcus, none is essential. Our findings show that Gram-positive pathogens are killed by human neutrophils via different mechanisms involving serine proteases.
Collapse
Affiliation(s)
- Alistair J Standish
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
38
|
Hirche TO, Benabid R, Deslee G, Gangloff S, Achilefu S, Guenounou M, Lebargy F, Hancock RE, Belaaouaj A. Neutrophil elastase mediates innate host protection against Pseudomonas aeruginosa. THE JOURNAL OF IMMUNOLOGY 2008; 181:4945-54. [PMID: 18802098 DOI: 10.4049/jimmunol.181.7.4945] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
According to the widely accepted view, neutrophil elastase (NE), a neutrophil-specific serine protease, is a major contributor to Pseudomonas aeruginosa infection-associated host tissue inflammation and damage, which in severe cases can lead to death. Herein, we provide for the first time compelling evidence that the host rather employs NE to protect itself against P. aeruginosa infection. Using a clinically relevant model of pneumonia, targeted deficiency in NE increased the susceptibility of mice to P. aeruginosa. We found that NE was required for maximal intracellular killing of P. aeruginosa by neutrophils. In investigating the mechanism of NE-mediated killing of P. aeruginosa, we found that NE degraded the major outer membrane protein F, a protein with important functions, including porin activity, maintenance of structural integrity, and sensing of host immune system activation. Consistent with this, the use of an isogenic mutant deficient in outer membrane protein F negated the role of NE in host defense against P. aeruginosa infection.
Collapse
Affiliation(s)
- Tim O Hirche
- Institut National de la Santé et de la Recherche Médicale, Programme Avenir, IFR53, University of Reims Champagne-Ardenne, Reims, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Leukocyte elastase (LE), a neutrophil serine protease, is known to cause alveolar wall destruction and alveolar hemorrhage in the lung, but recent evidence suggests that it may also produce a significant inflammatory response. The purpose of the current study was to (1) examine the relationship between LE-induced lung injury and specific markers of inflammation and cytokine/chemokine, and to (2) determine the potential of activated protein C (APC), a potent immunomodulator, to block the inflammatory response to LE. We treated the C57BL/6 mice with LE (10 U/kg, i.t.) and assessed the lung inflammation over 72 h. Total cells, total protein, and neutrophils were increased and peaked at 16 h in bronchial alveolar lavage fluid. Macrophages were also increased and peaked at 24 h. Administration of LE up-regulated the synthesis of proinflammatory cytokines, IL-1beta and IL-6, chemokines, keratinocyte-derived chemokine, and macrophage inflammatory protein 2 in bronchial alveolar lavage fluid, and their peaks were at 6 h. Furthermore, the mice were treated with APC at 0.2, 2.0, and 10 mg/kg (i.v.) after instillation of LE. Therapeutic treatment of APC at 2.0 and 10 mg/kg significantly attenuated the increases in all these parameters. Lung histology revealed that, in addition to inflammation, alveolar hemorrhage and alveolar wall destruction induced by LE were also attenuated by APC. Finally, the expression of tissue plasminogen activator and plasminogen activator inhibitor in whole lung of mice exposed to LE, detected by means of reverse-transcriptase-polymerase chain reaction, were not influenced by the treatment with APC. These data demonstrate that intratracheal administration of LE to mice causes a transient inflammatory response, and APC can play a protective role against LE-induced lung injury.
Collapse
|
40
|
Owen CA. Roles for proteinases in the pathogenesis of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2008; 3:253-68. [PMID: 18686734 PMCID: PMC2629972 DOI: 10.2147/copd.s2089] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Since the early 1960s, a compelling body of evidence has accumulated to show that proteinases play critical roles in airspace enlargement in chronic obstructive pulmonary disease (COPD). However, until recently the causative enzymes and their exact roles in pathologic processes in COPD have not been clear. Recent studies of gene-targeted mice in murine models of COPD have confirmed roles for proteinases not only in airspace enlargement, but also in airway pathologies in COPD. These studies have also shed light on the specific proteinases involved in COPD pathogenesis, and the mechanisms by which these proteinases injure the lung. They have also identified important interactions between different classes of proteinases, and between proteinases and other molecules that amplify lung inflammation and injury. This review will discuss the biology of proteinases and the mechanisms by which they contribute to the pathogenesis of COPD. In addition, I will discuss the potential of proteinase inhibitors and anti-inflammatory drugs as new treatment strategies for COPD patients.
Collapse
Affiliation(s)
- Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Kelly E, Greene CM, McElvaney NG. Targeting neutrophil elastase in cystic fibrosis. Expert Opin Ther Targets 2008; 12:145-57. [PMID: 18208364 DOI: 10.1517/14728222.12.2.145] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is a lethal hereditary disease characterised by neutrophil-dominated lung inflammation. These abundant neutrophils produce neutrophil elastase (NE), a destructive serine protease that has direct actions on extracellular matrix proteins and has a role in the host response to inflammation and infection. OBJECTIVE This review examines the prospect of developing novel therapies for CF by targeting NE. The authors explore the functions of NE and of naturally-occurring and synthetic NE inhibitors. METHODS A literature search was conducted exploring the functions of NE and inhibitors of NE; naturally occurring and synthetic. CONCLUSIONS Targeting NE in CF offers therapeutic potential, but optimal inhibitors that can be delivered safely and effectively to the lung are still under development.
Collapse
Affiliation(s)
- Emer Kelly
- Beaumont Hospital, Department of Respiratory Research, RCSI Smurfit Building, Beaumont, Dublin 9, Ireland.
| | | | | |
Collapse
|
42
|
Manolov T, Tan TT, Forsgren A, Riesbeck K. Moraxella-dependent alpha 1-antichymotrypsin neutralization: a unique virulence mechanism. Am J Respir Cell Mol Biol 2007; 38:609-17. [PMID: 18096871 DOI: 10.1165/rcmb.2007-0289oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The acute phase reactant and protease inhibitor alpha(1)-antichymotrypsin is considered to play a protective role in the airways, but whether it interacts with respiratory bacteria is not known. We analyzed whether the common respiratory pathogens Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, and other bacterial species interact with antichymotrypsin. M. catarrhalis was the only species that bound antichymotrypsin among 25 bacterial species tested by flow cytometry and direct binding assay. We compared a series of clinical isolates in addition to wild-type and ubiquitous surface protein-deficient Moraxella to study the nature of antichymotrypsin binding by the bacteria. Experiments with Moraxella mutants revealed that ubiquitous surface proteins A1 and A2 were responsible for the interaction, and using recombinant fragments, a consensus sequence within ubiquitous surface proteins A1 and A2 was defined. Binding of iodine-labeled antichymotrypsin was dose dependent and strong (dissociation constant [K(d)] 24.9-44.8 nM). Moreover, a chymotrypsin activity assay showed that antichymotrypsin, when bound to the bacterial surface, was neutralized. Moraxella antichymotrypsin neutralization is a novel microbial virulence mechanism that may induce excessive inflammation resulting in more exposed extracellular matrix that is beneficial for bacterial colonization.
Collapse
Affiliation(s)
- Taras Manolov
- Medical Microbiology, Department of Laboratory Medicine, Malmö University Hospital, Lund University, SE-205 02 Malmö, Sweden
| | | | | | | |
Collapse
|
43
|
Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physiopathological functions. Biochimie 2007; 90:227-42. [PMID: 18021746 DOI: 10.1016/j.biochi.2007.10.009] [Citation(s) in RCA: 337] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 10/19/2007] [Indexed: 11/21/2022]
Abstract
Polymorphonuclear neutrophils form a primary line of defense against bacterial infections using complementary oxidative and non-oxidative pathways to destroy phagocytized pathogens. The three serine proteases elastase, proteinase 3 and cathepsin G, are major components of the neutrophil primary granules that participate in the non-oxidative pathway of intracellular pathogen destruction. Neutrophil activation and degranulation results in the release of these proteases into the extracellular medium as proteolytically active enzymes, part of them remaining exposed at the cell surface. Extracellular neutrophil serine proteases also help kill bacteria and are involved in the degradation of extracellular matrix components during acute and chronic inflammation. But they are also important as specific regulators of the immune response, controlling cellular signaling through the processing of chemokines, modulating the cytokine network, and activating specific cell surface receptors. Neutrophil serine proteases are also involved in the pathogenicity of a variety of human diseases. This review focuses on the structural and functional properties of these proteases that may explain their specific biological roles, and facilitate their use as molecular targets for new therapeutic strategies.
Collapse
|
44
|
Richter R, Bistrian R, Escher S, Forssmann WG, Vakili J, Henschler R, Spodsberg N, Frimpong-Boateng A, Forssmann U. Quantum proteolytic activation of chemokine CCL15 by neutrophil granulocytes modulates mononuclear cell adhesiveness. THE JOURNAL OF IMMUNOLOGY 2005; 175:1599-608. [PMID: 16034099 DOI: 10.4049/jimmunol.175.3.1599] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monocyte infiltration into inflammatory sites is generally preceded by neutrophils. We show here that neutrophils may support this process by activation of CCL15, a human chemokine circulating in blood plasma. Neutrophils were found to release CCL15 proteolytic activity in the course of hemofiltration of blood from renal insufficiency patients. Processing of CCL15 immunoreactivity (IR) in the pericellular space is suggested by a lack of proteolytic activity in blood and blood filtrate, but a shift of the retention time (t(R)) of CCL15-IR, detected by chromatographic separation of CCL15-IR in blood and hemofiltrate. CCL15 molecules with N-terminal deletions of 23 (delta23) and 26 (delta26) aa were identified as main proteolytic products in hemofiltrate. Neutrophil cathepsin G was identified as the principal protease to produce delta23 and delta26 CCL15. Also, elastase displays CCL15 proteolytic activity and produces a delta21 isoform. Compared with full-length CCL15, delta23 and delta26 isoforms displayed a significantly increased potency to induce calcium fluxes and chemotactic activity on monocytes and to induce adhesiveness of mononuclear cells to fibronectin. Thus, our findings indicate that activation of monocytes by neutrophils is at least in part induced by quantum proteolytic processing of circulating or endothelium-bound CCL15 by neutrophil cathepsin G.
Collapse
|
45
|
Korkmaz B, Poutrain P, Hazouard E, de Monte M, Attucci S, Gauthier FL. Competition between elastase and related proteases from human neutrophil for binding to alpha1-protease inhibitor. Am J Respir Cell Mol Biol 2005; 32:553-9. [PMID: 15764720 DOI: 10.1165/rcmb.2004-0374oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The protease-antiprotease imbalance that is characteristic of most inflammatory lung disorders depends on the spatial-temporal regulation of active inhibitor and protease concentrations in lung secretions. We have studied the competition between the three main serine proteases from human neutrophil primary granules in their binding to alpha1-Pi, the main serine proteases inhibitor in lung secretions. Elastase was the only target of alpha1-Pi when identical molar amounts of purified inhibitor and the three proteases were tested together. The other two proteases were only inhibited once elastase was saturated. Elastase remained the preferred target of inhibitors when bronchoalveolar lavage fluids from patients with lung pneumonia and acute respiratory distress syndrome were used as the source of inhibitors, in spite of the presence of additional inhibitors in lung secretions. Since neutrophil proteases are expressed at the neutrophil surface, we also measured residual activities of membrane-bound proteases after purified neutrophils were incubated with bronchoalveolar fluids. Again, elastase was the preferred target of the inhibitors. We conclude that protease 3 and cathepsin G are not controlled as efficiently as elastase in lung secretions, a feature that must be taken into account when developing inhibitor-based anti-inflammatory therapies.
Collapse
Affiliation(s)
- Brice Korkmaz
- INSERM U618 Protéases et Vectorisation pulmonaires, University François Rabelais, 10 Bd Tonnellé, 37032 Tours Cedex, France
| | | | | | | | | | | |
Collapse
|
46
|
Eggers CT, Murray IA, Delmar VA, Day AG, Craik CS. The periplasmic serine protease inhibitor ecotin protects bacteria against neutrophil elastase. Biochem J 2004; 379:107-18. [PMID: 14705961 PMCID: PMC1224055 DOI: 10.1042/bj20031790] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Accepted: 01/06/2004] [Indexed: 11/17/2022]
Abstract
Ecotin is a dimeric periplasmic protein from Escherichia coli that has been shown to inhibit potently many trypsin-fold serine proteases of widely varying substrate specificity. To help elucidate the physiological function of ecotin, we examined the family of ecotin orthologues, which are present in a subset of Gram-negative bacteria. Phylogenetic analysis suggested that ecotin has an exogenous target, possibly neutrophil elastase. Recombinant protein was expressed and purified from E. coli, Yersinia pestis and Pseudomonas aeruginosa, all species that encounter the mammalian immune system, and also from the plant pathogen Pantoea citrea. Notably, the Pa. citrea variant inhibits neutrophil elastase 1000-fold less potently than the other orthologues. All four orthologues are dimeric proteins that potently inhibit (<10 pM) the pancreatic digestive proteases trypsin and chymotrypsin, while showing more variable inhibition (5 pM to 24 microM) of the blood proteases Factor Xa, thrombin and urokinase-type plasminogen activator. To test whether ecotin does, in fact, protect bacteria from neutrophil elastase, an ecotin-deficient strain was generated in E. coli. This strain is significantly more sensitive in cell-killing assays to human neutrophil elastase, which causes increased permeability of the outer membrane that persists even during renewed bacterial growth. Ecotin affects primarily the ability of E. coli to recover and grow following treatment with neutrophil elastase, rather than the actual rate of killing. This suggests that an important part of the antimicrobial mechanism of neutrophil elastase may be a periplasmic bacteriostatic effect of protease that has translocated across the damaged outer membrane.
Collapse
Affiliation(s)
- Christopher T Eggers
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94143-92280, USA
| | | | | | | | | |
Collapse
|
47
|
López-Boado YS, Espinola M, Bahr S, Belaaouaj A. Neutrophil serine proteinases cleave bacterial flagellin, abrogating its host response-inducing activity. THE JOURNAL OF IMMUNOLOGY 2004; 172:509-15. [PMID: 14688361 DOI: 10.4049/jimmunol.172.1.509] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
After bacterial infection, neutrophils dominate the cellular infiltrate. Their main function is assumed to be killing invading pathogens and resolving the inflammation they cause. Activated neutrophils are also known to release a variety of molecules, including the neutrophil serine proteinases, extracellularly. The release of these proteinases during inflammation creates a proteolytic environment where degradation of different molecules modulates the inflammatory response. Flagellin, the structural component of flagella on many bacterial species, is a virulence factor with a strong proinflammatory activity on epithelial cells and other cell types. In this study we show that both human and mouse neutrophil serine proteinases cleave flagellin from Pseudomonas aeruginosa and other bacterial species. More important, cleavage of P. aeruginosa flagellin by the neutrophil serine proteinases neutrophil elastase and cathepsin G resulted in loss of the biological activity of this virulence factor, as evidenced by the lack of innate host defense gene expression in human epithelial cells. The finding that flagellin is susceptible to cleavage by neutrophil serine proteinases suggests a novel role for these enzymes in the inflammatory response to infection. Not only can these enzymes kill bacteria, but they also degrade their virulence factors to halt the inflammatory response they trigger.
Collapse
Affiliation(s)
- Yolanda S López-Boado
- Department of Internal Medicine (Molecular Medicine), Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
48
|
Hirche TO, Atkinson JJ, Bahr S, Belaaouaj A. Deficiency in neutrophil elastase does not impair neutrophil recruitment to inflamed sites. Am J Respir Cell Mol Biol 2003; 30:576-84. [PMID: 14565940 DOI: 10.1165/rcmb.2003-0253oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To reach the sites of inflammation, neutrophils traverse the endothelium, its underlying basement membrane, and other barriers depending on the localization of the insulting agent. Whether neutrophil elastase (NE) plays a role in neutrophil recruitment to inflamed sites is still debatable. By exploiting mice deficient in NE (NE(-/-)), we sought to address this dilemma. We recruited neutrophils to the lungs or the peritoneum of wild-type (WT) or NE(-/-) mice by intranasal or intraperitoneal challenge with Pseudomonas aeruginosa or its lipopolysaccharide. At designated times post-inoculation (0, 4, 24, and 48 h), groups of mice were killed to assess changes in leukocyte counts and inflammatory responses. NE(-/-) and WT mice had normal circulating leukocyte numbers including neutrophils and changes in the hemograms in the setting of acute inflammation were indistinguishable. Analyses of lung tissues or fluids from the lungs and peritoneum found that regardless of the inflammatory model, the leukocyte counts including neutrophils and the inflammatory response were similar in NE(-/-) and WT mice at all time points. In vitro, neutrophils isolated from the lungs or the peritoneum of NE(-/-) and WT mice had comparable chemotactic and respiratory-burst functions and migrated normally through Matrigel in response to various stimuli. Interestingly, preincubation of human peripheral blood neutrophils with NE physiologic inhibitors did not alter the migration of the cells through Matrigel. In sum, our findings present the first in vivo description that the absence of NE does not impair neutrophil recruitment to inflamed sites and that NE is not required for basement membrane transmigration of neutrophils.
Collapse
Affiliation(s)
- Tim O Hirche
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110-1093, USA
| | | | | | | |
Collapse
|
49
|
Uehara A, Muramoto K, Takada H, Sugawara S. Neutrophil serine proteinases activate human nonepithelial cells to produce inflammatory cytokines through protease-activated receptor 2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5690-6. [PMID: 12759451 DOI: 10.4049/jimmunol.170.11.5690] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protease-activated receptors (PARs) compose a family of G protein-coupled receptors activated by proteolysis with exposure of their tethered ligand. Recently, we reported that a neutrophil-derived serine proteinase, proteinase 3 (PR3), activated human oral epithelial cells through PAR-2. The present study examined whether other neutrophil serine proteinases, human leukocyte elastase (HLE), and cathepsin G (Cat G) activate nonepithelial cells, human gingival fibroblasts (HGF). HLE and Cat G as well as PR3 activated HGF to produce IL-8 and monocyte chemoattractant protein 1. Human oral epithelial cells but not HGF express mRNA and protein of secretory leukocyte protease inhibitor, an inhibitor of HLE and Cat G, and recombinant secretory leukocyte protease inhibitor clearly inhibited the activation of HGF induced by HLE and Cat G but not by PR3. HGF express PAR-1 and PAR-2 mRNA in the cells and the proteins on the cell surface. HLE and Cat G cleaved the peptide corresponding to the N terminus of PAR-2 with exposure of its tethered ligand. Treatment with trypsin, an agonist for PAR-2, and a synthetic PAR-2 agonist peptide induced intracellular Ca(2+) mobilization and rendered cells refractory to subsequent stimulation with HLE and Cat G. The production of cytokine induced by HLE and Cat G and the PAR-2 agonist peptide was completely abolished by inhibition of phospholipase C. These findings suggest that neutrophil serine proteinases have equal ability to activate human nonepithelial cells through PAR-2 to produce inflammatory cytokines and may control a number of inflammatory processes such as periodontitis.
Collapse
Affiliation(s)
- Akiko Uehara
- Department of Microbiology and Immunology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | | | | | | |
Collapse
|
50
|
Kapui Z, Varga M, Urban-Szabo K, Mikus E, Szabo T, Szeredi J, Batori S, Finance O, Aranyi P. Biochemical and pharmacological characterization of 2-(9-(2-piperidinoethoxy)-4-oxo-4H-pyrido[1,2-a]pyrimidin-2-yloxymethyl)-4-(1-methylethyl)-6-methoxy-1,2-benzisothiazol-3(2H)-one-1,1-dioxide (SSR69071), a novel, orally active elastase inhibitor. J Pharmacol Exp Ther 2003; 305:451-9. [PMID: 12606659 DOI: 10.1124/jpet.102.044263] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human leukocyte elastase (HLE) is a proteinase capable of degrading a variety of proteins. Under normal circumstances, the proteolytic activity of HLE is effectively controlled by its natural inhibitors. However, an imbalance between elastase and its endogenous inhibitors may result in several pathophysiological states such as chronic obstructive pulmonary disease, asthma, emphysema, cystic fibrosis, and chronic inflammatory diseases. It is anticipated that an orally active HLE inhibitor could be useful for the treatment of these diseases. 2-(9-(2-Piperidinoethoxy)-4-oxo-4H-pyrido[1,2-a]pyrimidin-2-yloxymethyl)-4-(1-methylethyl)-6-methoxy-1,2-benzisothiazol-3(2H)-one-1,1-dioxide (SSR69071) is a potent inhibitor of HLE, with the inhibition constant (K(i)) and the constant for inactivation process (k(on)) being 0.0168 +/- 0.0014 nM and 0.183 +/- 0.013 10(6)/mol sr, respectively. The dissociation rate constant, k(off), was 3.11 + 0.37 10(-6)/s. SSR69071 displays a higher affinity for human elastase than for rat (K(i) = 3 nM), mouse (K(i) = 1.8 nM), and rabbit (K(i) = 58 nM) elastases. Bronchoalveolar lavage fluid from mice orally treated with SSR69071 inhibits HLE (ex vivo), and in this model, SSR69071 has a dose-dependent efficacy with an ED(50) = 10.5 mg/kg p.o. SSR69071 decreases significantly the acute lung hemorrhage induced by HLE (ED(50) = 2.8 mg/kg p.o.) in mice. Furthermore, SSR69071 prevents carrageenan- (ED(30) = 2.2 mg/kg) and HLE-induced (ED(30) = 2.7 mg/kg) paw edema in rats after p.o. administration. In conclusion, SSR69071 is a selective, orally active, and potent inhibitor of HLE with good penetration in respiratory tissues.
Collapse
Affiliation(s)
- Zoltan Kapui
- Discovery Research, Sanofi-Synthelabo Internal Medicine Budapest Site, Chinoin, Sanofi-Synthelabo Group, Budapest, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|