1
|
Hampton TH, Barnaby R, Roche C, Nymon A, Fukutani KF, MacKenzie TA, Stanton BA. Gene expression responses of CF airway epithelial cells exposed to elexacaftor/tezacaftor/ivacaftor (ETI) suggest benefits beyond improved CFTR channel function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610162. [PMID: 39257747 PMCID: PMC11383677 DOI: 10.1101/2024.08.28.610162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The combination of elexacaftor/tezacaftor/ivacaftor (ETI, Trikafta) reverses the primary defect in Cystic Fibrosis (CF) by improving CFTR mediated Cl - and HCO 3 - secretion by airway epithelial cells (AEC), leading to improved lung function and less frequent exacerbations and hospitalizations. However, studies have shown that CFTR modulators like ivacaftor, a component of ETI, has numerous effects on CF cells beyond improved CFTR channel function. Because little is known about the effect of ETI on CF AEC gene expression we exposed primary human AEC to ETI for 48 hours and interrogated the transcriptome by RNA-seq and qPCR. ETI increased defensin gene expression ( DEFB1 ) an observation consistent with reports of decreased bacterial burden in the lungs of people with CF (pwCF). ETI also decreased MMP10 and MMP12 gene expression, suggesting that ETI may reduce proteolytic induced lung destruction in CF. ETI also reduced the expression of the stress response gene heme oxygenase ( HMOX1 ). qPCR analysis confirmed DEFB1, HMOX1, MMP10 and MMP12 gene expression results observed by RNA-seq. Gene pathway analysis revealed that ETI decreased inflammatory signaling, cellular proliferation and MHC Class II antigen presentation. Collectively, these findings suggest that the clinical observation that ETI reduces lung infections in pwCF is related in part to drug induced increases in DEFB1 , and that ETI may reduce lung damage by reducing MMP10 and MMP12 gene expression, which is predicted to reduce matrix metalloprotease activity. Moreover, pathway analysis also identified several genes responsible for the ETI induced reduction in inflammation observed in people with CF. New and Noteworthy Gene expression responses by CF AEC exposed to ETI suggest that in addition to improving CFTR channel function, ETI is likely to increase resistance to bacterial infection by increasing levels of beta defensin 1 (hBD-1). ETI may also reduce lung damage by suppressing MMP10, and reduce airway inflammation by repressing proinflammatory cytokine secretion by AEC cells.
Collapse
|
2
|
Mall MA, Davies JC, Donaldson SH, Jain R, Chalmers JD, Shteinberg M. Neutrophil serine proteases in cystic fibrosis: role in disease pathogenesis and rationale as a therapeutic target. Eur Respir Rev 2024; 33:240001. [PMID: 39293854 PMCID: PMC11409056 DOI: 10.1183/16000617.0001-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/09/2024] [Indexed: 09/20/2024] Open
Abstract
Chronic airway inflammation is a central feature in the pathogenesis of bronchiectasis (BE), which can be caused by cystic fibrosis (CFBE; hereafter referred to as CF lung disease) and non-CF-related conditions (NCFBE). Inflammation in both CF lung disease and NCFBE is predominantly driven by neutrophils, which release proinflammatory cytokines and granule proteins, including neutrophil serine proteases (NSPs). NSPs include neutrophil elastase, proteinase 3 and cathepsin G. An imbalance between NSPs and their antiproteases has been observed in people with CF lung disease and people with NCFBE. While the role of the protease/antiprotease imbalance is well established in both CF lung disease and NCFBE, effective therapies targeting NSPs are lacking. In recent years, the introduction of CF transmembrane conductance regulator (CFTR) modulator therapy has immensely improved outcomes in many people with CF (pwCF). Despite this, evidence suggests that airway inflammation persists, even in pwCF treated with CFTR modulator therapy. In this review, we summarise current data on neutrophilic inflammation in CF lung disease to assess whether neutrophilic inflammation and high, uncontrolled NSP levels play similar roles in CF lung disease and in NCFBE. We discuss similarities between the neutrophilic inflammatory profiles of people with CF lung disease and NCFBE, potentially supporting a similar therapeutic approach. Additionally, we present evidence suggesting that neutrophilic inflammation persists in pwCF treated with CFTR modulator therapy, at levels similar to those in people with NCFBE. Collectively, these findings highlight the ongoing need for new treatment strategies targeting neutrophilic inflammation in CF lung disease.
Collapse
Affiliation(s)
- Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Scott H Donaldson
- Department of Medicine, Division of Pulmonary Diseases and Critical Care Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Raksha Jain
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Michal Shteinberg
- Lady Davis Carmel Medical Center, Haifa, Israel
- The B. Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
3
|
Li T, Mao N, Xie Z, Wang J, Jin F, Li Y, Liu S, Cai W, Gao X, Wei Z, Yang F, Xu H, Liu H, Zhang H, Xu D. Paeoniflorin mitigates MMP-12 inflammation in silicosis via Yang-Yin-Qing-Fei Decoction in murine models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155616. [PMID: 38669965 DOI: 10.1016/j.phymed.2024.155616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Silicosis presents a significant clinical challenges and economic burdens, with Traditional Chinese Medicine (TCM) emerging as a potential therapeutic avenue. However, the precise effects and mechanisms of TCM in treating silicosis remain uncertain and subject to debate. OBJECTIVE The study aims to elucidate the therapeutic role and mechanisms of the Yang-Yin-Qing-Fei Decoction (YYQFD) and its key component, paeoniflorin, in silicosis using a murine model. METHODS Silicotic mice were treated with YYQFD, pirfenidone (PFD), or paeoniflorin. RAW264.7 cells and mouse lung fibroblasts (MLF) were stimulated with silica, matrix metalloproteinase-12 (MMP-12), or TGF-β1, followed by treatment with paeoniflorin, PFD, or relevant inhibitors. YYQFD constituents were characterized using High-Performance Liquid Chromatography (HPLC). Lung fibrosis severity was assessed via histopathological examination, micro-CT imaging, lung functions, and Western blot analysis. Transcriptome sequencing and bioinformatics analysis were employed to delineate the gene expression profile and target genes modulated by YYQFD in silicosis. RESULTS Treatment with YYQFD ameliorated silica-induced lung fibrosis. Transcriptome sequencing identified MMP-12 as a potential common target of YYQFD and PFD. Additionally, a potential pro-inflammatory role of MMP-12, regulated by silica-induced TLR4 signaling pathways, was revealed. Paeoniflorin, one of the most distinctive compounds in YYQFD, attenuated silica-induced MMP-12 increase and its derived inflammatory factors in macrophages through a direct binding effect. Notably, paeoniflorin treatment exerted anti-fibrotic effects by inhibiting MMP-12-derived inflammatory factors and TGF-β1-induced myofibroblast differentiation in silica-exposed mice. CONCLUSIONS This study underscores paeoniflorin as one of the most principal bioactive compounds in YYQFD, highlighting its capacity to attenuate lung inflammation driven by macrophage-derived MMP-12 and reduce lung fibrosis both in vivo and in vitro.
Collapse
Affiliation(s)
- Tian Li
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Na Mao
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Zihao Xie
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Jianing Wang
- College of traditional Chinese medicine, North China University of Science and Technology, Tangshan, Hebei Province 063210, China
| | - Fuyu Jin
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Yaqian Li
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Shupeng Liu
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Wenchen Cai
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Xuemin Gao
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China; National Health Commission (NHC) Key Laboratory of Pneumoconiosis, Taiyuan 030001, Shanxi province, China
| | - Zhongqiu Wei
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Fang Yang
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Hong Xu
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China; National Health Commission (NHC) Key Laboratory of Pneumoconiosis, Taiyuan 030001, Shanxi province, China; Health Science Center, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Heliang Liu
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China.
| | - Haibo Zhang
- Department of Anesthesiology and Pain Medicine, Department of Physiology, Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Dingjie Xu
- College of traditional Chinese medicine, North China University of Science and Technology, Tangshan, Hebei Province 063210, China.
| |
Collapse
|
4
|
Bakalović G, Bokonjić D, Mihajlović D, Čolić M, Mališ V, Drakul M, Tomić S, Jojić I, Rakočević S, Popović D, Kozić L, Vasiljević M, Bekić M, Mašić S, Ljuboja O. Dysfunctions of Neutrophils in the Peripheral Blood of Children with Cystic Fibrosis. Biomedicines 2023; 11:1725. [PMID: 37371820 DOI: 10.3390/biomedicines11061725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Dysfunction of neutrophils in patients with cystic fibrosis (CF) is best characterized in bronchoalveolar lavage (BAL), whereas peripheral blood neutrophils are less examined, and the results are contradictory, especially in younger populations. Therefore, this work aimed to study functional and phenotypic changes in circulating neutrophils in children with CF. The study included 19 CF children (5-17 years) and 14 corresponding age-matched healthy children. Isolated neutrophils were cultured either alone or with different stimuli. Several functions were studied: apoptosis, NET-osis, phagocytosis, and production of reactive oxygen species (ROS), neutrophil elastase (NE), and 11 cytokines. In addition, the expression of 20 molecules involved in different functions of neutrophils was evaluated by using flow cytometry. CF neutrophils showed reduced apoptosis and lower production of NE and IL-18 compared to the healthy controls, whereas IL-8 was augmented. All of these functions were further potentiated after neutrophil stimulation, which included higher ROS production and the up-regulation of CD11b and IL-10 expression. NET-osis was higher only when neutrophils from moderate-severe CF were treated with Pseudomonas aeruginosa, and the process correlated with forced expiratory volume in the first second (FEV1). Phagocytosis was not significantly changed. In conclusion, circulating neutrophils from children with CF showed fewer impaired changes in phenotype than in function. Functional abnormalities, which were already present at the baseline levels in neutrophils, depended on the type of stimuli that mimicked different activation states of these cells at the site of infection.
Collapse
Affiliation(s)
- Ganimeta Bakalović
- Pediatric Clinic, Clinical Center of the University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
| | - Dejan Bokonjić
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
- Department of Pediatrics, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Dušan Mihajlović
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Miodrag Čolić
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Vanja Mališ
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Marija Drakul
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia
| | - Ivan Jojić
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Sara Rakočević
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Darinka Popović
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Ljiljana Kozić
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Miloš Vasiljević
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Marina Bekić
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia
| | - Srđan Mašić
- Center for Biomedical Sciences, Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina
| | - Olivera Ljuboja
- Clinic for Children's Diseases, University Clinical Center of Banja Luka, 51000 Banja Luka, Bosnia and Herzegovina
| |
Collapse
|
5
|
Smith ET, Kruppa M, Johnson DA, Van Haeften J, Chen X, Leahy D, Peake J, Harris JM. High yield expression in Pichia pastoris of human neutrophil elastase fused to cytochrome B5. Protein Expr Purif 2023; 206:106255. [PMID: 36822453 PMCID: PMC10118287 DOI: 10.1016/j.pep.2023.106255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
Recombinant human neutrophil elastase (rHNE), a serine protease, was expressed in Pichia pastoris. Glycosylation sites were removed via bioengineering to prevent hyper-glycosylation (a common problem with this system) and the cDNA was codon optimized for translation in Pichia pastoris. The zymogen form of rHNE was secreted as a fusion protein with an N-terminal six histidine tag followed by the heme binding domain of Cytochrome B5 (CytB5) linked to the N-terminus of the rHNE sequence via an enteropeptidase cleavage site. The CytB5 fusion balanced the very basic rHNE (pI = 9.89) to give a colored fusion protein (pI = 6.87), purified via IMAC. Active rHNE was obtained via enteropeptidase cleavage, and purified via cation exchange chromatography, resulting in a single protein band on SDS PAGE (Mr = 25 KDa). Peptide mass fingerprinting analysis confirmed the rHNE amino acid sequence, the absence of glycosylation and the absence of an 8 amino acid C-terminal peptide as opposed to the 20 amino acids usually missing from the C-terminus of native enzyme. The yield of active rHNE was 0.41 mg/L of baffled shaker flask culture medium. Active site titration with alpha-1 antitrypsin, a potent irreversible elastase inhibitor, quantified the concentration of purified active enzyme. The Km of rHNE with methoxy-succinyl-AAPVpNA was identical with that of the native enzyme within the assay's limit of accuracy. This is the first report of full-length rHNE expression at high yields and low cost facilitating further studies on this major human neutrophil enzyme.
Collapse
Affiliation(s)
- Eliot T Smith
- Departments of Biomedical Sciences and Medical Education, James H. Quillen College of Medicine, East Tennessee State University, Box 70582, Johnson City, Tennessee, USA
| | - Michael Kruppa
- Departments of Biomedical Sciences and Medical Education, James H. Quillen College of Medicine, East Tennessee State University, Box 70582, Johnson City, Tennessee, USA
| | - David A Johnson
- Departments of Biomedical Sciences and Medical Education, James H. Quillen College of Medicine, East Tennessee State University, Box 70582, Johnson City, Tennessee, USA.
| | - Jessica Van Haeften
- Queensland University of Technology, Molecular Simulation Group, Institute of Health and Biomedical Innovation, Corner Blamey Street & Musk Avenue, Kelvin Grove Urban Village, Queensland, 4059, Australia
| | - Xingchen Chen
- Queensland University of Technology, Molecular Simulation Group, Institute of Health and Biomedical Innovation, Corner Blamey Street & Musk Avenue, Kelvin Grove Urban Village, Queensland, 4059, Australia
| | - Darren Leahy
- Queensland University of Technology, Molecular Simulation Group, Institute of Health and Biomedical Innovation, Corner Blamey Street & Musk Avenue, Kelvin Grove Urban Village, Queensland, 4059, Australia
| | - Jonathan Peake
- Queensland University of Technology, Molecular Simulation Group, Institute of Health and Biomedical Innovation, Corner Blamey Street & Musk Avenue, Kelvin Grove Urban Village, Queensland, 4059, Australia
| | - Jonathan M Harris
- Queensland University of Technology, Molecular Simulation Group, Institute of Health and Biomedical Innovation, Corner Blamey Street & Musk Avenue, Kelvin Grove Urban Village, Queensland, 4059, Australia
| |
Collapse
|
6
|
The Role of MMPs in the Era of CFTR Modulators: An Additional Target for Cystic Fibrosis Patients? Biomolecules 2023; 13:biom13020350. [PMID: 36830719 PMCID: PMC9952876 DOI: 10.3390/biom13020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Cystic fibrosis (CF) is a high-prevalence disease characterized by significant lung remodeling, responsible for high morbidity and mortality worldwide. The lung structural changes are partly due to proteolytic activity associated with inflammatory cells such as neutrophils and macrophages. Matrix metalloproteases (MMPs) are the major proteases involved in CF, and recent literature data focused on their potential role in the pathogenesis of the disease. In fact, an imbalance of proteases and antiproteases was observed in CF patients, resulting in dysfunction of protease activity and loss of lung homeostasis. Currently, many steps forward have been moved in the field of pharmacological treatment with the recent introduction of triple-combination therapy targeting the CFTR channel. Despite CFTR modulator therapy potentially being effective in up to 90% of patients with CF, there are still patients who are not eligible for the available therapies. Here, we introduce experimental drugs to provide updates on therapy evolution regarding a proportion of CF non-responder patients to current treatment, and we summarize the role of MMPs in pathogenesis and as future therapeutic targets of CF.
Collapse
|
7
|
Efficacy of selective histone deacetylase 6 inhibition in mouse models of Pseudomonas aeruginosa infection: A new glimpse for reducing inflammation and infection in cystic fibrosis. Eur J Pharmacol 2022; 936:175349. [DOI: 10.1016/j.ejphar.2022.175349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022]
|
8
|
Cheng Y, Li G, Smedley CJ, Giel MC, Kitamura S, Woehl JL, Bianco G, Forli S, Homer JA, Cappiello JR, Wolan DW, Moses JE, Sharpless KB. Diversity oriented clicking delivers β-substituted alkenyl sulfonyl fluorides as covalent human neutrophil elastase inhibitors. Proc Natl Acad Sci U S A 2022; 119:e2208540119. [PMID: 36070343 PMCID: PMC9478681 DOI: 10.1073/pnas.2208540119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023] Open
Abstract
Diversity Oriented Clicking (DOC) is a discovery method geared toward the rapid synthesis of functional libraries. It combines the best attributes of both classical and modern click chemistries. DOC strategies center upon the chemical diversification of core "SuFExable" hubs-exemplified by 2-Substituted-Alkynyl-1-Sulfonyl Fluorides (SASFs)-enabling the modular assembly of compounds through multiple reaction pathways. We report here a range of stereoselective Michael-type addition pathways from SASF hubs including reactions with secondary amines, carboxylates, 1H-1,2,3-triazole, and halides. These high yielding conjugate addition pathways deliver unprecedented β-substituted alkenyl sulfonyl fluorides as single isomers with minimal purification, greatly enriching the repertoire of DOC and holding true to the fundamentals of modular click chemistry. Further, we demonstrate the potential for biological function - a key objective of click chemistry - of this family of SASF-derived molecules as covalent inhibitors of human neutrophil elastase.
Collapse
Affiliation(s)
- Yunfei Cheng
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Gencheng Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Marie-Claire Giel
- La Trobe Institute for Molecular Science, Melbourne, VIC 3086, Australia
| | - Seiya Kitamura
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Jordan L. Woehl
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Giulia Bianco
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Joshua A. Homer
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - John R. Cappiello
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Dennis W. Wolan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - John E. Moses
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - K. Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
9
|
Dave R, Randhawa G, Kim D, Simpson M, Hoare T. Microgels and Nanogels for the Delivery of Poorly Water-Soluble Drugs. Mol Pharm 2022; 19:1704-1721. [PMID: 35319212 DOI: 10.1021/acs.molpharmaceut.1c00967] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While microgels and nanogels are most commonly used for the delivery of hydrophilic therapeutics, the water-swollen structure, size, deformability, colloidal stability, functionality, and physicochemical tunability of microgels can also offer benefits for addressing many of the barriers of conventional vehicles for the delivery of hydrophobic therapeutics. In this review, we describe approaches for designing microgels with the potential to load and subsequently deliver hydrophobic drugs by creating compartmentalized microgels (e.g., core-shell structures), introducing hydrophobic domains in microgels, leveraging host-guest interactions, and/or applying "smart" environmentally responsive materials with switchable hydrophobicity. In particular, the challenge of promoting hydrophobic drug loading without compromising the inherent advantages of microgels as delivery vehicles and ensuring practically relevant release kinetics from such structures is highlighted, with an eye toward the practical translation of such vehicles to the clinic.
Collapse
Affiliation(s)
- Ridhdhi Dave
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Gurpreet Randhawa
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Daeun Kim
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Madeline Simpson
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
10
|
Barone S, Cassese E, Alfano AI, Brindisi M, Summa V. Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology. J Med Chem 2022; 65:3080-3097. [PMID: 35148101 PMCID: PMC8883472 DOI: 10.1021/acs.jmedchem.1c02067] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
Compelling new support
has been provided for histone deacetylase
isoform 6 (HDAC6) as a common thread in the generation of the dysregulated
proinflammatory and fibrotic phenotype in cystic fibrosis (CF). HDAC6
also plays a crucial role in bacterial clearance or killing as a direct
consequence of its effects on CF immune responses. Inhibiting HDAC6
functions thus eventually represents an innovative and effective strategy
to tackle multiple aspects of CF-associated lung disease. In this
Perspective, we not only showcase the latest evidence linking HDAC(6)
activity and expression with CF phenotype but also track the new dawn
of HDAC(6) modulators in CF and explore potentialities and future
perspectives in the field.
Collapse
Affiliation(s)
- Simona Barone
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Emilia Cassese
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Antonella Ilenia Alfano
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Vincenzo Summa
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| |
Collapse
|
11
|
AbdulWahab A, Allangawi M, Thomas M, Bettahi I, Sivaraman SK, Jerobin J, Chandra P, Ramanjaneya M, Abou-Samra AB. Sputum and Plasma Neutrophil Elastase in Stable Adult Patients With Cystic Fibrosis in Relation to Chronic Pseudomonas Aeruginosa Colonization. Cureus 2021; 13:e15948. [PMID: 34221778 PMCID: PMC8238017 DOI: 10.7759/cureus.15948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background and purpose Neutrophil elastase (NE) has been implicated in the pathogenesis of airway inflammation in cystic fibrosis (CF) patients and it impairs defenses against Pseudomonas aeruginosa (PA) infection or colonization. Sputum NE may act as a biomarker of neutrophilic inflammation in CF patients. This study aimed to determine sputum and plasma total NE levels in clinically stable adult CF patients and control subjects, and their correlation to PA colonization and lung functions. Methods This is a cross-sectional study. Total NE was measured on spontaneously expectorated sputum and plasma obtained from 21 CF patients, aged 18-40 years, during routine visits to the adult CF clinic. This was compared to plasma obtained from 22 matching healthy controls. The levels of NE were measured by the magnetic bead-based multiplex assay. Results Sputum and plasma NE levels had a significant positive correlation (Pearson r=0.533, P=0.013) with PA colonization. Sixteen CF patients (76.2%) were chronically colonized with PA. Both median sputum and plasma NE were found to be higher in CF patients with PA as compared with non-PA patients, even though this difference was statistically insignificant. Sputum and plasma NE levels did not correlate with the percentage predicted forced expiratory volume in one second (FEV1), the forced vital capacity (FVC), and FEV1/FVC and no association with PA. Conclusion The findings suggest that clinically stable adult CF patients colonized with PA may have higher NE levels in both plasma and sputum as compared to non-PA CF patients and probably total NE does not influence lung functions.
Collapse
Affiliation(s)
| | | | | | - Ilham Bettahi
- Qatar Metabolic Institute, Interim Translational Research Institute, Academic Health System, Hamad General Hospital, Doha, QAT
| | - Siveen K Sivaraman
- Interim Translational Research Institute, Academic Health System, Hamad General Hospital, Doha, QAT
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Interim Translational Research Institute, Academic Health System, Hamad General Hospital, Doha, QAT
| | - Prem Chandra
- Medical Research Center, Hamad Medical Hospital, Doha, QAT
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Interim Translational Research Institute, Academic Health System, Hamad General Hospital, Doha, QAT
| | | |
Collapse
|
12
|
Dysfunctional Inflammation in Cystic Fibrosis Airways: From Mechanisms to Novel Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22041952. [PMID: 33669352 PMCID: PMC7920244 DOI: 10.3390/ijms22041952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/27/2022] Open
Abstract
Cystic fibrosis (CF) is an inherited disorder caused by mutations in the gene encoding for the cystic fibrosis transmembrane conductance regulator (CFTR) protein, an ATP-gated chloride channel expressed on the apical surface of airway epithelial cells. CFTR absence/dysfunction results in defective ion transport and subsequent airway surface liquid dehydration that severely compromise the airway microenvironment. Noxious agents and pathogens are entrapped inside the abnormally thick mucus layer and establish a highly inflammatory environment, ultimately leading to lung damage. Since chronic airway inflammation plays a crucial role in CF pathophysiology, several studies have investigated the mechanisms responsible for the altered inflammatory/immune response that, in turn, exacerbates the epithelial dysfunction and infection susceptibility in CF patients. In this review, we address the evidence for a critical role of dysfunctional inflammation in lung damage in CF and discuss current therapeutic approaches targeting this condition, as well as potential new treatments that have been developed recently. Traditional therapeutic strategies have shown several limitations and limited clinical benefits. Therefore, many efforts have been made to develop alternative treatments and novel therapeutic approaches, and recent findings have identified new molecules as potential anti-inflammatory agents that may exert beneficial effects in CF patients. Furthermore, the potential anti-inflammatory properties of CFTR modulators, a class of drugs that directly target the molecular defect of CF, also will be critically reviewed. Finally, we also will discuss the possible impact of SARS-CoV-2 infection on CF patients, with a major focus on the consequences that the viral infection could have on the persistent inflammation in these patients.
Collapse
|
13
|
Wang X, Wang M, Chen S, Wei B, Gao Y, Huang L, Liu C, Huang T, Yu M, Zhao SH, Li X. Ammonia exposure causes lung injuries and disturbs pulmonary circadian clock gene network in a pig study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 205:111050. [PMID: 32827960 DOI: 10.1016/j.ecoenv.2020.111050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 06/11/2023]
Abstract
Ammonia toxicity to respiratory system in pig faming is of particular concern, but the molecular mechanism remains still unclear. The present study was devoted to assess the impacts of the ammonia exposure on the lung tissues based on a pig study using 80 ppm ammonia exposing to piglets for different days. The histology analysis revealed ammonia exposure induced lung injury and inflammatory response, as indicated by epithelial-mesenchymal transition (EMT), significant thickening of alveolar septa, infiltration of inflammatory cells and excessive mucus production. The transcriptome analysis revealed many more up-regulated genes in exposure groups when compared with the control group, and these genes were significantly enriched in the GO term of extracellular exosome, proteolysis, and regulation of circadian rhythm. The study discovered the induction of seven genes (CRY2, CIART, CREM, NR1D1, NR1D2, PER1 and PER3) that encode repressors of circadian clock. One gene (ARNTL) that encodes activator of circadian clock was down-regulated after ammonia exposure. The results of this study suggest that ammonia exposure disturbed the pulmonary circadian clock gene expression, which may establish new evidence for further understanding the toxicity of ammonia to lungs.
Collapse
Affiliation(s)
- Xiaotong Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mengyao Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuangzhao Chen
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Baoxin Wei
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yun Gao
- College of Engineering, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Longhui Huang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chun Liu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Tao Huang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mei Yu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shu-Hong Zhao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaoping Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
14
|
Sager TM, Umbright CM, Mustafa GM, Yanamala N, Leonard HD, McKinney WG, Kashon ML, Joseph P. Tobacco Smoke Exposure Exacerbated Crystalline Silica-Induced Lung Toxicity in Rats. Toxicol Sci 2020; 178:375-390. [PMID: 32976597 PMCID: PMC7825013 DOI: 10.1093/toxsci/kfaa146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Smoking may modify the lung response to silica exposure including cancer and silicosis. Nevertheless, the precise role of exposure to tobacco smoke (TS) on the lung response to crystalline silica (CS) exposure and the underlying mechanisms need further clarification. The objectives of the present study were to determine the role of TS on lung response to CS exposure and the underlying mechanism(s). Male Fischer 344 rats were exposed by inhalation to air, CS (15 mg/m3, 6 h/day, 5 days), TS (80 mg/m3, 3 h/day, twice weekly, 6 months), or CS (15 mg/m3, 6 h/day, 5 days) followed by TS (80 mg/m3, 3 h/day, twice weekly, 6 months). The rats were euthanized 6 months and 3 weeks following initiation of the first exposure and the lung response was assessed. Silica exposure resulted in significant lung toxicity as evidenced by lung histological changes, enhanced neutrophil infiltration, increased lactate dehydrogenase levels, enhanced oxidant production, and increased cytokine levels. The TS exposure alone had only a minimal effect on these toxicity parameters. However, the combined exposure to TS and CS exacerbated the lung response, compared with TS or CS exposure alone. Global gene expression changes in the lungs correlated with the lung toxicity severity. Bioinformatic analysis of the gene expression data demonstrated significant enrichment in functions, pathways, and networks relevant to the response to CS exposure which correlated with the lung toxicity detected. Collectively our data demonstrated an exacerbation of CS-induced lung toxicity by TS exposure and the molecular mechanisms underlying the exacerbated toxicity.
Collapse
Affiliation(s)
- Tina M Sager
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Christina M Umbright
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Gul Mehnaz Mustafa
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Naveena Yanamala
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Howard D Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Walter G McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Michael L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| | - Pius Joseph
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, West Virginia 26505
| |
Collapse
|
15
|
Toprak Kanık E, Yilmaz O, Ozdogru E, Alper H, Ulman C, Kanık A, Simsek Y, Yuksel H. Relevance between clinical status and exhaled molecules related to neutrophilic inflammation in pediatric cystic fibrosis. J Breath Res 2020; 14:046007. [PMID: 31896101 DOI: 10.1088/1752-7163/ab670d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is characterized with chronic inflammation with neutrophil and related cytokines in airway secretions. We aimed to measure the levels of neutrophil related inflammatory markers as nitric oxide, IL-8, IL-17, leukotriene B4 and neutrophil elastase as well as e-cadherin in exhaled breath condensate (EBC), and to determine their relation with clinical findings. METHODS We consecutively enrolled cystic fibrosis patients into our clinics between the age of six and eighteen years who could cooperate for exhaled breath condensate to this case-control study (n = 30). The age and sex matched control group (n = 26) was enrolled. Spirometry was performed during the stable period and EBC samples were obtained for measurement of the markers. RESULTS The mean age of the subjects enrolled was 12.1(4.2) years and 40% were positive for P.Aeruginosa in sputum. Subjects who had P.Aeruginosa in sputum cultures had significantly lower FEV1, FVC and FEF 25/75 values compared to the ones without P.Aeruginosa (p = 0.002, p = 0.002 and p = 0.005 respectively). EBC neutrophil elastase levels were significantly higher in the CF patients compared to non-CF controls (3.11 ± 4.71 versus 0.90 ± 2.68, p = 0.04). Nitric oxide, IL-17, IL-8, e-cadherin, neutrophil elastase or leukotriene B4 levels in EBC of CF patients were not related to P.Aeruginosa s infection, FEV1 levels or hospital admission in the last year. CONCLUSION In our study, neutrophil elastase levels in EBC are higher in CF patients compared to non-CF controls. This is independent of acute infection and is evidence to the persistence of neutrophilic lung injury. However, EBC NO, IL-8, IL-17, e-cadherin, neutrophil elastase and leukotriene B4 levels as inflammatory markers, are not correlated with disease progression or clinical findings.
Collapse
Affiliation(s)
- E Toprak Kanık
- Celal Bayar University Medical Faculty, Pediatric Allergy and Pulmonology, Manisa, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Ahmad S, Saleem M, Riaz N, Lee YS, Diri R, Noor A, Almasri D, Bagalagel A, Elsebai MF. The Natural Polypeptides as Significant Elastase Inhibitors. Front Pharmacol 2020; 11:688. [PMID: 32581778 PMCID: PMC7291377 DOI: 10.3389/fphar.2020.00688] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Human neutrophil elastase (HNE) is a major cause of the destruction of tissues in cases of several different chronic andinflammatory diseases. Overexpression of the elastase enzyme plays a significant role in the pathogenesis of various diseases including chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome, rheumatoid arthritis, the rare disease cyclic hematopoiesis (or cyclic neutropenia), infections, sepsis, cystic fibrosis, myocardial ischemia/reperfusion injury and asthma, inflammation, and atherosclerosis. Human neutrophil elastase is secreted by human neutrophils due to different stimuli. Medicine-based inhibition of the over-activation of neutrophils or production and activity of elastase have been suggested to mend inflammatory diseases. Although the development of new elastase inhibitors is an essential strategy for treating the different inflammatory diseases, it has been a challenge to specifically target the activity of elastase because of its overlapping functions with those of other serine proteases. This review article highlights the reported natural polypeptides as potential inhibitors of elastase enzyme. The mechanism of action, structural features, and activity of the polypeptides have also been correlated wherever they were available.
Collapse
Affiliation(s)
- Shabir Ahmad
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.,Department of Chemistry, Post-Graduate College, Bahawalpur, Pakistan
| | - Muhammad Saleem
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Naheed Riaz
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Yong Sup Lee
- Department of Life and Nanopharmaceutical Sciences & Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Reem Diri
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Noor
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Diena Almasri
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alaa Bagalagel
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Fahmi Elsebai
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.,Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
17
|
Giacalone VD, Dobosh BS, Gaggar A, Tirouvanziam R, Margaroli C. Immunomodulation in Cystic Fibrosis: Why and How? Int J Mol Sci 2020; 21:ijms21093331. [PMID: 32397175 PMCID: PMC7247557 DOI: 10.3390/ijms21093331] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/09/2023] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by unconventional mechanisms of inflammation, implicating a chronic immune response dominated by innate immune cells. Historically, therapeutic development has focused on the mutated cystic fibrosis transmembrane conductance regulator (CFTR), leading to the discovery of small molecules aiming at modulating and potentiating the presence and activity of CFTR at the plasma membrane. However, treatment burden sustained by CF patients, side effects of current medications, and recent advances in other therapeutic areas have highlighted the need to develop novel disease targeting of the inflammatory component driving CF lung damage. Furthermore, current issues with standard treatment emphasize the need for directed lung therapies that could minimize systemic side effects. Here, we summarize current treatment used to target immune cells in the lungs, and highlight potential benefits and caveats of novel therapeutic strategies.
Collapse
Affiliation(s)
- Vincent D. Giacalone
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Brian S. Dobosh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.G.); (C.M.)
- Pulmonary Section, Birmingham VA Medical Center, Birmingham, AL 35233, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Correspondence:
| | - Camilla Margaroli
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.G.); (C.M.)
| |
Collapse
|
18
|
Margaroli C, Garratt LW, Horati H, Dittrich AS, Rosenow T, Montgomery ST, Frey DL, Brown MR, Schultz C, Guglani L, Kicic A, Peng L, Scholte BJ, Mall MA, Janssens HM, Stick SM, Tirouvanziam R. Elastase Exocytosis by Airway Neutrophils Is Associated with Early Lung Damage in Children with Cystic Fibrosis. Am J Respir Crit Care Med 2020; 199:873-881. [PMID: 30281324 DOI: 10.1164/rccm.201803-0442oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RATIONALE Neutrophils are recruited to the airways of individuals with cystic fibrosis (CF). In adolescents and adults with CF, airway neutrophils actively exocytose the primary granule protease elastase (NE), whose extracellular activity correlates with lung damage. During childhood, free extracellular NE activity is measurable only in a subset of patients, and the exocytic function of airway neutrophils is unknown. OBJECTIVES To measure NE exocytosis by airway neutrophils in relation to free extracellular NE activity and lung damage in children with CF. METHODS We measured lung damage using chest computed tomography coupled with the Perth-Rotterdam Annotated Grid Morphometric Analysis for Cystic Fibrosis scoring system. Concomitantly, we phenotyped blood and BAL fluid leukocytes by flow and image cytometry, and measured free extracellular NE activity using spectrophotometric and Förster resonance energy transfer assays. Children with airway inflammation linked to aerodigestive disorder were enrolled as control subjects. MEASUREMENTS AND MAIN RESULTS Children with CF but not disease control children harbored BAL fluid neutrophils with high exocytosis of primary granules, before the detection of bronchiectasis. This measure of NE exocytosis correlated with lung damage (R = 0.55; P = 0.0008), whereas the molecular measure of free extracellular NE activity did not. This discrepancy may be caused by the inhibition of extracellular NE by BAL fluid antiproteases and its binding to leukocytes. CONCLUSIONS NE exocytosis by airway neutrophils occurs in all children with CF, and its cellular measure correlates with early lung damage. These findings implicate live airway neutrophils in early CF pathogenesis, which should instruct biomarker development and antiinflammatory therapy in children with CF.
Collapse
Affiliation(s)
- Camilla Margaroli
- 1 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,2 Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia
| | | | - Hamed Horati
- 4 Department of Pediatric Pulmonology, Erasmus University Medical Center/Sophia Children's Hospital, Rotterdam, the Netherlands
| | - A Susanne Dittrich
- 5 Department of Translational Pulmonology, Translational Lung Research Center, German Center for Lung Research, and.,6 Department of Pulmonology, and Critical Care Medicine, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | | | | | - Dario L Frey
- 5 Department of Translational Pulmonology, Translational Lung Research Center, German Center for Lung Research, and
| | - Milton R Brown
- 1 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,2 Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Carsten Schultz
- 7 Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Lokesh Guglani
- 1 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,2 Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Anthony Kicic
- 3 Telethon Kids Institute, Perth, Australia.,8 Department of Respiratory Medicine, Perth Children's Hospital, Perth, Western Australia, Australia.,9 Faculty of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Limin Peng
- 10 Department of Biostatistics, Emory University School of Public Health, Atlanta, Georgia
| | - Bob J Scholte
- 4 Department of Pediatric Pulmonology, Erasmus University Medical Center/Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Marcus A Mall
- 5 Department of Translational Pulmonology, Translational Lung Research Center, German Center for Lung Research, and.,11 Berlin Institute of Health, Berlin, Germany; and.,12 Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité-Universitätmedizin Berlin, Berlin, Germany
| | - Hettie M Janssens
- 4 Department of Pediatric Pulmonology, Erasmus University Medical Center/Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Stephen M Stick
- 3 Telethon Kids Institute, Perth, Australia.,8 Department of Respiratory Medicine, Perth Children's Hospital, Perth, Western Australia, Australia.,9 Faculty of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Rabindra Tirouvanziam
- 1 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,2 Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
19
|
Mejías JC, Forrest OA, Margaroli C, Frey Rubio DA, Viera L, Li J, Xu X, Gaggar A, Tirouvanziam R, Roy K. Neutrophil-targeted, protease-activated pulmonary drug delivery blocks airway and systemic inflammation. JCI Insight 2019; 4:131468. [PMID: 31661469 DOI: 10.1172/jci.insight.131468] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Pulmonary drug delivery presents a unique opportunity to target lower airway inflammation, which is often characterized by the massive recruitment of neutrophils from blood. However, specific therapies are lacking modulation of airway neutrophil function, and difficult challenges must be overcome to achieve therapeutic efficacy against pulmonary inflammation, notably drug hydrophobicity, mucociliary and macrophage-dependent clearance, and high extracellular protease burden. Here, we present a multistage, aerodynamically favorable delivery platform that uses extracellular proteolysis to its advantage to deliver nanoparticle-embedded hydrophobic drugs to neutrophils within the lower airways. Our design consists of a self-regulated nanoparticle-in-microgel system, in which microgel activation is triggered by extracellular elastase (degranulated by inflammatory neutrophils), and nanoparticles are loaded with Nexinhib20, a potent neutrophil degranulation inhibitor. Successful in vivo delivery of Nexinhib20 to the airways and into neutrophils promoted resolution of the inflammatory response by dampening neutrophil recruitment and degranulation, proinflammatory cytokine production in both airway and systemic compartments, as well as the presence of neutrophil-derived pathological extracellular vesicles in the lung fluid. Our findings showcase a new platform that overcomes challenges in pulmonary drug delivery and allows customization to match the proteolytic footprint of given diseases.
Collapse
Affiliation(s)
- Joscelyn C Mejías
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,National Science Foundation (NSF) Engineering Research Center for Cell Manufacturing Technologies, Atlanta, Georgia, USA
| | - Osric A Forrest
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.,Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.,Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - David A Frey Rubio
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,National Science Foundation (NSF) Engineering Research Center for Cell Manufacturing Technologies, Atlanta, Georgia, USA
| | - Liliana Viera
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jindong Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xin Xu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.,Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,National Science Foundation (NSF) Engineering Research Center for Cell Manufacturing Technologies, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Mejías JC, Roy K. In-vitro and in-vivo characterization of a multi-stage enzyme-responsive nanoparticle-in-microgel pulmonary drug delivery system. J Control Release 2019; 316:393-403. [PMID: 31715279 DOI: 10.1016/j.jconrel.2019.09.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/29/2019] [Indexed: 12/19/2022]
Abstract
Although the lung is an obvious target for site-specific delivery of many therapeutics for respiratory airway diseases such as asthma, COPD, and cystic fibrosis, novel strategies are needed to avoid key physiologic barriers for efficient delivery and controlled release of therapeutics to the lungs. Specifically, deposition into the deep lung requires particles with a 1-5μm aerodynamic diameter; however, particles with a geometric diameter less than 6μm are rapidly cleared by alveolar macrophages. Additionally, epithelial, endothelial, and fibroblast cells prefer smaller (< 300nm) nanoparticles for efficient endocytosis. Here we address these contradictory design requirements by using a nanoparticle-inside-microgel system (Nano-in-Microgel). Using an improved maleimide-thiol based Michael Addition during (water-in-oil) Emulsion (MADE) method, we fabricated both trypsin-responsive and neutrophil elastase-responsive polymeric Nano-in-Microgel to show the versatility of the system in easily exchanging enzyme-responsive crosslinkers for disease-specific proteases. By varying the initial macromer concentration, from 20 to 50% w/v, the size distribution means ranged from 4-8μm, enzymatic degradation of the microgels is within 30min, and in vitro macrophage phagocytosis is lower for the higher % w/v. We further demonstrated that in vivo lung delivery of the multi-stage carriers through the pulmonary route yields particle retention up to several hours and followed by clearance within in naïve mice. Our results provide a further understanding of how enzymatically-degradable multi-stage polymeric carriers can be used for pulmonary drug delivery.
Collapse
Affiliation(s)
- Joscelyn C Mejías
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
21
|
Castellani S, D'Oria S, Diana A, Polizzi AM, Di Gioia S, Mariggiò MA, Guerra L, Favia M, Vinella A, Leonetti G, De Venuto D, Gallo C, Montemurro P, Conese M. G-CSF and GM-CSF Modify Neutrophil Functions at Concentrations found in Cystic Fibrosis. Sci Rep 2019; 9:12937. [PMID: 31506515 PMCID: PMC6736848 DOI: 10.1038/s41598-019-49419-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023] Open
Abstract
The role of colony stimulating factors (CSFs) in cystic fibrosis (CF) circulating neutrophils has not been thoroughly evaluated, considering that the neutrophil burden of lung inflammation in these subjects is very high. The aim of this study was to assess granulocyte-CSF (G-CSF) and granulocyte-macrophage-CSF (GM-CSF) levels in CF patients in various clinical conditions and how these cytokines impact on activation and priming of neutrophils. G-CSF and GM-CSF levels were measured in sputum and serum samples of stable CF patients (n = 21) and in CF patients with acute exacerbation before and after a course of antibiotic therapy (n = 19). CSFs were tested on non CF neutrophils to investigate their effects on reactive oxygen species (ROS) production, degranulation (CD66b, elastase, lactoferrin, MMP-9), and chemotaxis. At very low concentrations found in CF patients (0.005–0.1 ng/ml), both cytokines inhibited ROS production, while higher concentrations (1–5 ng/ml) exerted a stimulatory effect. While either CSF induced elastase and MMP-9 secretion, lactoferrin levels were increased only by G-CSF. Chemotaxis was inhibited by GM-CSF, but was increased by G-CSF. However, when present together at low concentrations, CSFs increased basal and fMLP-stimulated ROS production and chemotaxis. These results suggest the CSF levels that circulating neutrophils face before extravasating into the lungs of CF patients may enhance their function contributing to the airway damage.
Collapse
Affiliation(s)
- Stefano Castellani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Susanna D'Oria
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Anna Diana
- Department of Biomedical Sciences and Human Oncology, Section of Medical Genetics, University of Bari, Bari, Italy
| | - Angela Maria Polizzi
- Department of Biomedical Sciences and Human Oncology, Section of Medical Genetics, University of Bari, Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maria Addolorata Mariggiò
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Angela Vinella
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Giuseppina Leonetti
- Cystic Fibrosis Regional Center, Department of Biomedical and Human Oncology, Pediatrics Section, U.O. "B. Trambusti", Policlinico, University of Bari, Bari, Italy
| | - Domenica De Venuto
- Cystic Fibrosis Regional Center, Department of Biomedical and Human Oncology, Pediatrics Section, U.O. "B. Trambusti", Policlinico, University of Bari, Bari, Italy
| | - Crescenzio Gallo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pasqualina Montemurro
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy.
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
22
|
SuFEx-enabled, agnostic discovery of covalent inhibitors of human neutrophil elastase. Proc Natl Acad Sci U S A 2019; 116:18808-18814. [PMID: 31484779 DOI: 10.1073/pnas.1909972116] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sulfur fluoride exchange (SuFEx) has emerged as the new generation of click chemistry. We report here a SuFEx-enabled, agnostic approach for the discovery and optimization of covalent inhibitors of human neutrophil elastase (hNE). Evaluation of our ever-growing collection of SuFExable compounds toward various biological assays unexpectedly revealed a selective and covalent hNE inhibitor: benzene-1,2-disulfonyl fluoride. Synthetic derivatization of the initial hit led to a more potent agent, 2-(fluorosulfonyl)phenyl fluorosulfate with IC50 0.24 μM and greater than 833-fold selectivity over the homologous neutrophil serine protease, cathepsin G. The optimized, yet simple benzenoid probe only modified active hNE and not its denatured form.
Collapse
|
23
|
Bhattacharjee O, Ayyangar U, Kurbet AS, Ashok D, Raghavan S. Unraveling the ECM-Immune Cell Crosstalk in Skin Diseases. Front Cell Dev Biol 2019; 7:68. [PMID: 31134198 PMCID: PMC6514232 DOI: 10.3389/fcell.2019.00068] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/09/2019] [Indexed: 01/06/2023] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and proteoglycans secreted by keratinocytes, fibroblasts and immune cells. The function of the skin ECM has expanded from being a scaffold that provides structural integrity, to a more dynamic entity that is constantly remodeled to maintain tissue homeostasis. The ECM functions as ligands for cell surface receptors such as integrins, dystroglycans, and toll-like receptors (TLRs) and regulate cellular signaling and immune cell dynamics. The ECM also acts as a sink for growth factors and cytokines, providing critical cues during epithelial morphogenesis. Dysregulation in the organization and deposition of ECMs lead to a plethora of pathophysiological conditions that are exacerbated by aberrant ECM-immune cell interactions. In this review, we focus on the interplay between ECM and immune cells in the context of skin diseases and also discuss state of the art therapies that target the key molecular players involved.
Collapse
Affiliation(s)
- Oindrila Bhattacharjee
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Uttkarsh Ayyangar
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Ambika S. Kurbet
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Driti Ashok
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Srikala Raghavan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| |
Collapse
|
24
|
Sesma JI, Wu B, Stuhlmiller TJ, Scott DW. SPX-101 is stable in and retains function after exposure to cystic fibrosis sputum. J Cyst Fibros 2019; 18:244-250. [DOI: 10.1016/j.jcf.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/16/2022]
|
25
|
Integration of Genome-Wide DNA Methylation and Transcription Uncovered Aberrant Methylation-Regulated Genes and Pathways in the Peripheral Blood Mononuclear Cells of Systemic Sclerosis. Int J Rheumatol 2018; 2018:7342472. [PMID: 30245726 PMCID: PMC6139224 DOI: 10.1155/2018/7342472] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/16/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
Objective. Systemic sclerosis (SSc) is a systemic connective tissue disease of unknown etiology. Aberrant gene expression and epigenetic modifications in circulating immune cells have been implicated in the pathogenesis of SSc. This study is to delineate the interaction network between gene transcription and DNA methylation in PBMC of SSc patients and to identify methylation-regulated genes which are involved in the pathogenesis of SSc. Methods. Genome-wide mRNA transcription and global DNA methylation analysis were performed on PBMC from 18 SSc patients and 19 matched normal controls (NC) using Illumina BeadChips. Differentially expressed genes (DEGs) and differentially methylated positions (DMPs) were integrative analyzed to identify methylation-regulated genes and associated molecular pathways. Results. Transcriptome analysis distinguished 453 DEGs (269 up- and 184 downregulated) in SSc from NC. Global DNA methylation analysis identified 925 DMPs located on 618 genes. Integration of the two lists revealed only 20 DEGs which harbor inversely correlated DMPs, including 12 upregulated (ELANE, CTSG, LTBR, C3AR1, CSTA, SPI1, ODF3B, SAMD4A, PLAUR, NFE2, ZYX, and CTSZ) and eight downregulated genes (RUNX3, PRF1, PRKCH, PAG1, RASSF5, FYN, CXCR6, and F2R). These potential methylation-regulated DEGs (MeDEGs) are enriched in the pathways related to immune cell migration, proliferation, activation, and inflammation activities. Using a machine learning algorism, we identified six out of the 20 MeDEGs, including F2R, CXCR6, FYN, LTBR, CTSG, and ELANE, which distinguished SSc from NC with 100% accuracy. Four genes (F2R, FYN, PAG1, and PRKCH) differentially expressed in SSc with interstitial lung disease (ILD) compared to SSc without ILD. Conclusion. The identified MeDEGs may represent novel candidate factors which lead to the abnormal activation of immune regulatory pathways in the pathogenesis of SSc. They may also be used as diagnostic biomarkers for SSc and clinical complications.
Collapse
|
26
|
Ringholz FC, Higgins G, Hatton A, Sassi A, Moukachar A, Fustero-Torre C, Hollenhorst M, Sermet-Gaudelus I, Harvey BJ, McNally P, Urbach V. Resolvin D1 regulates epithelial ion transport and inflammation in cystic fibrosis airways. J Cyst Fibros 2018; 17:607-615. [DOI: 10.1016/j.jcf.2017.11.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022]
|
27
|
Giovannoni MP, Schepetkin IA, Quinn MT, Cantini N, Crocetti L, Guerrini G, Iacovone A, Paoli P, Rossi P, Bartolucci G, Menicatti M, Vergelli C. Synthesis, biological evaluation, and molecular modelling studies of potent human neutrophil elastase (HNE) inhibitors. J Enzyme Inhib Med Chem 2018; 33:1108-1124. [PMID: 29969929 PMCID: PMC6032016 DOI: 10.1080/14756366.2018.1480615] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We report the synthesis and biological evaluation of a new series of 3- or 4-(substituted)phenylisoxazolones as HNE inhibitors. Due to tautomerism of the isoxazolone nucleus, two isomers were obtained as final compounds (2-NCO and 5-OCO) and the 2-NCO derivatives were the most potent with IC50 values in the nanomolar range (20–70 nM). Kinetic experiments indicated that 2-NCO 7d and 5-OCO 8d are both competitive HNE inhibitors. Molecular modelling on 7d and 8d suggests for the latter a more crowded region about the site of the nucleophilic attack, which could explain its lowered activity. In addition molecular dynamics (MD) simulations showed that the isomer 8d appears more prone to form H-bond interactions which, however, keep the reactive sites quite distant for the attack by Ser195. By contrast the amide 7d appears more mobile within the active pocket, since it makes single H-bond interactions affording a favourable orientation for the nucleophilic attack.
Collapse
Affiliation(s)
- Maria Paola Giovannoni
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Igor A Schepetkin
- b Department of Microbiology and Immunology , Montana State University , Bozeman , MT , USA
| | - Mark T Quinn
- b Department of Microbiology and Immunology , Montana State University , Bozeman , MT , USA
| | - Niccolò Cantini
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Letizia Crocetti
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Gabriella Guerrini
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Antonella Iacovone
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Paola Paoli
- c Department of Industrial Engineering , University of Florence , Florence , Italy
| | - Patrizia Rossi
- c Department of Industrial Engineering , University of Florence , Florence , Italy
| | - Gianluca Bartolucci
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Marta Menicatti
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| | - Claudia Vergelli
- a NEUROFARBA, Pharmaceutical and Nutraceutical Section , University of Florence , Sesto Fiorentino , Italy
| |
Collapse
|
28
|
Afratis NA, Klepfish M, Karamanos NK, Sagi I. The apparent competitive action of ECM proteases and cross-linking enzymes during fibrosis: Applications to drug discovery. Adv Drug Deliv Rev 2018; 129:4-15. [PMID: 29627371 DOI: 10.1016/j.addr.2018.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/11/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Progressive loss of organ function in most organs is associated with fibrosis, a tissue state associated with abnormal matrix buildup. If highly progressive, the fibrotic process eventually leads to organ failure and death. Fibrosis is a basic connective tissue lesion defined by the increase in the amount of fibrillar extracellular matrix (ECM) components in a tissue or organ. In addition, intrinsic changes in important structural cells can induce the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. ECM enzymes belonging to the family of matrix metalloproteinases (MMPs) and lysyl oxidases (LOXs) play a crucial role in ECM remodeling and regeneration. MMPs have a catalytic role in degradation of ECM, whereas LOX/LOXLs mediate ECM, especially collagen, cross-linking and stiffening. Importantly, enzymes from both families are elevated during the fibrotic response to tissue injury and its resolution. Yet, the apparent molecular competition or antagonistic activities of these enzyme families during the various stages of fibrosis is often overlooked. In this review, we discuss the diverse roles of MMPs and LOX/LOXL2 in chronic organ fibrosis. Finally, we review contemporary therapeutic strategies for fibrosis treatment, based on neutralization of MMP and LOX activity, as well as the development of novel drug delivery approaches.
Collapse
Affiliation(s)
- Nikolaos A Afratis
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mordehay Klepfish
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26500, Greece
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
29
|
Dittrich AS, Kühbandner I, Gehrig S, Rickert-Zacharias V, Twigg M, Wege S, Taggart CC, Herth F, Schultz C, Mall MA. Elastase activity on sputum neutrophils correlates with severity of lung disease in cystic fibrosis. Eur Respir J 2018; 51:13993003.01910-2017. [PMID: 29545279 DOI: 10.1183/13993003.01910-2017] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 02/14/2018] [Indexed: 01/06/2023]
Abstract
Neutrophil elastase (NE) is a key risk factor for severity of cystic fibrosis (CF) lung disease. Recent studies identified increased NE activity on the surface of airway neutrophils from CF-like mice and patients with CF. However, the role of surface-bound NE in CF lung disease remains unknown. We determined the relationship between surface-bound NE activity and severity of lung disease in CF.Surface-bound NE activity was measured on sputum neutrophils from 35 CF patients and eight healthy controls using novel lipidated Förster resonance energy transfer reporters and correlated with free NE activity, neutrophil counts, interleukin-8, myeloperoxidase and antiproteases in sputum supernatant, and with lung function parameters.Surface-bound NE activity was increased in CF compared to healthy controls (p<0.01) and correlated with free NE activity (p<0.05) and other inflammation markers (p<0.001). Surface-bound and free NE activity correlated with forced expiratory volume in 1 s % predicted (p<0.01 and p<0.05), but only surface-bound NE activity correlated with plethysmographic functional residual capacity % pred (p<0.01) in patients with CF.We demonstrate that surface-bound NE activity on airway neutrophils correlates with severity of lung disease in patients with CF. Our results suggest that surface-bound NE activity may play an important role in the pathogenesis and serve as novel biomarker in CF lung disease.
Collapse
Affiliation(s)
- A Susanne Dittrich
- Dept of Translational Pulmonology and Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Centre, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Dept of Pneumology and Critical Care Medicine, Thoraxklinik at the University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Iris Kühbandner
- Dept of Translational Pulmonology and Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Centre, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Dept of Pneumology and Critical Care Medicine, Thoraxklinik at the University Hospital Heidelberg, Heidelberg, Germany
| | - Stefanie Gehrig
- Dept of Translational Pulmonology and Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Centre, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Verena Rickert-Zacharias
- Dept of Translational Pulmonology and Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Centre, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Matthew Twigg
- Airway Innate Immunity Group (AiIR), Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Sabine Wege
- Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Dept of Pneumology and Critical Care Medicine, Thoraxklinik at the University Hospital Heidelberg, Heidelberg, Germany
| | - Clifford C Taggart
- Airway Innate Immunity Group (AiIR), Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Felix Herth
- Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Dept of Pneumology and Critical Care Medicine, Thoraxklinik at the University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Schultz
- Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Marcus A Mall
- Dept of Translational Pulmonology and Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Centre, University of Heidelberg, Heidelberg, Germany .,Translational Lung Research Centre Heidelberg (TLRC), German Centre for Lung Research (DZL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany.,Dept of Paediatric Pulmonology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
30
|
Bodas M, Mazur S, Min T, Vij N. Inhibition of histone-deacetylase activity rescues inflammatory cystic fibrosis lung disease by modulating innate and adaptive immune responses. Respir Res 2018; 19:2. [PMID: 29301535 PMCID: PMC5755330 DOI: 10.1186/s12931-017-0705-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Background Chronic lung disease resulting from dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) and NFκB-mediated neutrophilic-inflammation forms the basis of CF-related mortality. Here we aimed to evaluate if HDAC inhibition controls Pseudomonas-aeruginosa-lipopolysaccharide (Pa-LPS) induced airway inflammation and CF-lung disease. Methods For in vitro experiments, HEK293-cells were transfected with IL-8 or NFκB-firefly luciferase, and SV40-renilla- luciferase reporter constructs or ΔF508-CFTR-pCEP, followed by treatment with suberoylanilide hydroxamic acid (SAHA), Trichostatin-A (TSA) and/or TNFα. For murine studies, Cftr+/+ or Cftr−/− mice (n = 3) were injected/instilled with Pa-LPS and/or treated with SAHA or vehicle control. The progression of lung disease was monitored by quantifying changes in inflammatory markers (NFκB), cytokines (IL-6/IL-10), neutrophil activity (MPO, myeloperoxidase and/or NIMP-R14) and T-reg numbers. Results SAHA treatment significantly (p < 0.05) suppresses TNFα-induced NFκB and IL-8 reporter activities in HEK293-cells. Moreover, SAHA, Tubacin (selective HDAC6-inhibitor) or HDAC6-shRNAs controls CSE-induced ER-stress activities (p < 0.05). In addition, SAHA restores trafficking of misfolded-ΔF508-CFTR, by inducing protein levels of both B and C forms of CFTR. Murine studies using Cftr+/+ or Cftr−/− mice verified that SAHA controls Pa-LPS induced IL-6 levels, and neutrophil (MPO levels and/or NIMP-R14), NFκB-(inflammation) and Nrf2 (oxidative-stress marker) activities, while promoting FoxP3+ T-reg activity. Conclusion In summary, SAHA-mediated HDAC inhibition modulates innate and adaptive immune responses involved in pathogenesis and progression of inflammatory CF-lung disease. Electronic supplementary material The online version of this article (10.1186/s12931-017-0705-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA.,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven Mazur
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Integrated Research Facility at Fort Detrick, Fort Detrick, Frederick, MD, USA
| | - Taehong Min
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Genentech, 1 DNA Way, San Francisco, CA, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA. .,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,VIJ Biotech LLC, Baltimore, Maryland, USA.
| |
Collapse
|
31
|
Hägglund S, Blodörn K, Näslund K, Vargmar K, Lind SB, Mi J, Araínga M, Riffault S, Taylor G, Pringle J, Valarcher JF. Proteome analysis of bronchoalveolar lavage from calves infected with bovine respiratory syncytial virus-Insights in pathogenesis and perspectives for new treatments. PLoS One 2017; 12:e0186594. [PMID: 29036182 PMCID: PMC5643112 DOI: 10.1371/journal.pone.0186594] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
Human and bovine respiratory syncytial viruses (HRSV/BRSV) are major causes of severe lower respiratory tract infections in children and calves, respectively. Shared epidemiological, clinical, pathological and genetic characteristics of these viruses make comparative research highly relevant. To characterise the host response against BRSV infection, bronchoalveolar lavage supernatant (BAL) from i) non-vaccinated, BRSV-infected ii) vaccinated, BRSV-infected and iii) non-infected calves was analysed by tandem mass spectrometry. Proteins were semi-quantified and protein expression was validated by immunoblotting. Correlations between selected proteins and pathology, clinical signs and virus shedding were investigated. Calves with BRSV-induced disease had increased total protein concentrations and a decreased number of proteins identified in BAL. The protein profile was characterised by neutrophil activation and a reduction in identified antioxidant enzymes. The presence of neutrophils in alveolar septa, the expression level of neutrophil-related or antioxidant proteins and LZTFL1 correlated significantly with disease. Citrullinated histone 3, an indicator of extracellular traps (ETs), was only detected in non-vaccinated, BRSV-infected animals. By bringing disequilibrium in the release and detoxification of reactive oxygen species, generating ETs and causing elastine degradation, exaggerated neutrophil responses might exacerbate RSV-induced disease. Neutrophil-mitigating or antioxidant treatments should be further explored.
Collapse
Affiliation(s)
- Sara Hägglund
- Swedish University of Agricultural Sciences, Host Pathogen Interaction Group, Dept. of Clinical Sciences, Uppsala, Sweden
- * E-mail:
| | - Krister Blodörn
- Swedish University of Agricultural Sciences, Host Pathogen Interaction Group, Dept. of Clinical Sciences, Uppsala, Sweden
| | - Katarina Näslund
- Swedish University of Agricultural Sciences, Host Pathogen Interaction Group, Dept. of Clinical Sciences, Uppsala, Sweden
| | - Karin Vargmar
- Swedish University of Agricultural Sciences, Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Uppsala, Sweden
| | - Sara Bergström Lind
- Uppsala University, Science for Life Laboratory, Analytical Chemistry, Department of Chemistry-BMC, Uppsala, Sweden
| | - Jia Mi
- Uppsala University, Science for Life Laboratory, Analytical Chemistry, Department of Chemistry-BMC, Uppsala, Sweden
- Binzhou Medical University, Medicine and Pharmarcy Research Center, Yantai, China
| | - Mariluz Araínga
- University of Nebraska Medical Center (UNMC), Omaha, Nebraska, United States of America
| | - Sabine Riffault
- INRA, Unité de Virologie et Immunologie Moléculaires, Université Paris-Saclay, Jouy-en-Josas, France
| | - Geraldine Taylor
- The Pirbright Institute Ash Road, Pirbright, Surrey, United Kingdom
| | - John Pringle
- Swedish University of Agricultural Sciences, Host Pathogen Interaction Group, Dept. of Clinical Sciences, Uppsala, Sweden
| | - Jean François Valarcher
- Swedish University of Agricultural Sciences, Host Pathogen Interaction Group, Dept. of Clinical Sciences, Uppsala, Sweden
| |
Collapse
|
32
|
Chmelař J, Kotál J, Langhansová H, Kotsyfakis M. Protease Inhibitors in Tick Saliva: The Role of Serpins and Cystatins in Tick-host-Pathogen Interaction. Front Cell Infect Microbiol 2017; 7:216. [PMID: 28611951 PMCID: PMC5447049 DOI: 10.3389/fcimb.2017.00216] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/11/2017] [Indexed: 11/23/2022] Open
Abstract
The publication of the first tick sialome (salivary gland transcriptome) heralded a new era of research of tick protease inhibitors, which represent important constituents of the proteins secreted via tick saliva into the host. Three major groups of protease inhibitors are secreted into saliva: Kunitz inhibitors, serpins, and cystatins. Kunitz inhibitors are anti-hemostatic agents and tens of proteins with one or more Kunitz domains are known to block host coagulation and/or platelet aggregation. Serpins and cystatins are also anti-hemostatic effectors, but intriguingly, from the translational perspective, also act as pluripotent modulators of the host immune system. Here we focus especially on this latter aspect of protease inhibition by ticks and describe the current knowledge and data on secreted salivary serpins and cystatins and their role in tick-host-pathogen interaction triad. We also discuss the potential therapeutic use of tick protease inhibitors.
Collapse
Affiliation(s)
- Jindřich Chmelař
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia
| | - Jan Kotál
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| | - Helena Langhansová
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| |
Collapse
|
33
|
Airway mucus, inflammation and remodeling: emerging links in the pathogenesis of chronic lung diseases. Cell Tissue Res 2017; 367:537-550. [PMID: 28108847 DOI: 10.1007/s00441-016-2562-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
Airway mucus obstruction is a hallmark of many chronic lung diseases including rare genetic disorders such as cystic fibrosis (CF) and primary ciliary dyskinesia, as well as common lung diseases such as asthma and chronic obstructive pulmonary disease (COPD), which have emerged as a leading cause of morbidity and mortality worldwide. However, the role of excess airway mucus in the in vivo pathogenesis of these diseases remains poorly understood. The generation of mice with airway-specific overexpression of epithelial Na+ channels (ENaC), exhibiting airway surface dehydration (mucus hyperconcentration), impaired mucociliary clearance (MCC) and mucus plugging, led to a model of muco-obstructive lung disease that shares key features of CF and COPD. In this review, we summarize recent progress in the understanding of causes of impaired MCC and in vivo consequences of airway mucus obstruction that can be inferred from studies in βENaC-overexpressing mice. These studies confirm that mucus hyperconcentration on airway surfaces plays a critical role in the pathophysiology of impaired MCC, mucus adhesion and airway plugging that cause airflow obstruction and provide a nidus for bacterial infection. In addition, these studies support the emerging concept that excess airway mucus per se, probably via several mechanisms including hypoxic epithelial necrosis, retention of inhaled irritants or allergens, and potential immunomodulatory effects, is a potent trigger of chronic airway inflammation and associated lung damage, even in the absence of bacterial infection. Finally, these studies suggest that improvement of mucus clearance may be a promising therapeutic strategy for a spectrum of muco-obstructive lung diseases.
Collapse
|
34
|
Cystic fibrosis lung environment and Pseudomonas aeruginosa infection. BMC Pulm Med 2016; 16:174. [PMID: 27919253 PMCID: PMC5139081 DOI: 10.1186/s12890-016-0339-5] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/24/2016] [Indexed: 12/20/2022] Open
Abstract
Background The airways of patients with cystic fibrosis (CF) are highly complex, subject to various environmental conditions as well as a distinct microbiota. Pseudomonas aeruginosa is recognized as one of the most important pulmonary pathogens and the predominant cause of morbidity and mortality in CF. A multifarious interplay between the host, pathogens, microbiota, and the environment shapes the course of the disease. There have been several excellent reviews detailing CF pathology, Pseudomonas and the role of environment in CF but only a few reviews connect these entities with regards to influence on the overall course of the disease. A holistic understanding of contributing factors is pertinent to inform new research and therapeutics. Discussion In this article, we discuss the deterministic alterations in lung physiology as a result of CF. We also revisit the impact of those changes on the microbiota, with special emphasis on P. aeruginosa and the influence of other non-genetic factors on CF. Substantial past and current research on various genetic and non-genetic aspects of cystic fibrosis has been reviewed to assess the effect of different factors on CF pulmonary infection. A thorough review of contributing factors in CF and the alterations in lung physiology indicate that CF lung infection is multi-factorial with no isolated cause that should be solely targeted to control disease progression. A combinatorial approach may be required to ensure better disease outcomes. Conclusion CF lung infection is a complex disease and requires a broad multidisciplinary approach to improve CF disease outcomes. A holistic understanding of the underlying mechanisms and non-genetic contributing factors in CF is central to development of new and targeted therapeutic strategies.
Collapse
|
35
|
Ehrhardt H, Zimmer KP. The need for coordination of research activities in pediatric lung diseases. Mol Cell Pediatr 2016; 3:26. [PMID: 27465412 PMCID: PMC4963328 DOI: 10.1186/s40348-016-0060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 07/21/2016] [Indexed: 11/10/2022] Open
Affiliation(s)
- Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstr. 12, D-35392, Gießen, Germany.
| | - Klaus-Peter Zimmer
- Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstr. 12, D-35392, Gießen, Germany
| |
Collapse
|
36
|
Margaroli C, Tirouvanziam R. Neutrophil plasticity enables the development of pathological microenvironments: implications for cystic fibrosis airway disease. Mol Cell Pediatr 2016; 3:38. [PMID: 27868161 PMCID: PMC5136534 DOI: 10.1186/s40348-016-0066-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/04/2016] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The pathological course of several chronic inflammatory diseases, including cystic fibrosis, chronic obstructive pulmonary disease, and rheumatoid arthritis, features an aberrant innate immune response dominated by neutrophils. In cystic fibrosis, neutrophil burden and activity of neutrophil elastase in the extracellular fluid have been identified as strong predictors of lung disease severity. REVIEW Although neutrophils are generally considered to be rigid, pre-programmed effector leukocytes, recent studies suggest extensive plasticity in how neutrophil functions unfold upon recruitment to peripheral tissues, and how they choose their ultimate fate. Indeed, upon migration to cystic fibrosis airways, neutrophils display dysregulated lifespan, metabolic activation, and altered effector and regulatory functions, consistent with profound adaptation and phenotypic reprogramming. Licensed by signals present in cystic fibrosis airway microenvironment to survive and develop these novel functions, neutrophils orchestrate, in partnership with the epithelium and with the resident microbiota, the evolution of a pathological microenvironment. This microenvironment is defined by altered proteolytic, redox, and metabolic balance and the presence of stable luminal structures in which neutrophils and microbes coexist. CONCLUSIONS The elucidation of molecular mechanisms driving neutrophil plasticity in vivo will open new treatment opportunities designed to modulate, rather than block, the crucial adaptive functions fulfilled by neutrophils. This review aims to outline emerging mechanisms of neutrophil plasticity and their participation in the building of pathological microenvironments in the context of cystic fibrosis and other diseases with similar features.
Collapse
Affiliation(s)
- Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA.
| |
Collapse
|