1
|
Lalmanach G, Rigoux B, David A, Tahri-Joutey M, Lecaille F, Marchand-Adam S, Saidi A. Human cystatin C in fibrotic diseases. Clin Chim Acta 2024; 565:120016. [PMID: 39461496 DOI: 10.1016/j.cca.2024.120016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Human cystatin C (hCC), which has a pervasive distribution within body fluids and is ubiquitously expressed by numerous cells and tissues, is a highly potent extracellular inhibitor of cysteine proteases. Besides measurement of serum creatinine, which is the most widely used technique for appraising glomerular filtration rate (GFR), hCC has emerged as a relevant GFR biomarker, because its quantification in serum is less sensitive to interferences with factors such as age, muscle mass or diet. Moreover, there are growing body of evidence that hCC overexpression and/or oversecretion, which is primarily driven by TGF-β1, occur during fibrogenesis (cardiac, liver, oral, and lung fibrosis). Even though molecular mechanisms and signaling pathways governing the regulation of hCC remain to be deciphered more acutely, current data sustain that hCC expression relates to myofibrogenesis and that hCC could be a specific and valuable biomarker of fibrotic disease.
Collapse
Affiliation(s)
- Gilles Lalmanach
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France.
| | - Baptiste Rigoux
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Alexis David
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Mounia Tahri-Joutey
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Fabien Lecaille
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Sylvain Marchand-Adam
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France; The University Hospital Center of Tours (CHRU Tours), Pulmonology Department, Tours, France
| | - Ahlame Saidi
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| |
Collapse
|
2
|
Bioassay-Guided Alkaloids Isolation from Camellia sinensis and Colchicum luteum: In Silico and In Vitro Evaluations for Protease Inhibition. Molecules 2023; 28:molecules28062459. [PMID: 36985431 PMCID: PMC10058905 DOI: 10.3390/molecules28062459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023] Open
Abstract
Bioassay-guided isolation from Camellia sinensis (Theaceae) and Colchicum luteum (Liliaceae) utilizing an in vitro model of protease assay revealed colchicine (1) and caffeine (2) from chloroform fractions, respectively. Their structures were validated using spectral techniques. The purified compounds were further optimized with Gaussian software utilizing the B3LYP functional and 6-31G(d,p) basis set. The result files were utilized to determine several global reactivity characteristics to explain the diverse behavior of the compounds. Colchicine (1) showed a higher inhibition of protease activity (63.7 ± 0.5 %age with IC50 = 0.83 ± 0.07 mM), compared with caffeine (2) (39.2 ± 1.3 %age). In order to determine the type of inhibition, compound 1 was further studied, and, based on Lineweaver–Burk/Dixon plots and their secondary replots, it was depicted that compound 1 was a non-competitive inhibitor of this enzyme, with a Ki value of 0.690 ± 0.09 mM. To elucidate the theoretical features of protease inhibition, molecular docking studies were performed against serine protease (PDB #1S0Q), which demonstrated that compound 1 had a strong interaction with the different amino acid residues located on the active site of this understudied enzyme, with a high docking score of 16.2 kcal/mol.
Collapse
|
3
|
Chesseron S, Saidi A, Lecaille F, Lalmanach G, Bigot P. [Alteration of pulmonary epithelial permeability by cathepsin S in chronic obstructive pulmonary disease]. Rev Mal Respir 2023; 40:250-253. [PMID: 36828678 DOI: 10.1016/j.rmr.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 02/24/2023]
Abstract
Smoking is accountable for most of the chronic obstructive pulmonary disease (COPD) cases. COPD, which is characterized by the development of chronic bronchitis, could be associated with emphysema. In active smokers, there is an overexpression of cathepsin S, a cysteine protease, which participates in the development of emphysema via its elastinolytic activity. Likewise, we demonstrated that cathepsin S could degrade one or more protein constituents of cell junctions. This deleterious proteolytic activity leads to an alteration of the integrity of the lung epithelial barrier, which in turn could aggravate chronic inflammation and promote the exacerbation phases associated with infections.
Collapse
Affiliation(s)
- S Chesseron
- University of Tours, Tours, France; Inserm, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Mechanisms in Inflammation", 10, boulevard Tonnellé, 37032 Tours cedex, France
| | - A Saidi
- University of Tours, Tours, France; Inserm, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Mechanisms in Inflammation", 10, boulevard Tonnellé, 37032 Tours cedex, France
| | - F Lecaille
- University of Tours, Tours, France; Inserm, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Mechanisms in Inflammation", 10, boulevard Tonnellé, 37032 Tours cedex, France
| | - G Lalmanach
- University of Tours, Tours, France; Inserm, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Mechanisms in Inflammation", 10, boulevard Tonnellé, 37032 Tours cedex, France
| | - P Bigot
- University of Tours, Tours, France; Inserm, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Mechanisms in Inflammation", 10, boulevard Tonnellé, 37032 Tours cedex, France.
| |
Collapse
|
4
|
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
|
5
|
Wang K, Liao Y, Li X, Wang R, Zeng Z, Cheng M, Gao L, Xu D, Wen F, Wang T, Chen J. Inhibition of neutrophil elastase prevents cigarette smoke exposure-induced formation of neutrophil extracellular traps and improves lung function in a mouse model of chronic obstructive pulmonary disease. Int Immunopharmacol 2023; 114:109537. [PMID: 36495695 DOI: 10.1016/j.intimp.2022.109537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an important public health challenge worldwide, and is usually caused by significant exposure to noxious agents, particularly cigarette smoke. Recent studies have revealed that excessive production of neutrophil extracellular traps (NETs) in the airways is associated with disease severity in COPD patients. NETs are extracellular neutrophil-derived structures composed of chromatin fibers decorated with histones and granule proteases including neutrophil elastase (NE). However, the effective prevention of NET formation in COPD remains elusive. Here, we demonstrated that treatment with GW311616A, a potent and selective inhibitor of NE, prevented cigarette smoke extract (CSE)-induced NET formation in human neutrophils by blocking NE nuclear translocation and subsequent chromatin decondensation. Inhibition of NE also abrogated CSE-induced ROS production and migration impairment of neutrophils. Administration of GW311616A in vivo substantially reduced pulmonary generation of NETs while attenuating the key pathological changes in COPD, including airway leukocyte infiltration, mucus-secreting goblet cell hyperplasia, and emphysema-like alveolar destruction in a mouse model of COPD induced by chronic cigarette smoke exposure. Mice treated with GW311616A also showed significant attenuation of neutrophil numbers and percentages and the levels of neutrophil chemotactic factors (LTB4, KC, and CXCL5) and proinflammatory cytokines (IL-1β, and TNF-α) in bronchoalveolar lavage fluid compared to mice treated with cigarette smoke exposure only. Furthermore, GW311616A treatment considerably improved lung function in the COPD mouse model, including preventing the decline of FEV100/FVC and delta PEF as well as inhibiting the increase in FRC, TLC, and FRC/TLC. Overall, our study suggests that NE plays a critical role in cigarette smoke-induced NET formation by neutrophils and that inhibition of NE is a promising strategy to suppress NET-mediated pathophysiological changes in COPD.
Collapse
Affiliation(s)
- Ke Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yue Liao
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoou Li
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Ran Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zijian Zeng
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Mengxin Cheng
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Lijuan Gao
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Dan Xu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Fuqiang Wen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.
| | - Jun Chen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Bigot P, Chesseron S, Saidi A, Sizaret D, Parent C, Petit-Courty A, Courty Y, Lecaille F, Lalmanach G. Cleavage of Occludin by Cigarette Smoke-Elicited Cathepsin S Increases Permeability of Lung Epithelial Cells. Antioxidants (Basel) 2022; 12:antiox12010005. [PMID: 36670867 PMCID: PMC9854811 DOI: 10.3390/antiox12010005] [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: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an irreversible disease mainly caused by smoking. COPD is characterized by emphysema and chronic bronchitis associated with enhanced epithelial permeability. HYPOTHESIS Lung biopsies from smokers revealed a decreased expression level of occludin, which is a protein involved in the cohesion of epithelial tight junctions. Moreover, the occludin level correlated negatively with smoking history (pack-years), COPD grades, and cathepsin S (CatS) activity. Thus, we examined whether CatS could participate in the modulation of the integrity of human lung epithelial barriers. METHODS AND RESULTS Cigarette smoke extract (CSE) triggered the upregulation of CatS by THP-1 macrophages through the mTOR/TFEB signaling pathway. In a co-culture model, following the exposure of macrophages to CSE, an enhanced level of permeability of lung epithelial (16HBE and NHBE) cells towards FITC-Dextran was observed, which was associated with a decrease in occludin level. Similar results were obtained using 16HBE and NHBE cells cultured at the air-liquid interface. The treatment of THP-1 macrophages by CatS siRNAs or by a pharmacological inhibitor restored the barrier function of epithelial cells, suggesting that cigarette smoke-elicited CatS induced an alteration of epithelial integrity via the proteolytic injury of occludin. CONCLUSIONS Alongside its noteworthy resistance to oxidative stress induced by cigarette smoke oxidants and its deleterious elastin-degrading potency, CatS may also have a detrimental effect on the barrier function of epithelial cells through the cleavage of occludin. The obtained data emphasize the emerging role of CatS in smoking-related lung diseases and strengthen the relevance of targeting CatS in the treatment of emphysema and COPD.
Collapse
Affiliation(s)
- Paul Bigot
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Simon Chesseron
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Ahlame Saidi
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Damien Sizaret
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Pathological Anatomy and Cytology, The University Hospital Center of Tours, 37000 Tours, France
| | - Christelle Parent
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Aerosol therapy and Biotherapeutics for Respiratory Diseases”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Agnès Petit-Courty
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Yves Courty
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Fabien Lecaille
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Gilles Lalmanach
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
- Correspondence: ; Tel.: +33-2-47-36-61-51
| |
Collapse
|
7
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
8
|
Lecaille F, Chazeirat T, Saidi A, Lalmanach G. Cathepsin V: Molecular characteristics and significance in health and disease. Mol Aspects Med 2022; 88:101086. [PMID: 35305807 DOI: 10.1016/j.mam.2022.101086] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 12/31/2022]
Abstract
Human cysteine cathepsins form a family of eleven proteases (B, C, F, H, K, L, O, S, V, W, X/Z) that play important roles in a considerable number of biological and pathophysiological processes. Among them, cathepsin V, also known as cathepsin L2, is a lysosomal enzyme, which is mainly expressed in cornea, thymus, heart, brain, and skin. Cathepsin V is a multifunctional endopeptidase that is involved in both the release of antigenic peptides and the maturation of MHC class II molecules and participates in the turnover of elastin fibrils as well in the cleavage of intra- and extra-cellular substrates. Moreover, there is increasing evidence that cathepsin V may contribute to the progression of diverse diseases, due to the dysregulation of its expression and/or its activity. For instance, increased expression of cathepsin V is closely correlated with malignancies (breast cancer, squamous cell carcinoma, or colorectal cancer) as well vascular disorders (atherosclerosis, aortic aneurysm, hypertension) being the most prominent examples. This review aims to shed light on current knowledge on molecular aspects of cathepsin V (genomic organization, protein structure, substrate specificity), its regulation by protein and non-protein inhibitors as well to summarize its expression (tissue and cellular distribution). Then the core biological and pathophysiological roles of cathepsin V will be depicted, raising the question of its interest as a valuable target that can open up pioneering therapeutic avenues.
Collapse
Affiliation(s)
- Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| | - Thibault Chazeirat
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| |
Collapse
|
9
|
Wagner C, Balázs A, Schatterny J, Zhou-Suckow Z, Duerr J, Schultz C, Mall MA. Genetic Deletion of Mmp9 Does Not Reduce Airway Inflammation and Structural Lung Damage in Mice with Cystic Fibrosis-like Lung Disease. Int J Mol Sci 2022; 23:13405. [PMID: 36362203 PMCID: PMC9657231 DOI: 10.3390/ijms232113405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 09/10/2023] Open
Abstract
Elevated levels of matrix metalloprotease 9 (MMP-9) and neutrophil elastase (NE) are associated with bronchiectasis and lung function decline in patients with cystic fibrosis (CF). MMP-9 is a potent extracellular matrix-degrading enzyme which is activated by NE and has been implicated in structural lung damage in CF. However, the role of MMP-9 in the in vivo pathogenesis of CF lung disease is not well understood. Therefore, we used β-epithelial Na+ channel-overexpressing transgenic (βENaC-Tg) mice as a model of CF-like lung disease and determined the effect of genetic deletion of Mmp9 (Mmp9-/-) on key aspects of the pulmonary phenotype. We found that MMP-9 levels were elevated in the lungs of βENaC-Tg mice compared with wild-type littermates. Deletion of Mmp9 had no effect on spontaneous mortality, inflammatory markers in bronchoalveolar lavage, goblet cell metaplasia, mucus hypersecretion and emphysema-like structural lung damage, while it partially reduced mucus obstruction in βENaC-Tg mice. Further, lack of Mmp9 had no effect on increased inspiratory capacity and increased lung compliance in βENaC-Tg mice, whereas both lung function parameters were improved with genetic deletion of NE. We conclude that MMP-9 does not play a major role in the in vivo pathogenesis of CF-like lung disease in mice.
Collapse
Affiliation(s)
- Claudius Wagner
- Department of Translational Pulmonology, University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Anita Balázs
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jolanthe Schatterny
- Department of Translational Pulmonology, University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Zhe Zhou-Suckow
- Department of Translational Pulmonology, University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Julia Duerr
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Carsten Schultz
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
10
|
Kraposhina AY, Sobko EА, Demko IV, Kazmerchuk OV, Kacer AB, Abramov YI. The role of cathepsin S in the pathophysiology of bronchial asthma. BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-198-204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To date, the study of the role of proteases in the pathogenesis of various diseases remains relevant. The variety of cathepsin functions is associated with the peculiarities of their localization, expression, and regulation, due to which cathepsins are involved in development of many pathologies. Dysregulation of proteases, their inhibitors, and substrates can lead to the development of multiple organ dysfunction.The review presents data on the characteristics of the entire family of cathepsins and cathepsin S, in particular. The pathophysiological role of cathepsin S in the formation of bronchopulmonary pathologies, as well as in bronchial asthma is described, and intraand extracellular implementation mechanisms are considered. The authors believe it is this enzyme that could be targeted in targeted asthma therapy to prevent airway wall remodeling at the earliest stages of the disease. The literature search was carried out in the search engines Medline, eLibrary, Scopus, the Cochrane Library, and RSCI.
Collapse
Affiliation(s)
- A. Yu. Kraposhina
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | - E. А. Sobko
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | - I. V. Demko
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Krasnoyarsk Regional Clinical Hospital
| | | | - A. B. Kacer
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| | - Yu. I. Abramov
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| |
Collapse
|
11
|
The IRE1α-XBP1s Arm of the Unfolded Protein Response Activates N-Glycosylation to Remodel the Subepithelial Basement Membrane in Paramyxovirus Infection. Int J Mol Sci 2022; 23:ijms23169000. [PMID: 36012265 PMCID: PMC9408905 DOI: 10.3390/ijms23169000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract infections (LRTI) associated with decreased pulmonary function, asthma, and allergy. Recently, we demonstrated that RSV induces the hexosamine biosynthetic pathway via the unfolded protein response (UPR), which is a pathway controlling protein glycosylation and secretion of the extracellular matrix (ECM). Because the presence of matrix metalloproteinases and matricellular growth factors (TGF) is associated with severe LRTI, we studied the effect of RSV on ECM remodeling and found that RSV enhances the deposition of fibronectin-rich ECM by small airway epithelial cells in a manner highly dependent on the inositol requiring kinase (IRE1α)–XBP1 arm of the UPR. To understand this effect comprehensively, we applied pharmacoproteomics to understand the effect of the UPR on N-glycosylation and ECM secretion in RSV infection. We observe that RSV induces N-glycosylation and the secretion of proteins related to ECM organization, secretion, or proteins integral to plasma membranes, such as integrins, laminins, collagens, and ECM-modifying enzymes, in an IRE1α–XBP1 dependent manner. Using a murine paramyxovirus model that activates the UPR in vivo, we validate the IRE1α–XBP1-dependent secretion of ECM to alveolar space. This study extends understanding of the IRE1α–XBP1 pathway in regulating N-glycosylation coupled to structural remodeling of the epithelial basement membrane in RSV infection.
Collapse
|
12
|
Rickert-Zacharias V, Schultz M, Mall MA, Schultz C. Visualization of Ectopic Serine Protease Activity by Förster Resonance Energy Transfer-Based Reporters. ACS Chem Biol 2021; 16:2174-2184. [PMID: 34726893 DOI: 10.1021/acschembio.1c00168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Channel-activating proteases (CAPs) play a fundamental role in the regulation of sodium transport across epithelial tissues mainly via cleavage-mediated fine-tuning of the activity of the epithelial sodium channel (ENaC). Hyperactivity of CAPs and subsequently increased ENaC activity have been associated with various diseases, including cystic fibrosis (CF). To date, there is only a limited number of tools available to investigate CAP activity. Here, we developed ratiometric, peptide-based Förster resonance energy transfer (FRET) reporters useful to visualize and quantify the activity of ectopic serine proteases including the CAPs prostasin and matriptase in human and murine samples in a temporally and spatially resolved manner. Lipidated varieties were inserted into the outer leaflet of the plasma membrane to detect enzyme activity on the surface of individual cells, that is, close to the protease substrates. The FRET reporters (termed CAPRee) selectively detected the activity of ectopic serine proteases such as CAPs in solution and on the surface of human and murine cells. We found increased CAP activity on the surface of cells with a genetic background of CF. The new reporters will contribute to a better understanding of ectopic serine protease activity and their regulation under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Verena Rickert-Zacharias
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL) and University of Heidelberg, 69117 Heidelberg, Germany
- Faculty of Biosciences, Collaboration for Joint Ph.D. Degree between EMBL and Heidelberg University, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Madeleine Schultz
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Marcus A. Mall
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL) and University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité─Universitätsmedizin Berlin, 13353 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
- German Center for Lung Research (DZL), Associated
Partner Site, 13353 Berlin, Germany
| | - Carsten Schultz
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL) and University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon 97239, United States
| |
Collapse
|
13
|
Lepissier A, Addy C, Hayes K, Noel S, Bui S, Burgel PR, Dupont L, Eickmeier O, Fayon M, Leal T, Lopes C, Downey DG, Sermet-Gaudelus I. Inflammation biomarkers in sputum for clinical trials in cystic fibrosis: current understanding and gaps in knowledge. J Cyst Fibros 2021; 21:691-706. [PMID: 34772643 DOI: 10.1016/j.jcf.2021.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE Sputum biomarkers hold promise as a direct measure of inflammation within the cystic fibrosis (CF) lung, but variability in study design and sampling methodology have limited their use. A full evaluation of the reliability, validity and clinical relevance of individual biomarkers is required to optimise their use within CF clinical research. OBJECTIVES A biomarker Special Interest Working Group was established within the European Cystic Fibrosis Society-Clinical Trials Network Standardisation Committee, to perform a review of the evidence regarding sputum biomarkers in CF. METHODS From the 139 included articles, we identified 71 sputum biomarkers to undergo evaluation of their clinimetric properties, responsiveness, discriminant, concurrent and convergent validity. RESULTS Current evidence confirms the potential of sputum biomarkers as outcome measures in clinical trials. Inconsistency in responsiveness, concurrent and convergent validity require further research into these markers and processing standardisation before translation into wider use. Of the 71 biomarkers identified, Neutrophil Elastase (NE), IL-8, TNF-α and IL-1β, demonstrated validity and responsiveness to be currently considered for use in clinical trials. Other biomarkers show future promise, including IL-6, calprotectin, HMGB-1 and YKL-40. CONCLUSION A concerted international effort across the cystic fibrosis community is needed to promote high quality biomarker trial design, establish large population-based biomarker studies, and work together to create standards for collection, storage and analysis of sputum biomarkers.
Collapse
Affiliation(s)
- Agathe Lepissier
- Paediatric Center for Cystic Fibrosis, Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker Enfants Malades 149 rue de Sévres, Paris 75743, France; INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France; European Reference Network (ERN Lung)
| | - Charlotte Addy
- Northern Ireland Clinical Research Facility, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL; All Wales Adult Cystic Fibrosis Centre, University Hopsital Llandough, Penlan Road, CF64 2XX
| | - Kate Hayes
- Northern Ireland Clinical Research Facility, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Sabrina Noel
- INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France
| | - Stéphanie Bui
- Université de Bordeaux (INSERM U1045), CHU de Bordeaux, (CIC1401), F-33000 Bordeaux, France
| | - Pierre-Régis Burgel
- European Reference Network (ERN Lung); National Reference Cystic Fibrosis Center and Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, 75014, France; Institut Cochin, INSERM U1016 and Université de Paris; Paris 75014, France
| | - Lieven Dupont
- University Hospital Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | - Olaf Eickmeier
- Facharzt für Kinder- und Jugendmedizin, Universitätsklinikum Frankfurt a.M., Johann Wolfgang-Goethe-Universität, Allergologie, Pneumologie & Mukoviszidose, Theodor-Stern-Kai 7, 60590 Frankfurt/Main
| | - Michael Fayon
- Université de Bordeaux (INSERM U1045), CHU de Bordeaux, (CIC1401), F-33000 Bordeaux, France
| | - Teresinha Leal
- Louvain Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Carlos Lopes
- Departamento do Tórax, Hospital de Santa Maria, Lisbon
| | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Isabelle Sermet-Gaudelus
- Paediatric Center for Cystic Fibrosis, Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker Enfants Malades 149 rue de Sévres, Paris 75743, France; INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France; European Reference Network (ERN Lung); Service de Pneumologie et Allergologie Pédiatriques, Centre de Ressources et de Compétence de la Mucoviscidose, Hôpital Necker Enfants Malades 149 rue de Sévres, INSERM U1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris 75743, France.
| |
Collapse
|
14
|
Efimenko AY, Kalinina NI, Rubina KA, Semina EV, Sysoeva VY, Akopyan ZA, Tkachuk VA. Secretome of Multipotent Mesenchymal Stromal Cells as a Promising Treatment and for Rehabilitation of Patients with the Novel Coronaviral Infection. HERALD OF THE RUSSIAN ACADEMY OF SCIENCES 2021; 91:170-175. [PMID: 34131372 PMCID: PMC8192105 DOI: 10.1134/s101933162102012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 12/28/2020] [Accepted: 01/30/2021] [Indexed: 06/12/2023]
Abstract
As a rule, coronavirus infections are mild in healthy adults and do not require special approaches to treatment. However, highly pathogenic strains, particularly the recently isolated SARS-CoV2, which causes COVID-19 infection, in about 15% of cases lead to severe complications, including acute respiratory distress syndrome, which causes high patient mortality. In addition, a common complication of COVID-19 is the development of pulmonary fibrosis. Why is the novel coronavirus so pathogenic? What new treatments can be proposed to speed up the recovery and subsequent rehabilitation of the organism? In 2020, over 34 000 scientific articles were published on the structure, distribution, pathogenesis, and possible approaches to the treatment of infection caused by the novel SARS-CoV2 coronavirus. However, there are still no definitive answers to these questions, while the number of the diseased is increasing daily. One of the comprehensive approaches to the treatment of the consequences of the infection is the use of multipotent human mesenchymal stromal cells and products of their secretion (secretome). Acting at several stages of the development of the infection, the components of the secretome can suppress the interaction of the virus with endothelial cells, regulate inflammation, and stimulate lung tissue regeneration, preventing the development of fibrosis. The results of basic and clinical research on this topic are summarized, including our own experimental data, indicating that cell therapy approaches can be successfully applied to treat patients with COVID-19.
Collapse
Affiliation(s)
- A. Yu. Efimenko
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| | | | | | - E. V. Semina
- Moscow State University, Moscow, Russia
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | | | - Zh. A. Akopyan
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| | - V. A. Tkachuk
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| |
Collapse
|
15
|
Carroll EL, Bailo M, Reihill JA, Crilly A, Lockhart JC, Litherland GJ, Lundy FT, McGarvey LP, Hollywood MA, Martin SL. Trypsin-Like Proteases and Their Role in Muco-Obstructive Lung Diseases. Int J Mol Sci 2021; 22:5817. [PMID: 34072295 PMCID: PMC8199346 DOI: 10.3390/ijms22115817] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Trypsin-like proteases (TLPs) belong to a family of serine enzymes with primary substrate specificities for the basic residues, lysine and arginine, in the P1 position. Whilst initially perceived as soluble enzymes that are extracellularly secreted, a number of novel TLPs that are anchored in the cell membrane have since been discovered. Muco-obstructive lung diseases (MucOLDs) are characterised by the accumulation of hyper-concentrated mucus in the small airways, leading to persistent inflammation, infection and dysregulated protease activity. Although neutrophilic serine proteases, particularly neutrophil elastase, have been implicated in the propagation of inflammation and local tissue destruction, it is likely that the serine TLPs also contribute to various disease-relevant processes given the roles that a number of these enzymes play in the activation of both the epithelial sodium channel (ENaC) and protease-activated receptor 2 (PAR2). More recently, significant attention has focused on the activation of viruses such as SARS-CoV-2 by host TLPs. The purpose of this review was to highlight key TLPs linked to the activation of ENaC and PAR2 and their association with airway dehydration and inflammatory signalling pathways, respectively. The role of TLPs in viral infectivity will also be discussed in the context of the inhibition of TLP activities and the potential of these proteases as therapeutic targets.
Collapse
Affiliation(s)
- Emma L. Carroll
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| | - Mariarca Bailo
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - James A. Reihill
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| | - Anne Crilly
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - John C. Lockhart
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - Gary J. Litherland
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - Fionnuala T. Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, Belfast BT9 7BL, UK; (F.T.L.); (L.P.M.)
| | - Lorcan P. McGarvey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, Belfast BT9 7BL, UK; (F.T.L.); (L.P.M.)
| | - Mark A. Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, A91 HRK2 Dundalk, Ireland;
| | - S. Lorraine Martin
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| |
Collapse
|
16
|
McKelvey MC, Brown R, Ryan S, Mall MA, Weldon S, Taggart CC. Proteases, Mucus, and Mucosal Immunity in Chronic Lung Disease. Int J Mol Sci 2021; 22:5018. [PMID: 34065111 PMCID: PMC8125985 DOI: 10.3390/ijms22095018] [Citation(s) in RCA: 10] [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: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Dysregulated protease activity has long been implicated in the pathogenesis of chronic lung diseases and especially in conditions that display mucus obstruction, such as chronic obstructive pulmonary disease, cystic fibrosis, and non-cystic fibrosis bronchiectasis. However, our appreciation of the roles of proteases in various aspects of such diseases continues to grow. Patients with muco-obstructive lung disease experience progressive spirals of inflammation, mucostasis, airway infection and lung function decline. Some therapies exist for the treatment of these symptoms, but they are unable to halt disease progression and patients may benefit from novel adjunct therapies. In this review, we highlight how proteases act as multifunctional enzymes that are vital for normal airway homeostasis but, when their activity becomes immoderate, also directly contribute to airway dysfunction, and impair the processes that could resolve disease. We focus on how proteases regulate the state of mucus at the airway surface, impair mucociliary clearance and ultimately, promote mucostasis. We discuss how, in parallel, proteases are able to promote an inflammatory environment in the airways by mediating proinflammatory signalling, compromising host defence mechanisms and perpetuating their own proteolytic activity causing structural lung damage. Finally, we discuss some possible reasons for the clinical inefficacy of protease inhibitors to date and propose that, especially in a combination therapy approach, proteases represent attractive therapeutic targets for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| |
Collapse
|
17
|
McNulty MJ, Silberstein DZ, Kuhn BT, Padgett HS, Nandi S, McDonald KA, Cross CE. Alpha-1 antitrypsin deficiency and recombinant protein sources with focus on plant sources: Updates, challenges and perspectives. Free Radic Biol Med 2021; 163:10-30. [PMID: 33279618 DOI: 10.1016/j.freeradbiomed.2020.11.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Alpha-1 antitrypsin deficiency (A1ATD) is an autosomal recessive disease characterized by low plasma levels of A1AT, a serine protease inhibitor representing the most abundant circulating antiprotease normally present at plasma levels of 1-2 g/L. The dominant clinical manifestations include predispositions to early onset emphysema due to protease/antiprotease imbalance in distal lung parenchyma and liver disease largely due to unsecreted polymerized accumulations of misfolded mutant A1AT within the endoplasmic reticulum of hepatocytes. Since 1987, the only FDA licensed specific therapy for the emphysema component has been infusions of A1AT purified from pooled human plasma at the 2020 cost of up to US $200,000/year with the risk of intermittent shortages. In the past three decades various, potentially less expensive, recombinant forms of human A1AT have reached early stages of development, one of which is just reaching the stage of human clinical trials. The focus of this review is to update strategies for the treatment of the pulmonary component of A1ATD with some focus on perspectives for therapeutic production and regulatory approval of a recombinant product from plants. We review other competitive technologies for treating the lung disease manifestations of A1ATD, highlight strategies for the generation of data potentially helpful for securing FDA Investigational New Drug (IND) approval and present challenges in the selection of clinical trial strategies required for FDA licensing of a New Drug Approval (NDA) for this disease.
Collapse
Affiliation(s)
- Matthew J McNulty
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - David Z Silberstein
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Brooks T Kuhn
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA
| | | | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Carroll E Cross
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA; Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|
18
|
Editorial: Special Issue on "Therapeutic Approaches for Cystic Fibrosis". Int J Mol Sci 2020; 21:ijms21186657. [PMID: 32932926 PMCID: PMC7555172 DOI: 10.3390/ijms21186657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022] Open
|
19
|
Herman M, Tarran R. E-cigarettes, nicotine, the lung and the brain: multi-level cascading pathophysiology. J Physiol 2020; 598:5063-5071. [PMID: 32515030 DOI: 10.1113/jp278388] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Tobacco smoking is highly addictive and causes respiratory disease, cardiovascular disease and multiple types of cancer. Electronic-cigarettes (e-cigarettes) are non-combustible tobacco alternatives that aerosolize nicotine and flavouring agents in a propylene glycol-vegetable glycerine vehicle. They were originally envisaged as a tobacco cessation aid, but whether or not they help people to quit tobacco use is controversial. In this review, we have compared and contrasted what is known regarding the effects of nicotine on the lungs vs. the effects of nicotine in the brain in the context of addiction. Critically, both combustible tobacco products and e-cigarettes contain nicotine, a highly addictive, plant-derived alkaloid that binds to nicotinic acetylcholine receptors (nAChRs). Nicotine's reinforcing properties are primarily mediated by activation of the brain's mesolimbic reward circuitry and release of the neurotransmitter dopamine that contribute to the development of addiction. Moreover, nicotine addiction drives repeated intake that results in chronic pulmonary exposure to either tobacco smoke or e-cigarettes despite negative respiratory symptoms. Beyond the brain, nAChRs are also highly expressed in peripheral neurons, epithelia and immune cells, where their activation may cause harmful effects. Thus, nicotine, a key ingredient of both conventional and electronic cigarettes, produces neurological effects that drive addiction and may damage the lungs in the process, producing a complex, multilevel pathological state. We conclude that vaping needs to be studied by multi-disciplinary teams that include pulmonary and neurophysiologists as well as behaviourists and addiction specialists to fully understand their impact on human physiology.
Collapse
Affiliation(s)
- Melissa Herman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
20
|
Brown R, Nath S, Lora A, Samaha G, Elgamal Z, Kaiser R, Taggart C, Weldon S, Geraghty P. Cathepsin S: investigating an old player in lung disease pathogenesis, comorbidities, and potential therapeutics. Respir Res 2020; 21:111. [PMID: 32398133 PMCID: PMC7216426 DOI: 10.1186/s12931-020-01381-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
Dysregulated expression and activity of cathepsin S (CTSS), a lysosomal protease and a member of the cysteine cathepsin protease family, is linked to the pathogenesis of multiple diseases, including a number of conditions affecting the lungs. Extracellular CTSS has potent elastase activity and by processing cytokines and host defense proteins, it also plays a role in the regulation of inflammation. CTSS has also been linked to G-coupled protein receptor activation and possesses an important intracellular role in major histocompatibility complex class II antigen presentation. Modulated CTSS activity is also associated with pulmonary disease comorbidities, such as cancer, cardiovascular disease, and diabetes. CTSS is expressed in a wide variety of immune cells and is biologically active at neutral pH. Herein, we review the significance of CTSS signaling in pulmonary diseases and associated comorbidities. We also discuss CTSS as a plausible therapeutic target and describe recent and current clinical trials examining CTSS inhibition as a means for treatment.
Collapse
Affiliation(s)
- Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sridesh Nath
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Alnardo Lora
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ghassan Samaha
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ziyad Elgamal
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ryan Kaiser
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Clifford Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Patrick Geraghty
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA.
- Department of Cell Biology, State University of New York Downstate Medical Centre, Brooklyn, NY, USA.
| |
Collapse
|
21
|
Magréault S, Mankikian J, Marchand S, Diot P, Couet W, Flament T, Grégoire N. Pharmacokinetics of colistin after nebulization or intravenous administration of colistin methanesulphonate (Colimycin®) to cystic fibrosis patients. J Cyst Fibros 2020; 19:421-426. [DOI: 10.1016/j.jcf.2019.08.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 07/12/2019] [Accepted: 08/23/2019] [Indexed: 12/31/2022]
|
22
|
Ricard-Blum S, Miele AE. Omic approaches to decipher the molecular mechanisms of fibrosis, and design new anti-fibrotic strategies. Semin Cell Dev Biol 2020; 101:161-169. [DOI: 10.1016/j.semcdb.2019.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
|
23
|
Hagner M, Frey DL, Guerra M, Dittrich AS, Halls VS, Wege S, Herth FJF, Schultz C, Mall MA. New method for rapid and dynamic quantification of elastase activity on sputum neutrophils from patients with cystic fibrosis using flow cytometry. Eur Respir J 2020; 55:13993003.02355-2019. [PMID: 32139467 DOI: 10.1183/13993003.02355-2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/20/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Matthias Hagner
- Dept of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Equal contribution
| | - Dario L Frey
- Dept of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Equal contribution
| | - Matteo Guerra
- Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany.,EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany.,Equal contribution
| | - A Susanne Dittrich
- Dept of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Dept of Pulmonology and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Victoria S Halls
- Dept of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Sabine Wege
- Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Dept of Pulmonology and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Felix J F Herth
- Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Dept of Pulmonology and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Carsten Schultz
- Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany.,Dept of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA.,Equal contribution as senior author
| | - Marcus A Mall
- Dept of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany .,Translational Lung Research Center (TLRC), Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Dept of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Equal contribution as senior author
| |
Collapse
|
24
|
Wartenberg M, Andrault PM, Saidi A, Bigot P, Nadal-Desbarats L, Lecaille F, Lalmanach G. Oxidation of cathepsin S by major chemicals of cigarette smoke. Free Radic Biol Med 2020; 150:53-65. [PMID: 32084513 DOI: 10.1016/j.freeradbiomed.2020.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/06/2020] [Accepted: 02/17/2020] [Indexed: 01/04/2023]
Abstract
Lung cysteine cathepsin S (CatS) that is a potent elastase plays a deleterious role in alveolar remodeling during smoke-induced emphysema. Despite the presence of a reactive nucleophilic cysteine (Cys25) within its active site, most of its elastinolytic activity is preserved after exposure to cigarette smoke extract (CSE), a major source of sulfhydryl oxidants. This result led us to decipher CatS resistance to major and representative CSE oxidants: hydrogen peroxide, formaldehyde, acrolein and peroxynitrite. CatS was inactivated by hydrogen peroxide, peroxynitrite and acrolein in a time- and dose-dependent manner, while formaldehyde was a weaker oxidant. Hydrogen peroxide, but not CSE, formaldehyde, and peroxynitrite impaired the autocatalytic maturation of pro-CatS, whereas acrolein prevented the formation of mature CatS without hindering the initial step of the two-step autocatalytic process. Far-UV CD spectra analysis supported that oxidation by CSE and hydrogen peroxide did not led to a structural alteration of CatS, despite a notable increase of protein carbonylation, a major hallmark of oxidative damage. Evaluation of the oxidation status of Cys25 by specific biotinylated redox sensing probes suggested the formation of sulfenic acid followed by a slower conversion to sulfinic acid after incubation with hydrogen peroxide. Addition of reducing reagents (dithiothreitol, glutathione and N-acetyl cysteine) led to a partial recovery of CatS activity following incubation with CSE, hydrogen peroxide and peroxynitrite. Current results provide some mechanistic evidence of CatS stability and activity in the presence of CSE, supporting its harmful contribution to the pathophysiology of emphysema.
Collapse
Affiliation(s)
- Mylène Wartenberg
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France
| | - Pierre-Marie Andrault
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France
| | - Paul Bigot
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France
| | - Lydie Nadal-Desbarats
- Université de Tours, Tours, France; INSERM, UMR1253, Imagerie et Cerveau (iBrain), Team « Imageries, Biomarqueurs et Thérapies », Tours, France
| | - Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team « Mécanismes Protéolytiques dans l'Inflammation », Tours, France.
| |
Collapse
|
25
|
Regulation of the Proteolytic Activity of Cysteine Cathepsins by Oxidants. Int J Mol Sci 2020; 21:ijms21061944. [PMID: 32178437 PMCID: PMC7139492 DOI: 10.3390/ijms21061944] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
Besides their primary involvement in the recycling and degradation of proteins in endo-lysosomal compartments and also in specialized biological functions, cysteine cathepsins are pivotal proteolytic contributors of various deleterious diseases. While the molecular mechanisms of regulation via their natural inhibitors have been exhaustively studied, less is currently known about how their enzymatic activity is modulated during the redox imbalance associated with oxidative stress and their exposure resistance to oxidants. More specifically, there is only patchy information on the regulation of lung cysteine cathepsins, while the respiratory system is directly exposed to countless exogenous oxidants contained in dust, tobacco, combustion fumes, and industrial or domestic particles. Papain-like enzymes (clan CA, family C1, subfamily C1A) encompass a conserved catalytic thiolate-imidazolium pair (Cys25-His159) in their active site. Although the sulfhydryl group (with a low acidic pKa) is a potent nucleophile highly susceptible to chemical modifications, some cysteine cathepsins reveal an unanticipated resistance to oxidative stress. Besides an introductory chapter and peculiar attention to lung cysteine cathepsins, the purpose of this review is to afford a concise update of the current knowledge on molecular mechanisms associated with the regulation of cysteine cathepsins by redox balance and by oxidants (e.g., Michael acceptors, reactive oxygen, and nitrogen species).
Collapse
|
26
|
Alpha-1 Antitrypsin-A Target for MicroRNA-Based Therapeutic Development for Cystic Fibrosis. Int J Mol Sci 2020; 21:ijms21030836. [PMID: 32012925 PMCID: PMC7037267 DOI: 10.3390/ijms21030836] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder arising from mutations to the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Disruption to normal ion homeostasis in the airway results in impaired mucociliary clearance, leaving the lung more vulnerable to recurrent and chronic bacterial infections. The CF lung endures an excess of neutrophilic inflammation, and whilst neutrophil serine proteases are a crucial part of the innate host defence to infection, a surplus of neutrophil elastase (NE) is understood to create a net destructive effect. Alpha-1 antitrypsin (A1AT) is a key antiprotease in the control of NE protease activity but is ineffective in the CF lung due to the huge imbalance of NE levels. Therapeutic strategies to boost levels of protective antiproteases such as A1AT in the lung remain an attractive research strategy to limit the damage from excess protease activity. microRNAs are small non-coding RNA molecules that bind specific cognate sequences to inhibit expression of target mRNAs. The inhibition of miRNAs which target the SERPINA1 (A1AT-encoding gene) mRNA represents a novel therapeutic approach for CF inflammation. This could involve the delivery of antagomirs that bind and sequester the target miRNA, or target site blockers that bind miRNA recognition elements within the target mRNA to prevent miRNA interaction. Therefore, miRNA targeted therapies offer an alternative strategy to drive endogenous A1AT production and thus supplement the antiprotease shield of the CF lung.
Collapse
|
27
|
Stockley RA. Alpha-1 Antitrypsin Deficiency: Have We Got the Right Proteinase? CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:163-171. [PMID: 32396717 DOI: 10.15326/jcopdf.7.3.2019.0151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alpha-1 antitrypsin deficiency (AATD) has traditionally been associated with the development of early onset panlobular emphysema thought to reflect the direct interstitial damage caused by neutrophil elastase. Since this enzyme is highly sensitive to irreversible inhibition by alpha-1 antitrypsin (AAT), the logic of intravenous augmentation therapy has remained unquestioned and efficacy is supported by both observational studies and formal clinical trials. However, evidence suggests that although AAT augmentation modulates the progression of emphysema, it only slows it down. This raises the issue of whether our long-held beliefs of the cause of the susceptibility to develop emphysema in deficient individuals are correct. There are several aspects of our understanding of the disease that might benefit from a radical departure from traditional thought. This review addresses these concepts and alternative pathways that may be central to progression of emphysema.
Collapse
Affiliation(s)
- Robert A Stockley
- Lung Investigation Unit, University Hospitals, Birmingham National Health Service Foundation Trust, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
28
|
Nakajima K, Ono M, Radović U, Dizdarević S, Tomizawa SI, Kuroha K, Nagamatsu G, Hoshi I, Matsunaga R, Shirakawa T, Kurosawa T, Miyazaki Y, Seki M, Suzuki Y, Koseki H, Nakamura M, Suda T, Ohbo K. Lack of whey acidic protein (WAP) four-disulfide core domain protease inhibitor 2 (WFDC2) causes neonatal death from respiratory failure in mice. Dis Model Mech 2019; 12:dmm.040139. [PMID: 31562139 PMCID: PMC6899016 DOI: 10.1242/dmm.040139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Respiratory failure is a life-threatening problem for pre-term and term infants, yet many causes remain unknown. Here, we present evidence that whey acidic protein (WAP) four-disulfide core domain protease inhibitor 2 (Wfdc2), a protease inhibitor previously unrecognized in respiratory disease, may be a causal factor in infant respiratory failure. Wfdc2 transcripts are detected in the embryonic lung and analysis of a Wfdc2-GFP knock-in mouse line shows that both basal and club cells, and type II alveolar epithelial cells (AECIIs), express Wfdc2 neonatally. Wfdc2-null-mutant mice display progressive atelectasis after birth with a lethal phenotype. Mutant lungs have multiple defects, including impaired cilia and the absence of mature club cells from the tracheo-bronchial airways, and malformed lamellar bodies in AECIIs. RNA sequencing shows significant activation of a pro-inflammatory pathway, but with low-quantity infiltration of mononuclear cells in the lung. These data demonstrate that Wfdc2 function is vitally important for lung aeration at birth and that gene deficiency likely causes failure of the lung mucosal barrier.
Collapse
Affiliation(s)
- Kuniko Nakajima
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Michio Ono
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Uroš Radović
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Selma Dizdarević
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shin-Ichi Tomizawa
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kazushige Kuroha
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Go Nagamatsu
- Department of Stem Cell Biology, Kyushu University, Faculty of Medical Sciences, 3-1-1, Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Ikue Hoshi
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Risa Matsunaga
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takayuki Shirakawa
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takeyuki Kurosawa
- Department of Respiratory Medicine, Toho University, School of Medicine, 5-21-16, Ohmorinishi, Ohta-ku, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Masataka Nakamura
- Human Gene Sciences Center, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Toshio Suda
- Cancer Science Institute of Singapore, National Singapore University Centre for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599.,International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Kazuyuki Ohbo
- Department of Histology and Cell Biology, Yokohama City University, School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
29
|
Andrault PM, Schamberger AC, Chazeirat T, Sizaret D, Renault J, Staab-Weijnitz CA, Hennen E, Petit-Courty A, Wartenberg M, Saidi A, Baranek T, Guyetant S, Courty Y, Eickelberg O, Lalmanach G, Lecaille F. Cigarette smoke induces overexpression of active human cathepsin S in lungs from current smokers with or without COPD. Am J Physiol Lung Cell Mol Physiol 2019; 317:L625-L638. [PMID: 31553637 DOI: 10.1152/ajplung.00061.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cigarette smoking has marked effects on lung tissue, including induction of oxidative stress, inflammatory cell recruitment, and a protease/antiprotease imbalance. These effects contribute to tissue remodeling and destruction resulting in loss of lung function in chronic obstructive pulmonary disease (COPD) patients. Cathepsin S (CatS) is a cysteine protease that is involved in the remodeling/degradation of connective tissue and basement membrane. Aberrant expression or activity of CatS has been implicated in a variety of diseases, including arthritis, cancer, cardiovascular, and lung diseases. However, little is known about the effect of cigarette smoking on both CatS expression and activity, as well as its role in smoking-related lung diseases. Here, we evaluated the expression and activity of human CatS in lung tissues from never-smokers and smokers with or without COPD. Despite the presence of an oxidizing environment, CatS expression and activity were significantly higher in current smokers (both non-COPD and COPD) compared with never-smokers, and correlated positively with smoking history. Moreover, we found that the exposure of primary human bronchial epithelial cells to cigarette smoke extract triggered the activation of P2X7 receptors, which in turns drives CatS upregulation. The present data suggest that excessive CatS expression and activity contribute, beside other proteases, to the deleterious effects of cigarette smoke on pulmonary homeostasis.
Collapse
Affiliation(s)
- Pierre-Marie Andrault
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Andrea C Schamberger
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Thibault Chazeirat
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Damien Sizaret
- Université de Tours, Tours, France.,Centre Hospitalier Régional Universitaire de Tours, Service d'Anatomie et Cytologie Pathologique, Tours, France
| | | | - Claudia A Staab-Weijnitz
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Elisabeth Hennen
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Agnès Petit-Courty
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Mylène Wartenberg
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Thomas Baranek
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Serge Guyetant
- Université de Tours, Tours, France.,Centre Hospitalier Régional Universitaire de Tours, Service d'Anatomie et Cytologie Pathologique, Tours, France
| | - Yves Courty
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Gilles Lalmanach
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| | - Fabien Lecaille
- Université de Tours, Tours, France.,INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Team Mécanismes Protéolytiques dans l'Inflammation, Tours, France
| |
Collapse
|
30
|
Bari E, Ferrarotti I, Di Silvestre D, Grisoli P, Barzon V, Balderacchi A, Torre ML, Rossi R, Mauri P, Corsico AG, Perteghella S. Adipose Mesenchymal Extracellular Vesicles as Alpha-1-Antitrypsin Physiological Delivery Systems for Lung Regeneration. Cells 2019; 8:E965. [PMID: 31450843 PMCID: PMC6770759 DOI: 10.3390/cells8090965] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence shows that Mesenchymal Stem/Stromal Cells (MSCs) exert their therapeutic effects by the release of secretome, made of both soluble proteins and nano/microstructured extracellular vesicles (EVs). In this work, for the first time, we proved by a proteomic investigation that adipose-derived (AD)-MSC-secretome contains alpha-1-antitrypsin (AAT), the main elastase inhibitor in the lung, 72 other proteins involved in protease/antiprotease balance, and 46 proteins involved in the response to bacteria. By secretome fractionation, we proved that AAT is present both in the soluble fraction of secretome and aggregated and/or adsorbed on the surface of EVs, that can act as natural carriers promoting AAT in vivo stability and activity. To modulate secretome composition, AD-MSCs were cultured in different stimulating conditions, such as serum starvation or chemicals (IL-1β and/or dexamethasone) and the expression of the gene encoding for AAT was increased. By testing in vitro the anti-elastase activity of MSC-secretome, a dose-dependent effect was observed; chemical stimulation of AD-MSCs did not increase their secretome anti-elastase activity. Finally, MSC-secretome showed anti-bacterial activity on Gram-negative bacteria, especially for Klebsiellapneumoniae. These preliminary results, in addition to the already demonstrated immunomodulation, pave the way for the use of MSC-secretome in the treatment of AAT-deficiency lung diseases.
Collapse
Affiliation(s)
- Elia Bari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Ilaria Ferrarotti
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, Pneumology Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy
| | - Dario Di Silvestre
- Institute for Biomedical Technologies, F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Pietro Grisoli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Valentina Barzon
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, Pneumology Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy
| | - Alice Balderacchi
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, Pneumology Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
- PharmaExceed S.r.l., Piazza Castello, 19, 27100 Pavia, Italy.
| | - Rossana Rossi
- Institute for Biomedical Technologies, F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies, F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Angelo Guido Corsico
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, Pneumology Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, 27100 Pavia, Italy
- PharmaExceed S.r.l., Piazza Castello, 19, 27100 Pavia, Italy
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
- PharmaExceed S.r.l., Piazza Castello, 19, 27100 Pavia, Italy
| |
Collapse
|
31
|
Takahashi Y, Kobayashi T, D'Alessandro-Gabazza CN, Toda M, Fujiwara K, Okano T, Fujimoto H, Asayama K, Takeshita A, Yasuma T, Nishihama K, Inoue R, Qin L, Takei Y, Taguchi O, Gabazza EC. Protective Role of Matrix Metalloproteinase-2 in Allergic Bronchial Asthma. Front Immunol 2019; 10:1795. [PMID: 31428095 PMCID: PMC6687911 DOI: 10.3389/fimmu.2019.01795] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022] Open
Abstract
Inflammation, reversible obstruction, and hyperresponsiveness of the airways are characteristic findings of bronchial asthma. Several evidence has demonstrated the involvement of matrix metalloproteinase-2 in allergic airway inflammation. Matrix metalloproteinase-2 may promote aberrant tissue remodeling in late stages of allergic airway inflammation. However, whether matrix metalloproteinase-2 is detrimental or protective in early stages of allergic airway inflammation remains unclear. To evaluate this here we compared the severity of allergic bronchial asthma between mice overexpressing human matrix metalloproteinase-2 and wild type mice. After sensitization and challenge with an allergen, mice overexpressing the human matrix metalloproteinase-2 showed a significant reduction in airway hyperresponsiveness and in the expression of Th2 cytokines and IgE compared to their wild type counterparts. An inhibitor of matrix metalloproteinases abolished this beneficial effect of human matrix metalloproteinase-2 overexpression. Allergen-sensitized and challenged human matrix metalloproteinase-2 transgenic mice had enhanced percentage of M1 macrophages with increased expression of inducible nitric oxide synthase and STAT1 activation in the lungs compared to their wild type counterparts. There was no difference in the percentage of regulatory T cells between mouse groups. The results of this study showed that matrix metalloproteinase-2 is protective in allergic bronchial asthma by promoting polarization of macrophages to M1 phenotype.
Collapse
Affiliation(s)
- Yoshinori Takahashi
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | | | - Masaaki Toda
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kentaro Fujiwara
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Tomohito Okano
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hajime Fujimoto
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kentaro Asayama
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Atsuro Takeshita
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Taro Yasuma
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kota Nishihama
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Ryo Inoue
- Central Institute for Experimental Animals, Kawasaki-ku, Japan
| | - Liqiang Qin
- Department of Nephrology, Taizhou Hospital, Wenzhou Medical University, Lihai, China
| | - Yoshiyuki Takei
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Osamu Taguchi
- Center for Physical and Mental Health, Mie University Graduate School of Medicine, Tsu, Japan
| | - Esteban C Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
32
|
Bari E, Ferrarotti I, Torre ML, Corsico AG, Perteghella S. Mesenchymal stem/stromal cell secretome for lung regeneration: The long way through "pharmaceuticalization" for the best formulation. J Control Release 2019; 309:11-24. [PMID: 31326462 DOI: 10.1016/j.jconrel.2019.07.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
Pulmonary acute and chronic diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and pulmonary hypertension, are considered to be major health issues worldwide. Cellular therapies with Mesenchymal Stem Cells (MSCs) offer a new therapeutic approach for chronic and acute lung diseases related to their anti-inflammatory, immunomodulatory, regenerative, pro-angiogenic and anti-fibrotic properties. Such therapeutic effects can be attributed to MSC-secretome, made of free soluble proteins and extracellular vesicles (EVs). This review summarizes the recent findings related to the efficacy and safety of MSC-derived products in pre-clinical models of lung diseases, pointing out the biologically active substances contained into MSC-secretome and their mechanisms involved in tissue regeneration. A perspective view is then provided about the missing steps required for the secretome "pharmaceuticalization" into a high quality, safe and effective medicinal product, as well as the formulation strategies required for EV non-invasive route of administration, such as inhalation.
Collapse
Affiliation(s)
- Elia Bari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy
| | - Ilaria Ferrarotti
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy.
| | - Angelo Guido Corsico
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy
| |
Collapse
|
33
|
Biguanide is a modifiable pharmacophore for recruitment of endogenous Zn 2+ to inhibit cysteinyl cathepsins: review and implications. Biometals 2019; 32:575-593. [PMID: 31044334 PMCID: PMC6647370 DOI: 10.1007/s10534-019-00197-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/13/2019] [Indexed: 01/28/2023]
Abstract
Excessive activities of cysteinyl cathepsins (CysCts) contribute to the progress of many diseases; however, therapeutic inhibition has been problematic. Zn2+ is a natural inhibitor of proteases with CysHis dyads or CysHis(Xaa) triads. Biguanide forms bidentate metal complexes through the two imino nitrogens. Here, it is discussed that phenformin (phenylethyl biguanide) is a model for recruitment of endogenous Zn2+ to inhibit CysHis/CysHis(X) peptidolysis. Phenformin is a Zn2+-interactive, anti-proteolytic agent in bioassay of living tissue. Benzoyl-L-arginine amide (BAA) is a classical substrate of papain-like proteases; the amide bond is scissile. In this review, the structures of BAA and the phenformin-Zn2+ complex were compared in silico. Their chemistry and dimensions are discussed in light of the active sites of papain-like proteases. The phenyl moieties of both structures bind to the "S2" substrate-binding site that is typical of many proteases. When the phenyl moiety of BAA binds to S2, then the scissile amide bond is directed to the position of the thiolate-imidazolium ion pair, and is then hydrolyzed. However, when the phenyl moiety of phenformin binds to S2, then the coordinated Zn2+ is directed to the identical position; and catalysis is inhibited. Phenformin stabilizes a "Zn2+ sandwich" between the drug and protease active site. Hundreds of biguanide derivatives have been synthesized at the 1 and 5 nitrogen positions; many more are conceivable. Various substituent moieties can register with various arrays of substrate-binding sites so as to align coordinated Zn2+ with catalytic partners of diverse proteases. Biguanide is identified here as a modifiable pharmacophore for synthesis of therapeutic CysCt inhibitors with a wide range of potencies and specificities. Phenformin-Zn2+ Complex.
Collapse
|
34
|
Wartenberg M, Saidi A, Galibert M, Joulin-Giet A, Burlaud-Gaillard J, Lecaille F, Scott CJ, Aucagne V, Delmas AF, Lalmanach G. Imaging of extracellular cathepsin S activity by a selective near infrared fluorescence substrate-based probe. Biochimie 2019; 166:84-93. [PMID: 30914255 DOI: 10.1016/j.biochi.2019.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/19/2019] [Indexed: 02/05/2023]
Abstract
We designed a near-infrared fluorescent substrate-based probe (SBP), termed MG101, for monitoring extracellular cathepsin S (CatS) activity. We conceived a fused peptide hairpin loop-structure, combining a CatS recognition domain, an electrostatic zipper (with complementary charges of a polyanionic (D-Glu)5 segment and a polycationic (D-Arg)5 motif, as well as a N and C terminal Förster resonance energy transfer pair (donor: AlexaFluor680; quencher: BHQ3) to facilitate activity-dependent imaging. MG101 showed excellent stability since no fluorescence release corresponding to a self-dequenching was observed in the presence of either 2 M NaCl or after incubation at a broad range of pH (2.2-8.2). Cathepsins B, D, G, H, and K, neutrophil elastase and proteinase 3 did not cleave MG101, while CatS, and to a lesser extent CatL, hydrolysed MG101 at pH 5.5. However MG101 was fully selective for CatS at pH 7.4 (kcat/Km = 140,000 M-1 s-1) and sensitive to low concentration of CatS (<1 nM). The selectivity of MG101 was successfully endorsed ex vivo, as it was hydrolysed in cell lysates derived from wild-type but not knockout CatS murine spleen. Furthermore, application of the SBP probe with confocal microscopy confirmed the secretion of active CatS from THP-1 macrophages, which could be abrogated by pharmacological CatS inhibitors. Taken together, present data highlight MG101 as a novel near-infrared fluorescent SBP for the visualization of extracellular active CatS from macrophages and other cell types.
Collapse
Affiliation(s)
- Mylène Wartenberg
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Mathieu Galibert
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Alix Joulin-Giet
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Julien Burlaud-Gaillard
- Université de Tours, Tours, France; Plateforme IBiSA de Microscopie Electronique, Université de Tours, Tours, France
| | - Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Vincent Aucagne
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Agnès F Delmas
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France.
| |
Collapse
|
35
|
Vidak E, Javoršek U, Vizovišek M, Turk B. Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 2019; 8:cells8030264. [PMID: 30897858 PMCID: PMC6468544 DOI: 10.3390/cells8030264] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/17/2022] Open
Abstract
For a long time, cysteine cathepsins were considered primarily as proteases crucial for nonspecific bulk proteolysis in the endolysosomal system. However, this view has dramatically changed, and cathepsins are now considered key players in many important physiological processes, including in diseases like cancer, rheumatoid arthritis, and various inflammatory diseases. Cathepsins are emerging as important players in the extracellular space, and the paradigm is shifting from the degrading enzymes to the enzymes that can also specifically modify extracellular proteins. In pathological conditions, the activity of cathepsins is often dysregulated, resulting in their overexpression and secretion into the extracellular space. This is typically observed in cancer and inflammation, and cathepsins are therefore considered valuable diagnostic and therapeutic targets. In particular, the investigation of limited proteolysis by cathepsins in the extracellular space is opening numerous possibilities for future break-through discoveries. In this review, we highlight the most important findings that establish cysteine cathepsins as important players in the extracellular space and discuss their roles that reach beyond processing and degradation of extracellular matrix (ECM) components. In addition, we discuss the recent developments in cathepsin research and the new possibilities that are opening in translational medicine.
Collapse
Affiliation(s)
- Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Matej Vizovišek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich Otto-Stern-Weg 3, 8093 Zürich, Switzerland.
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
Curcumin inhibits the TGF-β1-dependent differentiation of lung fibroblasts via PPARγ-driven upregulation of cathepsins B and L. Sci Rep 2019; 9:491. [PMID: 30679571 PMCID: PMC6345753 DOI: 10.1038/s41598-018-36858-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/26/2018] [Indexed: 01/15/2023] Open
Abstract
Pulmonary fibrosis is a progressive disease characterized by a widespread accumulation of myofibroblasts and extracellular matrix components. Growing evidences support that cysteine cathepsins, embracing cathepsin B (CatB) that affects TGF-β1-driven Smad pathway, along with their extracellular inhibitor cystatin C, participate in myofibrogenesis. Here we established that curcumin, a potent antifibrotic drug used in traditional Asian medicine, impaired the expression of both α-smooth muscle actin and mature TGF-β1 and inhibited the differentiation of human lung fibroblasts (CCD-19Lu cells). Curcumin induced a compelling upregulation of CatB and CatL. Conversely cystatin C was downregulated, which allowed the recovery of the peptidase activity of secreted cathepsins and the restoration of the proteolytic balance. Consistently, the amount of both insoluble and soluble type I collagen decreased, reaching levels similar to those observed for undifferentiated fibroblasts. The signaling pathways activated by curcumin were further examined. Curcumin triggered the expression of nuclear peroxisome proliferator-activated receptor γ (PPARγ). Contrariwise PPARγ inhibition, either by an antagonist (2-chloro-5-nitro-N-4-pyridinyl-benzamide) or by RNA silencing, restored TGF-β1-driven differentiation of curcumin-treated CCD-19Lu cells. PPARγ response element (PPRE)-like sequences were identified in the promoter regions of both CatB and CatL. Finally, we established that the transcriptional induction of CatB and CatL depends on the binding of PPARγ to PPRE sequences as a PPARγ/Retinoid X Receptor-α heterodimer.
Collapse
|
37
|
Vizovišek M, Fonović M, Turk B. Cysteine cathepsins in extracellular matrix remodeling: Extracellular matrix degradation and beyond. Matrix Biol 2019; 75-76:141-159. [DOI: 10.1016/j.matbio.2018.01.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/14/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022]
|
38
|
Mallia-Milanes B, Dufour A, Philp C, Solis N, Klein T, Fischer M, Bolton CE, Shapiro S, Overall CM, Johnson SR. TAILS proteomics reveals dynamic changes in airway proteolysis controlling protease activity and innate immunity during COPD exacerbations. Am J Physiol Lung Cell Mol Physiol 2018; 315:L1003-L1014. [PMID: 30284925 DOI: 10.1152/ajplung.00175.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dysregulated protease activity is thought to cause parenchymal and airway damage in chronic obstructive pulmonary disease (COPD). Multiple proteases have been implicated in COPD, and identifying their substrates may reveal new disease mechanisms and treatments. However, as proteases interact with many substrates that may be protease inhibitors or proteases themselves, these webs of protease interactions make the wider consequences of therapeutically targeting proteases difficult to predict. We therefore used a systems approach to determine protease substrates and protease activity in COPD airways. Protease substrates were determined by proteomics using the terminal amine isotopic labeling of substrates (TAILS) methodology in paired sputum samples during stable COPD and exacerbations. Protease activity and specific protein degradation in airway samples were assessed using Western blotting, substrate assays, and ex vivo cleavage assays. Two hundred ninety-nine proteins were identified in human COPD sputum, 125 of which were proteolytically processed, including proteases, protease inhibitors, mucins, defensins, and complement and other innate immune proteins. During exacerbations, airway neutrophils and neutrophil proteases increased and more proteins were cleaved, particularly at multiple sites, consistent with degradation and inactivation. During exacerbations, different substrates were processed, including protease inhibitors, mucins, and complement proteins. Exacerbations were associated with increasing airway elastase activity and increased processing of specific elastase substrates, including secretory leukocyte protease inhibitor. Proteolysis regulates multiple processes including elastase activity and innate immune proteins in COPD airways and differs during stable disease and exacerbations. The complexity of protease, inhibitor, and substrate networks makes the effect of protease inhibitors hard to predict which should be used cautiously.
Collapse
Affiliation(s)
- Brendan Mallia-Milanes
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom
| | - Antoine Dufour
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Christopher Philp
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| | - Nestor Solis
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Theo Klein
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Marlies Fischer
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| | - Charlotte E Bolton
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom
| | - Steven Shapiro
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Christopher M Overall
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Simon R Johnson
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| |
Collapse
|
39
|
Taylor S, Dirir O, Zamanian RT, Rabinovitch M, Thompson AAR. The Role of Neutrophils and Neutrophil Elastase in Pulmonary Arterial Hypertension. Front Med (Lausanne) 2018; 5:217. [PMID: 30131961 PMCID: PMC6090899 DOI: 10.3389/fmed.2018.00217] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/16/2018] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe vasculopathy characterized by the presence of fibrotic lesions in the arterial wall and the loss of small distal pulmonary arteries. The vasculopathy is accompanied by perivascular inflammation and increased protease levels, with neutrophil elastase notably implicated in aberrant vascular remodeling. However, the source of elevated elastase levels in PAH remains unclear. A major source of neutrophil elastase is the neutrophil, an understudied cell population in PAH. The principal function of neutrophils is to destroy invading pathogens by means of phagocytosis and NET formation, but proteases, chemokines, and cytokines implicated in PAH can be released by and/or prime and activate neutrophils. This review focuses on the contribution of inflammation to the development and progression of the disease, highlighting studies implicating neutrophils, neutrophil elastase, and other neutrophil proteases in PAH. The roles of cytokines, chemokines, and neutrophil elastase in the disease are discussed and we describe new insight into the role neutrophils potentially play in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Shalina Taylor
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States
| | - Omar Dirir
- Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Roham T. Zamanian
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States
- Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Marlene Rabinovitch
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States
| | - A. A. Roger Thompson
- Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
40
|
Tber Z, Wartenberg M, Jacques JE, Roy V, Lecaille F, Warszycki D, Bojarski AJ, Lalmanach G, Agrofoglio LA. Selective inhibition of human cathepsin S by 2,4,6-trisubstituted 1,3,5-triazine analogs. Bioorg Med Chem 2018; 26:4310-4319. [PMID: 30049585 DOI: 10.1016/j.bmc.2018.07.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022]
Abstract
We report herein the synthesis and biological evaluation of a new series of 2,4,6-trisubstituted 1,3,5-triazines as reversible inhibitors of human cysteine cathepsins. The desired products bearing morpholine and N-Boc piperidine, respectively, were obtained in three to four steps from commercially available trichlorotriazine. Seventeen hitherto unknown compounds were evaluated in vitro against various cathepsins for their inhibitory properties. Among them, compound 7c (4-(morpholin-4-yl)-6-[4-(trifluoromethoxy)anilino]-1,3,5-triazine-2-carbonitrile) was identified as the most potent and selective inhibitor of cathepsin S (Ki = 2 ± 0.3 nM). Also 7c impaired the autocatalytic maturation of procathepsin S. Molecular docking studies support that 7c bound within the active site of cathepsin S, by interacting with Gly23, Cys25 and Trp26 (S1 subsite), with Asn67, Gly69 and Phe70 (S2 subsite) and with Gln19 (S1' pocket).
Collapse
Affiliation(s)
- Zahira Tber
- Université d'Orléans, CNRS, ICOA, UMR 7311, F-45067 Orléans, France
| | - Mylène Wartenberg
- INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France
| | | | - Vincent Roy
- Université d'Orléans, CNRS, ICOA, UMR 7311, F-45067 Orléans, France.
| | - Fabien Lecaille
- INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France
| | - Dawid Warszycki
- Medicinal Chemistry Department, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej J Bojarski
- Medicinal Chemistry Department, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Gilles Lalmanach
- INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France
| | | |
Collapse
|
41
|
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a devastating chronic, progressive and irreversible disease that remains refractory to current therapies. Matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitors of MMPs (TIMPs), have been implicated in the development of pulmonary fibrosis since decades. Coagulation signalling deregulation, which influences several key inflammatory and fibro-proliferative responses, is also essential in IPF pathogenesis, and a growing body of evidence indicates that Protease-Activated Receptors (PARs) inhibition in IPF may be promising for future evaluation. Therefore, proteases and anti-proteases aroused great biomedical interest over the past years, owing to the identification of their potential roles in lung fibrosis. During these last decades, numerous other proteases and anti-proteases have been studied in lung fibrosis, such as matriptase, Human airway trypsin-like protease (HAT), Hepatocyte growth factor activator (HGFA)/HGFA activator inhibitor (HAI) system, Plasminogen activator inhibitor (PAI)-1, Protease nexine (PN)-1, cathepsins, calpains, and cystatin C. Herein, we provide a general overview of the proteases and anti-proteases unbalance during lung fibrogenesis and explore potential therapeutics for IPF.
Collapse
|
42
|
Horani A, Ustione A, Huang T, Firth AL, Pan J, Gunsten SP, Haspel JA, Piston DW, Brody SL. Establishment of the early cilia preassembly protein complex during motile ciliogenesis. Proc Natl Acad Sci U S A 2018; 115:E1221-E1228. [PMID: 29358401 PMCID: PMC5819421 DOI: 10.1073/pnas.1715915115] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Motile cilia are characterized by dynein motor units, which preassemble in the cytoplasm before trafficking into the cilia. Proteins required for dynein preassembly were discovered by finding human mutations that result in absent ciliary motors, but little is known about their expression, function, or interactions. By monitoring ciliogenesis in primary airway epithelial cells and MCIDAS-regulated induced pluripotent stem cells, we uncovered two phases of expression of preassembly proteins. An early phase, composed of HEATR2, SPAG1, and DNAAF2, preceded other preassembly proteins and was independent of MCIDAS regulation. The early preassembly proteins colocalized within perinuclear foci that also contained dynein arm proteins. These proteins also interacted based on immunoprecipitation and Förster resonance energy transfer (FRET) studies. FRET analysis of HEAT domain deletions and human mutations showed that HEATR2 interacted with itself and SPAG1 at multiple HEAT domains, while DNAAF2 interacted with SPAG1. Human mutations in HEATR2 did not affect this interaction, but triggered the formation of p62/Sequestosome-1-positive aggregates containing the early preassembly proteins, suggesting that degradation of an early preassembly complex is responsible for disease and pointing to key regions required for HEATR2 scaffold stability. We speculate that HEATR2 is an early scaffold for the initiation of dynein complex assembly in motile cilia.
Collapse
Affiliation(s)
- Amjad Horani
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110;
| | - Alessandro Ustione
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tao Huang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Amy L Firth
- Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033
| | - Jiehong Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Sean P Gunsten
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jeffrey A Haspel
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
43
|
Menou A, Flajolet P, Duitman J, Justet A, Moog S, Jaillet M, Tabèze L, Solhonne B, Garnier M, Mal H, Mordant P, Castier Y, Cazes A, Sallenave J, A. Mailleux A, Crestani B. Human airway trypsin‐like protease exerts potent, antifibrotic action in pulmonary fibrosis. FASEB J 2018; 32:1250-1264. [DOI: 10.1096/fj.201700583r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Awen Menou
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Pauline Flajolet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - JanWillem Duitman
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Aurélien Justet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Sophie Moog
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Madeleine Jaillet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Laure Tabèze
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Brigitte Solhonne
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Marc Garnier
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Departement d'Anesthésie et Réanimation, (AP‐HP) Hôpital Tenon Paris France
| | - Hervé Mal
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie et Transplantation Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Pierre Mordant
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Chirurgie Thoracique et Vasculaire Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Yves Castier
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Chirurgie Thoracique et Vasculaire Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Aurélie Cazes
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Departement d'Anatomie Pathologique Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Jean‐Michel Sallenave
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Arnaud A. Mailleux
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Bruno Crestani
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| |
Collapse
|