1
|
Huang C, Qiu H, Xu C, Tan Z, Jin M, Hu J, Huang Z, Zhou Y, Ge S, Hu X. Downregulation of tropomyosin 2 promotes the progression of lung adenocarcinoma by regulating neutrophil infiltration through neutrophil elastase. Cell Death Dis 2025; 16:264. [PMID: 40199876 PMCID: PMC11978998 DOI: 10.1038/s41419-025-07531-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Lung adenocarcinoma (LUAD) is a common malignant tumor in the lung that seriously endangers the health of people worldwide. The neutrophil-associated inflammatory microenvironment contributes to the activation of tumor cells. In this study, we report a role of tumor-associated neutrophils (TANs) promote tumor progression of LUAD by crosstalk between neutrophils and tumor cells. Mechanistically, in co-culture with tumor cells, downregulation of TPM2 on tumor cells increases neutrophil elastase (ELANE) levels in neutrophils regulated by p38/ MAPK signaling activation, and ELANE promotes tumor cell progression through the Hippo pathway. Furthermore, downregulation of TPM2 activates ELANE of neutrophils to facilitate ERK1/2 activation, thus enhancing IL1β and IL8 secretion for chemoattraction of more neutrophils to tumor microenvironment. The new studies identify an accomplice role for the interaction between TPM2 and ELANE in promoting LUAD progression and provide potential strategies in the prevention and/or treatment of LUAD and other cancers.
Collapse
Affiliation(s)
- Caixiu Huang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Hao Qiu
- The First People's Hospital of Changde City, Changde, PR China
| | - Changting Xu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Zilong Tan
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Mei Jin
- Pingxiang People's Hospital, Pingxiang, PR China
| | - Jing Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | | | - Yuwei Zhou
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Shengyou Ge
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| | - Xiaoyuan Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| |
Collapse
|
2
|
Kant R, Mishra N, Kandhari K, Saba L, Tewari-Singh N, Petrash JM, Agarwal C, Agarwal R. Dexamethasone mitigates sulfur mustard-induced stem cell deficiency in vivo in rabbit limbal tissue by reducing inflammation and oxidative stress. Arch Toxicol 2025; 99:1531-1543. [PMID: 39904901 DOI: 10.1007/s00204-025-03961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Sulfur mustard (SM) exposure induces ocular injury primarily to the cornea, limbus, and sclera. Although corneal injuries have been studied in detail, there is a dearth of literature on the effects of SM on limbus, particularly mechanisms underlying its compromised functioning, causing limbal stem cell deficiency (LSCD). LSCD causes impaired corneal repair leading to persistent epithelial defects, mustard gas keratopathy, and prolonged inflammation, resulting in total blindness in case of severe damage. Notably, dexamethasone (Dex) has been reported to treat SM-induced corneal injuries effectively; however, its efficacy for SM-induced limbus injury has not been studied. Hence, delayed/persistent structural damage (H&E and trichrome staining) and loss of LSCs [ΔNp63; immunofluorescence (IF)] in the limbus at day 28 post-SM exposure were assessed. Thereafter, in-depth proteomic analysis (LC-MS/MS) of SM exposed, Dex treated, and control limbal tissues (New Zealand white male rabbits) was performed. SM exposure significantly modulated the expression profile of 66 proteins, of which 62 were significantly reversed with Dex; thus, markedly inhibiting/hindering SM-induced limbal injury. Ingenuity Pathway Analysis predicted the primary involvement of (1) inflammation and immune response-associated pathways via dysregulation of defensin-5, eosinophil peroxidase, corticostatin-6, myeloperoxidase, and cathepsin C; and (2) drug/toxin metabolism and oxidative stress via GSTs, and ALDH1As modulations. IF analysis confirmed that Dex treatment significantly reversed SM-induced increases in human neutrophil peptides, defensin-5, and cathepsin C expression by 68%, 77%, and 90%, respectively. Thus, Dex markedly mitigated SM-induced limbal tissue injuries and prevented LSCD, via SM-induced inflammatory and oxidative stress inhibition, in our studies.
Collapse
Affiliation(s)
- Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA
| | - Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA
| | - Laura Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA
| | - Neera Tewari-Singh
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - J Mark Petrash
- Department of Ophthalmology, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, 12850 E. Montview Blvd, C238, Room V20-2118, Aurora, CO, 80045, USA.
| |
Collapse
|
3
|
Korkmaz-Icöz S, Szabó G, Gieldon A, McDonald PP, Dashkevich A, Yildirim AÖ, Korkmaz B. Protective effects of neutrophil serine protease inhibition against ischemia-reperfusion injury in lung or heart transplantation. FEBS J 2025. [PMID: 39854149 DOI: 10.1111/febs.17411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/15/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025]
Abstract
Transplanted organs are inevitably exposed to ischemia-reperfusion (IR) injury, which is known to cause graft dysfunction. Functional and structural changes that follow IR tissue injury are mediated by neutrophils through the production of oxygen-derived free radicals, as well as from degranulation which entails the release of proteases and other pro-inflammatory mediators. Neutrophil serine proteases (NSPs) are believed to be the principal triggers of post-ischemic reperfusion damage. Extended preservation times for the transplanted donor organ correlate with heightened occurrences of vascular damage and graft dysfunction. Preservation with α1-antitrypsin, an endogenous inhibitor of NSPs, improves primary graft function after lung or heart transplantation. Furthermore, pre-operative pharmacological targeting of NSP activation in the recipient using chemical inhibitors suppresses neutrophilic inflammation in transplanted organs. Hence, effective control of NSPs in the graft and recipient is a promising strategy to prevent IR injury. In this review, we describe the pathological functions of NSPs in IR injury and discuss their pharmacological inhibition to prevent primary graft dysfunction in lung or heart transplantation.
Collapse
Affiliation(s)
- Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University Hospital Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), Germany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), Germany
| | | | | | - Alexey Dashkevich
- University Department of Cardiac Surgery, Leipzig Heart Center, Leipzig, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, Ludwig-Maximilians University (LMU), Munich, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases (CEPR)", Tours, France
- Université de Tours, France
| |
Collapse
|
4
|
Arbiv OA, Quon BS. Disarming the cavalry: targeting neutrophils to limit collateral damage in non-CF bronchiectasis. Eur Respir J 2025; 65:2401804. [PMID: 39746771 DOI: 10.1183/13993003.01804-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 01/04/2025]
Affiliation(s)
- Omri A Arbiv
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management, and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Clinician-Investigator Program, University of British Columbia, Vancouver, BC, Canada
| | - Bradley S Quon
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
5
|
Liu S, Yang X, Zhao H, Zhao X, Fan K, Liu G, Li X, Du C, Liu J, Ma J. Cathepsin C exacerbates EAE by promoting the expansion of Tfh cells and the formation of TLSs in the CNS. Brain Behav Immun 2025; 123:123-142. [PMID: 39243987 DOI: 10.1016/j.bbi.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4+ T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35-55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4+ Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both in vivo and in vitro. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaohan Yang
- Department of Morphology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Henan Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xinnan Zhao
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Kai Fan
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Gang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Cong Du
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jianmei Ma
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
6
|
Chen X, Lou Y, Zhou F, Shi D, Liu X, Tao F. Identification of novel indolinone derivatives as CTSC inhibitors to treat inflammatory bowel disease by modulating inflammatory factors. Eur J Med Chem 2024; 280:116914. [PMID: 39383651 DOI: 10.1016/j.ejmech.2024.116914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024]
Abstract
Novel inflammatory bowel disease (IBD) therapeutic drugs, mainly biologics that neutralize pro-inflammatory factors and janus kinase inhibitors that inhibit cytokine-mediated signal transduction, face problems including low efficacy rates, limited therapeutic benefits, and infection risks. It is an important task to find proteins that broadly regulate a variety of cytokines and to develop corresponding drugs. Cathepsin C (CTSC) mediates neutrophil-related inflammatory, participates in the recruitment and activation of inflammatory cells, and regulates cytokines levels, and is considered an ideal target for IBD treatment. In this study, starting from the in-house molecule, through medicinal chemistry and target-based design, a novel CTSC inhibitor B22 with IBD therapeutic efficacy was discovered. In vitro target verification and mechanism study indicated that B22 inhibit CTSC activity by binding to S2 pocket and S1 site, further inhibiting downstream serine protease activity. In addition, B22 exhibited anti-inflammatory activity and regulated various cytokines levels. In vivo studies highlighted B22 bears acceptable toxicity and suitable pharmacokinetic properties, and displays anti-inflammatory activity in IBD model. In conclusion, B22 is a potential anti-inflammatory molecule for IBD by targeting CTSC and deserves further research.
Collapse
Affiliation(s)
- Xing Chen
- School of Public Health, Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230032, PR China
| | - Yan Lou
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Feilong Zhou
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Daxing Shi
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Xinhua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China.
| | - Fangbiao Tao
- School of Public Health, Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
7
|
Shen X, Li N, Liu M, Han X, Wang Y, Jia J, Wu F, Chen H, Liu X. Design and synthesis of novel cathepsin C inhibitors with anti-inflammatory activity. RSC Med Chem 2024:d4md00730a. [PMID: 39635544 PMCID: PMC11612923 DOI: 10.1039/d4md00730a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Cathepsin C (Cat C) is a potential candidate for addressing inflammatory conditions associated with neutrophil serine proteases (NSPs). The high reactivity of electrophilic warheads and the metabolic instability of peptide structures are among the primary challenges in developing potent cathepsin C inhibitors. Compound 36, a lead compound derived from compound 1 through structure-based drug design and structure-activity relationship (SAR), exhibited strong Cat C inhibitory activity with an IC50 value of 437 nM. It also showed a substantial enhancement in overall anti-inflammatory activity, achieving an inhibitory effect on NO release at 4.1 μM. Furthermore, molecular docking was conducted to analyze the mode of action with Cat C. And cell thermal shift analysis (CETSA) revealed that this compound increases the temperature tolerance of Cat C in a concentration-dependent manner, suggesting strong binding to the target Cat C. Prolonged pharmacological inhibition activity may result in the depletion of active NSPs.
Collapse
Affiliation(s)
- Xiaobao Shen
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Nan Li
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Miao Liu
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Xuanzheng Han
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Yazhi Wang
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Jingwen Jia
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Fufang Wu
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Hongwei Chen
- Medical School, Fuyang Normal University Fuyang 236037 P. R. China
| | - Xinhua Liu
- School of Pharmacy, Anhui Medical University Hefei 230601 P. R. China
| |
Collapse
|
8
|
Domain R, Seren S, Jerke U, Makridakis M, Chen KJ, Zoidakis J, Rhimi M, Zhang X, Bonvent T, Croix C, Gonzalez L, Li D, Basso J, Paget C, Viaud-Massuard MC, Lalmanach G, Shi GP, Aghdassi A, Vlahou A, McDonald PP, Couillin I, Williams R, Kettritz R, Korkmaz B. Pharmacological inhibition of cathepsin S and of NSPs-AAP-1 (a novel, alternative protease driving the activation of neutrophil serine proteases). Biochem Pharmacol 2024; 229:116114. [PMID: 39455238 DOI: 10.1016/j.bcp.2024.116114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 10/28/2024]
Abstract
An uncontrolled activity of neutrophil serine proteases (NSPs) contributes to inflammatory diseases. Cathepsin C (CatC) is known to activate NSPs during neutrophilic differentiation and represents a promising pharmacological target in NSP-mediated diseases. In humans, Papillon-Lefèvre syndrome (PLS) patients have mutations in theirCTSC gene, resulting in the complete absence of CatC activity. Despite this, low residual NSP activities are detected in PLS neutrophils (<10% vs healthy individuals), suggesting the involvement of CatC-independent proteolytic pathway(s) in the activation of proNSPs. This prompted us to characterize CatC-independent NSP activation pathways by blocking proCatC maturation. In this study, we show that inhibition of intracellular CatS almost completely blocked CatC maturation in human promyeloid HL-60 cells. Despite this, NSP activation was not significantly reduced, confirming the presence of a CatC-independent activation pathway involving a CatC-like protease that we termed NSPs-AAP-1. Similarly, when human CD34+ progenitor cells were treated with CatS inhibitors during neutrophilic differentiation in vitro, CatC activity was nearly abrogated but ∼30% NSP activities remained, further supporting the existence of NSPs-AAP-1. Our data indicate that NSPs-AAP-1 is a cysteine protease that is inhibited by reversible nitrile compounds designed for CatC inhibition. We further established a proof of concept for the indirect, although incomplete, inhibition of NSPs by pharmacological targeting of CatC maturation using CatS inhibitors. This emphasizes the potential of CatS as a therapeutic target for inflammatory diseases. Thus, preventing proNSP maturation using a CatS inhibitor, alone or in combination with a CatC/NSPs-AAP-1 inhibitor, represents a promising approach to efficiently control the extent of tissue injury in neutrophil-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Roxane Domain
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Seda Seren
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Uwe Jerke
- Experimental and Clinical Research Center, Charité und Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany
| | - Manousos Makridakis
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Kuan-Ju Chen
- Research Department, Insmed Incorporated, Bridgewater, NJ, USA
| | - Jérôme Zoidakis
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Moez Rhimi
- INRAE UMR-1319, Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Tillia Bonvent
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Cécile Croix
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Loïc Gonzalez
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Dedong Li
- Research Department, Insmed Incorporated, Bridgewater, NJ, USA
| | - Jessica Basso
- Research Department, Insmed Incorporated, Bridgewater, NJ, USA
| | - Christophe Paget
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Marie-Claude Viaud-Massuard
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Gilles Lalmanach
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France
| | - Guo-Ping Shi
- Department of Medicine, Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ali Aghdassi
- Department of Medicine A - Gastroenterology, Nephrology, Endocrinology and Rheumatology, University Medicine Greifswald, Greifswald, Germany
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Isabelle Couillin
- CNRS UMR-7355, Experimental and Molecular Immunology and Neurogenetics, Université d'Orléans, Orleans, France
| | - Rich Williams
- The Patrick G Johnston Center for Cancer Research, Queen's University, Belfast, UK
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité und Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany; Nephrology and Intensive Care Medicine, Charité-Universitätsmedizin, Berlin, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, Research Center for Respiratory Diseases, Tours, France; Université de Tours, Tours, France.
| |
Collapse
|
9
|
Zuo Y, Gong S, Zhang L, Zhou J, Wu JL, Li N. A Deep Mining Strategy for Peptide Rapid Identification in Lactobacillus reuteri Based on LC-MS/MS Integrated with FBMN and De Novo Sequencing. Metabolites 2024; 14:467. [PMID: 39330474 PMCID: PMC11434120 DOI: 10.3390/metabo14090467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Lactobacillus reuteri (L. reuteri) is widely recognized as a probiotic that produces prebiotics. However, studies on bioactive peptides or amino acid (AA) derivatives produced by L. reuteri are still lacking, whereas many bioactive peptides and AA derivatives have been found in other Lactobacillus species. In addition, rapid identification of peptides is challenged by the large amount of data and is limited by the coverage of protein databases. In this study, we performed a rapid and thorough profile of peptides in L. reuteri incorporating Global Natural Products Social Molecular Networking (GNPS) platform database searching, de novo sequencing, and deep mining, based on feature-based molecular networking (FBMN). According to FBMN, it was found that peptides containing identical or similar AA compositions were grouped into the same clusters, especially cyclic dipeptides (CDPs). Therefore, the grouping characteristics of clusters, differences in precursor ions, and characteristic fragment ions were utilized for the mining of deeply unknown compounds. Through this strategy, a total of 192 compounds, including 184 peptides, were rapidly identified. Among them, 53 CDPs, including four novel ones, were found for the first time in L. reuteri. Then, one of the novel CDPs, cyclo(5-OMe-Glu-4-OH-Pro), was isolated and characterized, which was consistent with the identification results. Moreover, some of the identified peptides exhibited considerable interactions with seven anti-inflammatory-related target proteins through molecular docking. According to the binding energies of peptides with different AA consistencies, it was considered that the existence of unnatural AAs in CDPs might contribute to their anti-inflammatory activity. These results provide a valuable strategy for the rapid identification of peptides, including CDPs. This study also reveals the substance basis for the potential anti-inflammatory effects exerted by L. reuteri.
Collapse
Affiliation(s)
| | | | | | | | - Jian-Lin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China; (Y.Z.); (S.G.); (L.Z.); (J.Z.)
| | - Na Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China; (Y.Z.); (S.G.); (L.Z.); (J.Z.)
| |
Collapse
|
10
|
Nishibata Y, Arai S, Taniguchi M, Nakade I, Ogawa H, Kitano S, Hosoi Y, Shindo A, Nishiyama R, Masuda S, Nakazawa D, Tomaru U, Shimizu T, Sinko W, Nagakura T, Terada Y, Ishizu A. Cathepsin C inhibition reduces neutrophil serine protease activity and improves activated neutrophil-mediated disorders. Nat Commun 2024; 15:6519. [PMID: 39174512 PMCID: PMC11341692 DOI: 10.1038/s41467-024-50747-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 07/18/2024] [Indexed: 08/24/2024] Open
Abstract
Cathepsin C (CatC) is an enzyme which regulates the maturation of neutrophil serine proteases (NSPs) essential for neutrophil activation. Activated neutrophils are key players in the innate immune system, and are also implicated in the etiology of various inflammatory diseases. This study aims to demonstrate a therapeutic potential for CatC inhibitors against disorders in which activated neutrophil-derived neutrophil extracellular traps (NETs) play a significant role. We demonstrate that a CatC inhibitor, MOD06051, dose-dependently suppresses the cellular activity of NSPs, including neutrophil elastase (NE), in vitro. Neutrophils derived from MOD06051-administered rats exhibit significantly lower NE activity and NET-forming ability than controls. Furthermore, MOD06051 dose-dependently ameliorates vasculitis and significantly decreases NETs when administered to a rat model of myeloperoxidase (MPO)-antineutrophil cytoplasmic antibody-associated vasculitis (AAV). These findings suggest that CatC inhibition is a promising strategy to reduce neutrophil activation and improve activated neutrophil-mediated diseases such as MPO-AAV.
Collapse
Affiliation(s)
- Yuka Nishibata
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Suishin Arai
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Mai Taniguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Issei Nakade
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hodaka Ogawa
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Shota Kitano
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yumeka Hosoi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Ayano Shindo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Ryo Nishiyama
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | | | | | | | | | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
11
|
Chitsamankhun C, Siritongtaworn N, Fournier BPJ, Sriwattanapong K, Theerapanon T, Samaranayake L, Porntaveetus T. Cathepsin C in health and disease: from structural insights to therapeutic prospects. J Transl Med 2024; 22:777. [PMID: 39164687 PMCID: PMC11337848 DOI: 10.1186/s12967-024-05589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Cathepsin C (CTSC) is a lysosomal cysteine protease constitutively expressed at high levels in the lung, kidney, liver, and spleen. It plays a key role in the activation of serine proteases in cytotoxic T cells, natural killer cells (granzymes A and B), mast cells (chymase and tryptase) and neutrophils (cathepsin G, neutrophil elastase, proteinase 3) underscoring its pivotal significance in immune and inflammatory defenses. Here, we comprehensively review the structural attributes, synthesis, and function of CTSC, with a focus on its variants implicated in the etiopathology of several syndromes associated with neutrophil serine proteases, including Papillon-Lefevre syndrome (PLS), Haim-Munk Syndrome (HMS), and aggressive periodontitis (AP). These syndromes are characterized by palmoplantar hyperkeratosis, and early-onset periodontitis (severe gum disease) resulting in premature tooth loss. Due to the critical role played by CTSC in these and several other conditions it is being explored as a potential therapeutic target for autoimmune and inflammatory disorders. The review also discusses in depth the gene variants of CTSC, and in particular their postulated association with chronic obstructive pulmonary disease (COPD), COVID-19, various cancers, anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, sudden cardiac death (SCD), atherosclerotic vascular disease, and neuroinflammatory disease. Finally, the therapeutic potential of CTSC across a range of human diseases is discussed.
Collapse
Affiliation(s)
- Chakriya Chitsamankhun
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nutwara Siritongtaworn
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - B P J Fournier
- Dental Faculty, Oral Biology Department, Reference Center of Oral and Dental Rare Diseases, Rothschild Hospital, Université Paris Cité, Paris, France
| | - Kanokwan Sriwattanapong
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanakorn Theerapanon
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Lakshman Samaranayake
- Faculty of Dentistry, University of Hong Kong, Hospital Road, Hong Kong, Hong Kong
- Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Graduate Program in Geriatric and Special Patients Care, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
12
|
Brundage J, Barrios JP, Tison GH, Pirruccello JP. Genetics of Cardiac Aging Implicate Organ-Specific Variation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.02.24310874. [PMID: 39148824 PMCID: PMC11326326 DOI: 10.1101/2024.08.02.24310874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Heart structure and function change with age, and the notion that the heart may age faster for some individuals than for others has driven interest in estimating cardiac age acceleration. However, current approaches have limited feature richness (heart measurements; radiomics) or capture extraneous data and therefore lack cardiac specificity (deep learning [DL] on unmasked chest MRI). These technical limitations have been a barrier to efforts to understand genetic contributions to age acceleration. We hypothesized that a video-based DL model provided with heart-masked MRI data would capture a rich yet cardiac-specific representation of cardiac aging. In 61,691 UK Biobank participants, we excluded noncardiac pixels from cardiac MRI and trained a video-based DL model to predict age from one cardiac cycle in the 4-chamber view. We then computed cardiac age acceleration as the bias-corrected prediction of heart age minus the calendar age. Predicted heart age explained 71.1% of variance in calendar age, with a mean absolute error of 3.3 years. Cardiac age acceleration was linked to unfavorable cardiac geometry and systolic and diastolic dysfunction. We also observed links between cardiac age acceleration and diet, decreased physical activity, increased alcohol and tobacco use, and altered levels of 239 serum proteins, as well as adverse brain MRI characteristics. We found cardiac age acceleration to be heritable (h2g 26.6%); a genome-wide association study identified 8 loci related to linked to cardiomyopathy (near TTN, TNS1, LSM3, PALLD, DSP, PLEC, ANKRD1 and MYO18B) and an additional 16 loci (near MECOM, NPR3, KLHL3, HDGFL1, CDKN1A, ELN, SLC25A37, PI15, AP3M1, HMGA2, ADPRHL1, PGAP3, WNT9B, UHRF1 and DOK5). Of the discovered loci, 21 were not previously associated with cardiac age acceleration. Mendelian randomization revealed that lower genetically mediated levels of 6 circulating proteins (MSRA most strongly), as well as greater levels of 5 proteins (LXN most strongly) were associated with cardiac age acceleration, as were greater blood pressure and Lp(a). A polygenic score for cardiac age acceleration predicted earlier onset of arrhythmia, heart failure, myocardial infarction, and mortality. These findings provide a thematic understanding of cardiac age acceleration and suggest that heart- and vascular-specific factors are key to cardiac age acceleration, predominating over a more global aging program.
Collapse
Affiliation(s)
- James Brundage
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Joshua P. Barrios
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
| | - Geoffrey H. Tison
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Center for Biosignal Research, University of California San Francisco, San Francisco, CA, USA
| | - James P. Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Genetics Center, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Center for Biosignal Research, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
14
|
Herder C, Thorand B, Strom A, Rathmann W, Heier M, Koenig W, Morrison H, Ziegler D, Roden M, Peters A, Bönhof GJ, Maalmi H. Associations between multiple neurological biomarkers and distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Metab Res Rev 2024; 40:e3807. [PMID: 38872492 DOI: 10.1002/dmrr.3807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/25/2024] [Accepted: 04/22/2024] [Indexed: 06/15/2024]
Abstract
AIMS The aim of this study was to assess associations between neurological biomarkers and distal sensorimotor polyneuropathy (DSPN). MATERIALS AND METHODS Cross-sectional analyses were based on 1032 participants aged 61-82 years from the population-based KORA F4 survey, 177 of whom had DSPN at baseline. The prevalence of type 2 diabetes was 20%. Prospective analyses used data from 505 participants without DSPN at baseline, of whom 125 had developed DSPN until the KORA FF4 survey. DSPN was defined based on the examination part of the Michigan Neuropathy Screening Instrument. Serum levels of neurological biomarkers were measured using proximity extension assay technology. Associations between 88 biomarkers and prevalent or incident DSPN were estimated using Poisson regression with robust error variance and are expressed as risk ratios (RR) and 95% CI per 1-SD increase. Results were adjusted for multiple confounders and multiple testing using the Benjamini-Hochberg procedure. RESULTS Higher serum levels of CTSC (cathepsin C; RR [95% CI] 1.23 (1.08; 1.39), pB-H = 0.044) and PDGFRα (platelet-derived growth factor receptor A; RR [95% CI] 1.21 (1.08; 1.35), pB-H = 0.044) were associated with prevalent DSPN in the total study sample. CDH3, JAM-B, LAYN, RGMA and SCARA5 were positively associated with DSPN in the diabetes subgroup, whereas GCP5 was positively associated with DSPN in people without diabetes (all pB-H for interaction <0.05). None of the biomarkers showed an association with incident DSPN (all pB-H>0.05). CONCLUSIONS This study identified multiple novel associations between neurological biomarkers and prevalent DSPN, which may be attributable to functions of these proteins in neuroinflammation, neural development and myelination.
Collapse
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Partner Neuherberg, Partner Düsseldorf, Munich, Germany
- Institute for Medical Information Processing Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
| | - Wolfgang Rathmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- KORA Study Centre, University Hospital of Augsburg, Augsburg, Germany
| | - Wolfgang Koenig
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), Partner Site München Heart Alliance, Munich, Germany
| | - Helen Morrison
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Partner Neuherberg, Partner Düsseldorf, Munich, Germany
- Institute for Medical Information Processing Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Haifa Maalmi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich, Germany
| |
Collapse
|
15
|
Li Q, Wan C, Zhang Z, Liu G, Wang S. CTSC promoted the migration and invasion of glioma cells via activation of STAT3/SERPINA3 axis. Gene 2024; 893:147948. [PMID: 37925117 DOI: 10.1016/j.gene.2023.147948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/15/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Cathepsin C (CTSC) has been reported to be upregulated in several cancers, however, there are still many missing links about the role of CTSC in glioma. To address this knowledge gap, the present study employed bioinformatics analysis, Transwell assay, RT-qPCR and Western blot assays to investigate the expression level of CTSC in glioma tissues, its relationship with survival period, and its effect on the migration and invasion ability of glioma cells. The findings revealed that CTSC was upregulated in glioma and was associated with poor prognosis. Moreover, CTSC was found to promote cell migration and invasion abilities as well as epithelial-mesenchymal transition (EMT). A further study found that CTSC induced SERPINA3 and STAT3 expression in glioma cells. Additionally, we demonstrated that STAT3 signaling mediated upregulation of SERPINA3 expression by CTSC. In sum, our findings suggest that CTSC activates the STAT3/SERPINA3 axis to promote migration and invasion of glioma cells, which may lead to new potential therapeutic approaches for humans with cancer.
Collapse
Affiliation(s)
- Qi Li
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, China
| | - Chenguang Wan
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, China
| | - Zhifei Zhang
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, China
| | - Guangwei Liu
- Department of Otolaryngology, Tianjin Third Central Hospital, Tianjin, China
| | - Song Wang
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
16
|
Aghdassi AA, Pham C, Zierke L, Mariaule V, Korkmaz B, Rhimi M. Cathepsin C role in inflammatory gastroenterological, renal, rheumatic, and pulmonary disorders. Biochimie 2024; 216:175-180. [PMID: 37758158 DOI: 10.1016/j.biochi.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/27/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023]
Abstract
Cathepsin C (CatC, syn. Dipeptidyl peptidase I) is a lysosomal cysteine proteinase expressed in several tissues including inflammatory cells. This enzyme is important for maintaining multiple cellular functions and for processing immune cell-derived proteases. While mutations in the CatC gene were reported in Papillon-Lefèvre syndrome, a rare autosomal recessive disorder featuring hyperkeratosis and periodontitis, evidence from clinical and preclinical studies points toward pro-inflammatory effects of CatC in various disease processes that are mainly mediated by the activation of neutrophil serine proteinases. Moreover, tumor-promoting effects were ascribed to CatC. The aim of this review is to highlight current knowledge of the CatC as a potential therapeutic target in inflammatory disorders.
Collapse
Affiliation(s)
- Ali A Aghdassi
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Christine Pham
- Division of Rheumatology, Washington University in St. Louis, St. Louis, MO, USA
| | - Lukas Zierke
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Vincent Mariaule
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, University of Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032, Tours, France
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, University of Paris-Saclay, INRAE, Jouy-en-Josas, France.
| |
Collapse
|
17
|
Chalmers JD, Kettritz R, Korkmaz B. Dipeptidyl peptidase 1 inhibition as a potential therapeutic approach in neutrophil-mediated inflammatory disease. Front Immunol 2023; 14:1239151. [PMID: 38162644 PMCID: PMC10755895 DOI: 10.3389/fimmu.2023.1239151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024] Open
Abstract
Neutrophils have a critical role in the innate immune response to infection and the control of inflammation. A key component of this process is the release of neutrophil serine proteases (NSPs), primarily neutrophil elastase, proteinase 3, cathepsin G, and NSP4, which have essential functions in immune modulation and tissue repair following injury. Normally, NSP activity is controlled and modulated by endogenous antiproteases. However, disruption of this homeostatic relationship can cause diseases in which neutrophilic inflammation is central to the pathology, such as chronic obstructive pulmonary disease (COPD), alpha-1 antitrypsin deficiency, bronchiectasis, and cystic fibrosis, as well as many non-pulmonary pathologies. Although the pathobiology of these diseases varies, evidence indicates that excessive NSP activity is common and a principal mediator of tissue damage and clinical decline. NSPs are synthesized as inactive zymogens and activated primarily by the ubiquitous enzyme dipeptidyl peptidase 1, also known as cathepsin C. Preclinical data confirm that inactivation of this protease reduces activation of NSPs. Thus, pharmacological inhibition of dipeptidyl peptidase 1 potentially reduces the contribution of aberrant NSP activity to the severity and/or progression of multiple inflammatory diseases. Initial clinical data support this view. Ongoing research continues to explore the role of NSP activation by dipeptidyl peptidase 1 in different disease states and the potential clinical benefits of dipeptidyl peptidase 1 inhibition.
Collapse
Affiliation(s)
- James D. Chalmers
- Department of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, Research Center for Respiratory Diseases, University of Tours, Tours, France
| |
Collapse
|
18
|
Liu B, Korkmaz B, Kraft P, Mayer T, Sayour AA, Grundl MA, Domain R, Karck M, Szabó G, Korkmaz-Icöz S. Pharmacological inhibition of the cysteine protease cathepsin C improves graft function after heart transplantation in rats. J Transl Med 2023; 21:799. [PMID: 37946197 PMCID: PMC10636924 DOI: 10.1186/s12967-023-04659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Heart transplantation (HTX) is the standard treatment for end-stage heart failure. However, reperfusion following an ischemic period can contribute to myocardial injury. Neutrophil infiltration, along with the subsequent release of tissue-degrading neutrophil elastase (NE)-related serine proteases and oxygen-derived radicals, is associated with adverse graft outcomes. The inhibition of cathepsin C (CatC) has been shown to block NE-related protease activation. We hypothesized that the CatC inhibitor BI-9740 improves graft function after HTX. METHODS In a rat model of HTX, the recipient Lewis rats were orally administered with either a placebo (n = 12) or BI-9740 (n = 11, 20 mg/kg) once daily for 12 days. Donor hearts from untreated Lewis rats were explanted, preserved in a cardioplegic solution, and subsequently heterotopically implanted. In vivo left-ventricular (LV) graft function was assessed after 1 h of reperfusion. The proteolytic activity of neutrophil serine proteases was determined in bone marrow lysates from BI-9740-treated and control rats. Additionally, myocardial morphological changes were examined, and heart samples underwent immunohistochemistry and western blot analysis. RESULTS The NE-related proteolytic activity in bone marrow cell lysates was markedly decreased in the BI-9740-treated rats compared to those of the placebo group. Histopathological lesions, elevated CatC and myeloperoxidase-positive cell infiltration, and nitrotyrosine immunoreactivity with an increased number of poly(ADP-ribose) polymerase (PARP)-1-positive cells were lowered in the hearts of animals treated with BI-9740 compared to placebo groups. Regarding the functional parameters of the implanted graft, improvements were observed in both systolic function (LV systolic pressure 110 ± 6 vs 74 ± 6 mmHg; dP/dtmax 2782 ± 149 vs 2076 ± 167 mmHg/s, LV developed pressure, at an intraventricular volume of 200 µl, p < 0.05) and diastolic function in the hearts of BI-9740 treated animals compared with those receiving the only placebo. Furthermore, the administration of BI-9740 resulted in a shorter graft re-beating time compared to the placebo group. However, this study did not provide evidence of DNA fragmentation, the generation of both superoxide anions and hydrogen peroxide, correlating with the absence of protein alterations related to apoptosis, as evidenced by western blot in grafts after HTX. CONCLUSIONS We provided experimental evidence that pharmacological inhibition of CatC improves graft function following HTX in rats.
Collapse
Affiliation(s)
- Baoer Liu
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), 06120, Halle, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and Université de Tours, 37032, Tours, France
| | - Patricia Kraft
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Tobias Mayer
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Alex A Sayour
- Heart and Vascular Center, Semmelweis University, Budapest, 1122, Hungary
| | - Marc A Grundl
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach a.d. Riss, Germany
| | - Roxane Domain
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and Université de Tours, 37032, Tours, France
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), 06120, Halle, Germany
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120, Heidelberg, Germany.
- Department of Cardiac Surgery, University Hospital Halle (Saale), 06120, Halle, Germany.
| |
Collapse
|
19
|
Abstract
Mast cell granules are packed with proteases, which are released with other mediators by degranulating stimuli. Several of these proteases are targets of potentially therapeutic inhibitors based on hypothesized contributions to diseases, notably asthma and ulcerative colitis for β-tryptases, heart and kidney scarring for chymases, and airway infection for dipeptidyl peptidase-I. Small-molecule and antibody-based β-tryptase inhibitors showing preclinical promise were tested in early-phase human trials with some evidence of benefit. Chymase inhibitors were given safely in Phase II trials without demonstrating benefits, whereas dipeptidyl peptidase-I inhibitor improved bronchiectasis, in effects likely related to inactivation of the enzyme in neutrophils.
Collapse
|
20
|
Song J, Zhu N, Pan X, Guo L, Kong X. Expression and significance of cathepsin C and cathepsin D during pregnancy and Preeclampsia. Reprod Biol Endocrinol 2023; 21:92. [PMID: 37794357 PMCID: PMC10548605 DOI: 10.1186/s12958-023-01138-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Cathepsin C (Cat C) is involved in the inflammatory-immune system and can be degraded by cathepsin D (Cat D). Preeclampsia (PE) and the inflammation-immunity relationship is currently a hot research topic, but there are still few studies. The aim was to investigate the expression and significance of Cat C and D in the serum of nonpregnant women, patients in various stages of pregnancy and patients with PE, and in the placenta of patients with normal pregnancy and PE. METHODS Sixty young healthy nonpregnant women were selected: 180 normal pregnant women, including 60 each in the first, second, and third trimesters, and 100 women with PE, including 39 women with severe preeclampsia. The levels of Cat C and D in serum were detected by enzyme-linked immunosorbent assay (ELISA), and the expression levels of Cat C and D in placentas were detected by immunohistochemistry (IHC). RESULTS The serum of Cat C in the first trimester was significantly lower than that in the nonpregnant group (P < 0.001), whereas Cat D was significantly higher than that in the nonpregnant group (P < 0.01). The levels of Cat C and D in the second trimester and third trimester were significantly higher than those in the first trimester (P < 0.05), but there was no significant difference in Cat C and D between the second trimester and third trimester. The levels of Cat C in the serum and placentas of patients with PE were significantly higher than those in the third trimester (P < 0.001) and positively correlated with the severity of PE (P < 0.001), whereas the levels of Cat D in the serum and placentas of patients with PE were significantly lower than those in the third trimester (P < 0.001) and negatively correlated with the severity of PE (P < 0.001). Age, primigravida proportion, and body mass index were significantly higher in the PE group than in the control group (P < 0.05), which were high-risk factors for PE. CONCLUSIONS Cat C and D are associated with the maintenance of normal pregnancy. In patients with preeclampsia, a significant increase in Cat C and a significant decrease in Cat D levels may lead to the occurrence and development of preeclampsia.
Collapse
Affiliation(s)
- Jingzhe Song
- Department of Obstetrics and Gynecology, Clinical Medical College, Yangzhou University, 225001, Yangzhou, Jiangsu, China
| | - Nan Zhu
- Department of Obstetrics and Gynecology, Clinical Medical College, Yangzhou University, 225001, Yangzhou, Jiangsu, China.
- Graduate School of Dalian Medical University, Dalian, China.
| | - Xinchen Pan
- Department of Obstetrics and Gynecology, Clinical Medical College, Yangzhou University, 225001, Yangzhou, Jiangsu, China
| | - Lu Guo
- School of Nursing and School of Public Health, Yangzhou University, Yangzhou, China
| | - Xiang Kong
- Department of Obstetrics and Gynecology, Clinical Medical College, Yangzhou University, 225001, Yangzhou, Jiangsu, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu Province, China.
| |
Collapse
|
21
|
Abideen SA, Khan M, Al-Harbi AI, Ahmad S. Pharmacological inhibition of cathepsin C (CatC) as a potential approach for cancer therapeutics. J Biomol Struct Dyn 2023; 41:8682-8689. [PMID: 36264138 DOI: 10.1080/07391102.2022.2135603] [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: 07/26/2022] [Accepted: 10/08/2022] [Indexed: 10/24/2022]
Abstract
Studies have established that proteolytic enzyme inhibition holds significant promise in cancer prevention and treatment. Cathepsin C (CatC) is conserved lysosomal cysteine dipeptidyl aminopeptidase, which is the key for pro-inflammatory neutrophil serine protease activation and biological functioning. This makes CatC as a promising therapeutic drug target for the management of different cancer types. Considering this, using a wide range of computer aided drug-designing applications, several inhibitors are shortlisted against CatC active pocket, which interact with the enzyme with high affinity and form strong intermolecular interaction network. Compared to control, three molecules ASN_06916232, ASN_06917112 and ASN_06916892 are filtered as best binders of the CatC active pocket with binding energy value of -10.9 kcal/mol, -10.9 kcal/mol and -10.7 kcal/mol, respectively. These compounds interact with several important active side residues of CatC such as Ser233, Cys234, Gly277, Asn380 and His38. Furthermore, the complexes of these compounds with CatC reveal very stable dynamics with average RMSD value less than 3 Å. The binding energy analysis further indicates the compounds to have very stable van der Waals and electrostatic energies. In conclusion, these molecules are promising and require experimental validation to prove them as anti-CatC molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Syed Ainul Abideen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Murad Khan
- Shanghai Center for System Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alhanouf I Al-Harbi
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu, Saudi Arabia
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| |
Collapse
|
22
|
Thornton LB, Key M, Micchelli C, Stasic AJ, Kwain S, Floyd K, Moreno SN, Dominy BN, Whitehead DC, Dou Z. A cathepsin C-like protease mediates the post-translation modification of Toxoplasma gondii secretory proteins for optimal invasion and egress. mBio 2023; 14:e0017423. [PMID: 37326431 PMCID: PMC10470614 DOI: 10.1128/mbio.00174-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/01/2023] [Indexed: 06/17/2023] Open
Abstract
Microbial pathogens use proteases for their infections, such as digestion of proteins for nutrients and activation of their virulence factors. As an obligate intracellular parasite, Toxoplasma gondii must invade host cells to establish its intracellular propagation. To facilitate invasion, the parasites secrete invasion effectors from microneme and rhoptry, two unique organelles in apicomplexans. Previous work has shown that some micronemal invasion effectors experience a series of proteolytic cleavages within the parasite's secretion pathway for maturation, such as the aspartyl protease (TgASP3) and the cathepsin L-like protease (TgCPL), localized within the post-Golgi compartment and the endolysosomal system, respectively. Furthermore, it has been shown that the precise maturation of micronemal effectors is critical for Toxoplasma invasion and egress. Here, we show that an endosome-like compartment (ELC)-residing cathepsin C-like protease (TgCPC1) mediates the final trimming of some micronemal effectors, and its loss further results in defects in the steps of invasion, egress, and migration throughout the parasite's lytic cycle. Notably, the deletion of TgCPC1 completely blocks the activation of subtilisin-like protease 1 (TgSUB1) in the parasites, which globally impairs the surface-trimming of many key micronemal invasion and egress effectors. Additionally, we found that Toxoplasma is not efficiently inhibited by the chemical inhibitor targeting the malarial CPC ortholog, suggesting that these cathepsin C-like orthologs are structurally different within the apicomplexan phylum. Collectively, our findings identify a novel function of TgCPC1 in processing micronemal proteins within the Toxoplasma parasite's secretory pathway and expand the understanding of the roles of cathepsin C protease. IMPORTANCE Toxoplasma gondii is a microbial pathogen that is well adapted for disseminating infections. It can infect virtually all warm-blooded animals. Approximately one-third of the human population carries toxoplasmosis. During infection, the parasites sequentially secrete protein effectors from the microneme, rhoptry, and dense granule, three organelles exclusively found in apicomplexan parasites, to help establish their lytic cycle. Proteolytic cleavage of these secretory proteins is required for the parasite's optimal function. Previous work has revealed that two proteases residing within the parasite's secretory pathway cleave micronemal and rhoptry proteins, which mediate parasite invasion and egress. Here, we demonstrate that a cathepsin C-like protease (TgCPC1) is involved in processing several invasion and egress effectors. The genetic deletion of TgCPC1 prevented the complete maturation of some effectors in the parasites. Strikingly, the deletion led to a full inactivation of one surface-anchored protease, which globally impaired the trimming of some key micronemal proteins before secretion. Therefore, this finding represents a novel post-translational mechanism for the processing of virulence factors within microbial pathogens.
Collapse
Affiliation(s)
- L. Brock Thornton
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Melanie Key
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Chiara Micchelli
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Andrew J. Stasic
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Samuel Kwain
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| | - Katherine Floyd
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Silvia N.J. Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Brian N. Dominy
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| | - Daniel C. Whitehead
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Zhicheng Dou
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
23
|
Chen KJ, Zhang J, LaSala D, Basso J, Chun D, Zhou Y, McDonald PP, Perkins WR, Cipolla DC. Brensocatib, an oral, reversible inhibitor of dipeptidyl peptidase 1, mitigates interferon-α-accelerated lupus nephritis in mice. Front Immunol 2023; 14:1185727. [PMID: 37441081 PMCID: PMC10333524 DOI: 10.3389/fimmu.2023.1185727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Neutrophils have been implicated in initiating and perpetuating systemic lupus erythematosus and the resultant kidney damage in lupus nephritis (LN) patients, in part through an excessive release of neutrophil serine proteases (NSPs). NSP zymogens are activated by dipeptidyl peptidase 1 (DPP1) during neutrophil maturation and released by mature neutrophils in response to inflammatory stimuli. Thus, a potential strategy to attenuate disease progression in LN would be to inhibit DPP1. We tested whether brensocatib, a highly selective and reversible DPP1 inhibitor, could mitigate LN progression in an interferon-alpha (IFNα)-accelerated NZB/W F1 mouse model. To confirm brensocatib's pharmacodynamic effect on NSPs in this mouse strain, repeated dose studies were conducted for 7 and 14 days in naïve NZB/W F1 mice via oral gavage twice a day. Brensocatib at 2 and 20 mg/kg/day achieved a significant reduction in bone marrow NSP activities after 7 days of daily administration. To initiate LN disease progression, the mice were injected with an IFNα-expressing adenovirus. After 2 weeks, three brensocatib doses (or vehicle) were administered for 6 more weeks. Throughout the 8-week study, brensocatib treatment (20 mg/kg/day) significantly reduced the occurrence of severe proteinuria compared to the vehicle control. Brensocatib treatment also entailed a significant reduction in the urine albumin-to-creatinine ratio, indicating decreased kidney damage, as well as a significant reduction in blood urea nitrogen level, suggesting improved renal function. Based on kidney histopathology analysis, brensocatib treatment significantly lowered both the renal tubular protein score and the nephropathy score compared to the vehicle group. A trend towards reduced glomerulonephritis score with brensocatib treatment was also observed. Lastly, brensocatib significantly reduced LN mouse kidney infiltration by various inflammatory cells. In conclusion, these results suggest that brensocatib alters disease progression in LN mice and warrant further evaluation of DPP1 inhibition in LN.
Collapse
|
24
|
Jiang H, Dong Z, Xia X, Li X. Cathepsins in oral diseases: mechanisms and therapeutic implications. Front Immunol 2023; 14:1203071. [PMID: 37334378 PMCID: PMC10272612 DOI: 10.3389/fimmu.2023.1203071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Cathepsins are a type of lysosomal globulin hydrolase and are crucial for many physiological processes, including the resorption of bone matrix, innate immunity, apoptosis, proliferation, metastasis, autophagy, and angiogenesis. Findings regarding their functions in human physiological processes and disorders have drawn extensive attention. In this review, we will focus on the relationship between cathepsins and oral diseases. We highlight the structural and functional properties of cathepsins related to oral diseases, as well as the regulatory mechanisms in tissue and cells and their therapeutic uses. Elucidating the associated mechanism between cathepsins and oral diseases is thought to be a promising strategy for the treatment of oral diseases and may be a starting point for further studies at the molecular level.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Zuoxiang Dong
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xiaomin Xia
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Xue Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Xu T, Xie K, Wang C, Ivanovski S, Zhou Y. Immunomodulatory nanotherapeutic approaches for periodontal tissue regeneration. NANOSCALE 2023; 15:5992-6008. [PMID: 36896757 DOI: 10.1039/d2nr06149j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is an infection-induced inflammatory disease characterized by progressive destruction of tooth supporting tissues, which, if left untreated, can result in tooth loss. The destruction of periodontal tissues is primarily caused by an imbalance between the host immune protection and immune destruction mechanisms. The ultimate goal of periodontal therapy is to eliminate inflammation and promote the repair and regeneration of both hard and soft tissues, so as to restore the physiological structure and function of periodontium. Advancement in nanotechnologies has enabled the development of nanomaterials with immunomodulatory properties for regenerative dentistry. This review discusses the immune mechanisms of the major effector cells in the innate and adaptive immune systems, the physicochemical and biological properties of nanomaterials, and the research advancements in immunomodulatory nanotherapeutic approaches for the management of periodontitis and the regeneration of periodontal tissues. The current challenges, and prospects for future applications of nanomaterials are then discussed so that researchers at the intersections of osteoimmunology, regenerative dentistry and materiobiology will continue to advance the development of nanomaterials for improved periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tian Xu
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Kunke Xie
- Clinical Laboratory, Bo'Ai Hospital of Zhongshan, 6 Chenggui Road, East District, Zhongshan 528403, Guangdong, China
| | - Cong Wang
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Yinghong Zhou
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| |
Collapse
|
26
|
Carla Guarino, Seren S, Lemoine R, Hummel A, Margotin JE, El-Benna J, Hoarau C, Specks U, Jenne D, Korkmaz B. Constitutive and induced forms of membrane-bound proteinase 3 interact with antineutrophil cytoplasmic antibodies and promote immune activation of neutrophils. J Biol Chem 2023; 299:103072. [PMID: 36849007 PMCID: PMC10124916 DOI: 10.1016/j.jbc.2023.103072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023] Open
Abstract
Proteinase 3 (PR3) is the main target antigen of anti-neutrophil cytoplasmic antibodies (ANCA) in PR3-ANCA-associated vasculitis. A small fraction of PR3 is constitutively exposed on the surface of quiescent blood neutrophils in a proteolytically inactive form. When activated, neutrophils expose an induced form of membrane-bound PR3 (PR3mb) on their surface as well, which is enzymatically less active than unbound PR3 in solution due to its altered conformation. In this work, our objective was to understand the respective role of constitutive and induced PR3mb in the immune activation of neutrophils triggered by murine anti-PR3 mAbs and human PR3-ANCA. We quantified immune activation of neutrophils by the measurement of the production of superoxide anions and secreted protease activity in the cell supernatant before and after treatment of the cells by alpha-1 protease inhibitor (α1PI) that clears induced PR3mb from the cell surface. Incubation of TNFα-primed neutrophils with anti-PR3 antibodies resulted in a significant increase in superoxide anion production, membrane activation marker exposition, and secreted protease activity. When primed neutrophils were first treated with α1PI, we observed a partial reduction in antibody-induced neutrophil activation, suggesting that constitutive PR3mb is sufficient to activate neutrophils. The pre-treatment of primed neutrophils with purified antigen-binding fragments used as competitor significantly reduced cell activation by whole antibodies. This led us to the conclusion that PR3mb promoted immune activation of neutrophils. We propose that blocking and/or elimination of PR3mb offers a new therapeutic strategy to attenuate neutrophil activation in patients with PR3-ANCA-associated vasculitis.
Collapse
Affiliation(s)
- Carla Guarino
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, F-37032, Tours, France
| | - Seda Seren
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, F-37032, Tours, France
| | - Roxane Lemoine
- EA4245 "Transplantation, Immunology and Inflammation", University of Tours, France and Clinical immunology and allergology Service, Tours University Hospital, F-37032, Tours, France
| | - AmberM Hummel
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | - Jean-Edouard Margotin
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, F-37032, Tours, France
| | - Jamel El-Benna
- Université de Paris, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, Faculté de Médecine Xavier Bichat, F-75018, Paris, France
| | - Cyrille Hoarau
- EA4245 "Transplantation, Immunology and Inflammation", University of Tours, France and Clinical immunology and allergology Service, Tours University Hospital, F-37032, Tours, France
| | - Ulrich Specks
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | - DieterE Jenne
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, German Center for Lung Research (DZL), 81377 Munich and Max Planck Institute of Neurobiology, 82152 Planegg-Martinsried, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, F-37032, Tours, France.
| |
Collapse
|
27
|
Thornton LB, Key M, Micchelli C, Stasic AJ, Kwain S, Floyd K, Moreno SNJ, Dominy BN, Whitehead DC, Dou Z. A cathepsin C-like protease post-translationally modifies Toxoplasma gondii secretory proteins for optimal invasion and egress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.525043. [PMID: 36712013 PMCID: PMC9882377 DOI: 10.1101/2023.01.21.525043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Microbial pathogens use proteases for their infections, such as digestion of proteins for nutrients and activation of their virulence factors. As an obligate intracellular parasite, Toxoplasma gondii must invade host cells to establish its intracellular propagation. To facilitate invasion, the parasites secrete invasion effectors from microneme and rhoptry, two unique organelles in apicomplexans. Previous work has shown that some micronemal invasion effectors experience a series of proteolytic cleavages within the parasite's secretion pathway for maturation, such as the aspartyl protease (TgASP3) and the cathepsin L-like protease (TgCPL), localized within the post-Golgi compartment (1) and the endolysosomal system (2), respectively. Furthermore, it has been shown that the precise maturation of micronemal effectors is critical for Toxoplasma invasion and egress (1). Here, we show that an endosome-like compartment (ELC)-residing cathepsin C-like protease (TgCPC1) mediates the final trimming of some micronemal effectors, and its loss further results in defects in the steps of invasion, egress, and migration throughout the parasite's lytic cycle. Notably, the deletion of TgCPC1 completely blocks the activation of subtilisin-like protease 1 (TgSUB1) in the parasites, which globally impairs the surface-trimming of many key micronemal invasion and egress effectors. Additionally, we found that TgCPC1 was not efficiently inhibited by the chemical inhibitor targeting its malarial ortholog, suggesting that these cathepsin C-like orthologs are structurally different within the apicomplexan phylum. Taken together, our findings identify a novel function of TgCPC1 in the processing of micronemal proteins within the secretory pathway of Toxoplasma parasites and expand the understanding of the roles of cathepsin C protease. IMPORTANCE Toxoplasma gondii is a microbial pathogen that is well adapted for disseminating infections. It can infect virtually all warm-blooded animals. Approximately one-third of the human population carries toxoplasmosis. During infection, the parasites sequentially secrete protein effectors from the microneme, rhoptry, and dense granule, three organelles exclusively found in apicomplexan parasites, to help establish their lytic cycle. Proteolytic cleavage of these secretory proteins is required for the parasite's optimal function. Previous work has revealed that two proteases residing within the parasite's secretory pathway cleave micronemal and rhoptry proteins, which mediate parasite invasion and egress. Here, we demonstrate that a cathepsin C-like protease (TgCPC1) is involved in processing several invasion and egress effectors. The genetic deletion of TgCPC1 prevented the complete maturation of some effectors in the parasites. Strikingly, the deletion led to a full inactivation of one surface-anchored protease, which globally impaired the trimming of some key micronemal proteins before secretion. Therefore, this finding represents a novel post-translational mechanism for the processing of virulence factors within microbial pathogens.
Collapse
Affiliation(s)
- L. Brock Thornton
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Melanie Key
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Chiara Micchelli
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Andrew J. Stasic
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Samuel Kwain
- Department of Chemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Katherine Floyd
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Silvia N. J. Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Brian N. Dominy
- Department of Chemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Daniel C. Whitehead
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, United States of America
- Department of Chemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Zhicheng Dou
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, United States of America
| |
Collapse
|
28
|
Wątroba K, Pawełczak M, Kaźmierczak M. Dipeptide analogues of fluorinated aminophosphonic acid sodium salts as moderate competitive inhibitors of cathepsin C. Beilstein J Org Chem 2023; 19:434-439. [PMID: 37091732 PMCID: PMC10113521 DOI: 10.3762/bjoc.19.33] [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: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 04/25/2023] Open
Abstract
In this paper, we present the solvolysis reaction of dipeptide analogues of fluorinated aminophosphonates with simultaneous quantitative deprotection of the amino group. To the best of our knowledge, this work is the first reported example of the application of fluorinated aminophosphonates in cathepsin C inhibition studies. The new molecules show moderate inhibition of the cathepsin C enzyme, which opens the door to consider them as potential therapeutic agents. Overall, our findings provide a new avenue for the development of fluorinated aminophosphonate-based inhibitors.
Collapse
Affiliation(s)
- Karolina Wątroba
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | | | - Marcin Kaźmierczak
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
- Centre for Advanced Technologies, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 10, 61-614 Poznań, Poland
| |
Collapse
|
29
|
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: 6] [Impact Index Per Article: 2.0] [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
|
30
|
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: 3.0] [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
|
31
|
Scurt FG, Bose K, Hammoud B, Brandt S, Bernhardt A, Gross C, Mertens PR, Chatzikyrkou C. Old known and possible new biomarkers of ANCA-associated vasculitis. J Autoimmun 2022; 133:102953. [PMID: 36410262 DOI: 10.1016/j.jaut.2022.102953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/06/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) comprises a group of multisystem disorders involving severe, systemic, small-vessel vasculitis with short- and long term serious and life-threating complications. Despite the simplification of treatment, fundamental aspects concerning assessment of its efficacy and its adaptation to encountered complications or to the relapsing/remitting/subclinical disease course remain still unknown. The pathogenesis of AAV is complex and unique, and despite the progress achieved in the last years, much has not to be learnt. Foremost, there is still no accurate marker enabling us to monitoring disease and guide therapy. Therefore, the disease management relays often on clinical judgment and follows a" trial and error approach". In the recent years, an increasing number of new molecules s have been explored and used for this purpose including genomics, B- and T-cell subpopulations, complement system factors, cytokines, metabolomics, biospectroscopy and components of our microbiome. The aim of this review is to discuss both the role of known historical and clinically established biomarkers of AAV, as well as to highlight potential new ones, which could be used for timely diagnosis and monitoring of this devastating disease, with the goal to improve the effectiveness and ameliorate the complications of its demanding therapy.
Collapse
Affiliation(s)
- Florian G Scurt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany.
| | - K Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Ben Hammoud
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - S Brandt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - A Bernhardt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - C Gross
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Peter R Mertens
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | | |
Collapse
|
32
|
Huertas J, Lee HT. Multi‑faceted roles of cathepsins in ischemia reperfusion injury (Review). Mol Med Rep 2022; 26:368. [PMID: 36300202 PMCID: PMC9644425 DOI: 10.3892/mmr.2022.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Cathepsins are one of the most abundant proteases within the lysosomes with diverse physiological effects ranging from immune responses, cell death and intracellular protein degradation. Cathepsins are involved in extracellular and systemic functions such as systemic inflammation and extracellular matrix degradation. Ischemia reperfusion (IR) injury is responsible for numerous diseases including myocardial infarction, acute kidney injury, stroke and acute graft failure after transplant surgery. Inflammation plays a major role in the reperfusion phase of IR injury and previous research has shown that cathepsins are key mediators of the inflammation cascade as well as apoptosis. Taken together, cathepsins modulation could provide potential therapeutic approaches to attenuate IR injury. The present review summarized the current understanding of various cathepsin subtypes, their major physiologic functions, their roles in multi‑organ IR injury and detailed selective cathepsin inhibitors with therapeutic potential.
Collapse
Affiliation(s)
- Jaime Huertas
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| | - H. Thomas Lee
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| |
Collapse
|
33
|
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: 22] [Impact Index Per Article: 7.3] [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
|
34
|
Aymonnier K, Amsler J, Lamprecht P, Salama A, Witko‐Sarsat V. The neutrophil: A key resourceful agent in immune‐mediated vasculitis. Immunol Rev 2022; 314:326-356. [PMID: 36408947 DOI: 10.1111/imr.13170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The term "vasculitis" refers to a group of rare immune-mediated diseases characterized by the dysregulated immune system attacking blood vessels located in any organ of the body, including the skin, lungs, and kidneys. Vasculitides are classified according to the size of the vessel that is affected. Although this observation is not specific to small-, medium-, or large-vessel vasculitides, patients show a high circulating neutrophil-to-lymphocyte ratio, suggesting the direct or indirect involvement of neutrophils in these diseases. As first responders to infection or inflammation, neutrophils release cytotoxic mediators, including reactive oxygen species, proteases, and neutrophil extracellular traps. If not controlled, this dangerous arsenal can injure the vascular system, which acts as the main transport route for neutrophils, thereby amplifying the initial inflammatory stimulus and the recruitment of immune cells. This review highlights the ability of neutrophils to "set the tone" for immune cells and other cells in the vessel wall. Considering both their long-established and newly described roles, we extend their functions far beyond their direct host-damaging potential. We also review the roles of neutrophils in various types of primary vasculitis, including immune complex vasculitis, anti-neutrophil cytoplasmic antibody-associated vasculitis, polyarteritis nodosa, Kawasaki disease, giant cell arteritis, Takayasu arteritis, and Behçet's disease.
Collapse
Affiliation(s)
- Karen Aymonnier
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Jennifer Amsler
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology University of Lübeck Lübeck Germany
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital University College London London UK
| | | |
Collapse
|
35
|
Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim Biophys Acta Rev Cancer 2022; 1877:188762. [PMID: 35853517 DOI: 10.1016/j.bbcan.2022.188762] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/10/2022] [Accepted: 07/14/2022] [Indexed: 02/08/2023]
Abstract
Neutrophils are the frontline cells in response to microbial infections and are involved in a range of inflammatory disorders in the body. In recent years, neutrophils have gained considerable attention in their involvement of complex roles in tumor development and progression. Tumor-associated neutrophils (TANs) that accumulate in local region could be triggered by external stimuli from tumor microenvironment (TME) and switch between anti- and pro-tumor phenotypes. The anti-tumor neutrophils kill tumor cells through direct cytotoxic effects as well as indirect effects by activating adaptive immune responses. In contrast, the pro-tumor phenotype of neutrophils might be associated with cell proliferation, angiogenesis, and immunosuppression in TME. More recently, neutrophils have been proposed as a potential target in cancer therapy for their ability to diminish the pro-tumor pathways, such as by immune checkpoint blockade. This review discusses the complex roles of neutrophils in TME and highlights the strategies in neutrophil targeting in cancer treatment with a particular focus on the progresses of ongoing clinical trials involving neutrophil-targeted therapies.
Collapse
|
36
|
Chen X, Yan Y, Du J, Shen X, He C, Pan H, Zhu J, Liu X. Non-peptidyl non-covalent cathepsin C inhibitoEEr bearing a unique thiophene-substituted pyridine: Design, structure-activity relationship and anti-inflammatory activity in vivo. Eur J Med Chem 2022; 236:114368. [DOI: 10.1016/j.ejmech.2022.114368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/20/2022] [Accepted: 04/05/2022] [Indexed: 11/04/2022]
|
37
|
Tang W, Zhong Y, Wei Y, Deng Z, Mao J, Liu J, Valencak TG, Liu J, Xu H, Wang H. Ileum tissue single-cell mRNA sequencing elucidates the cellular architecture of pathophysiological changes associated with weaning in piglets. BMC Biol 2022; 20:123. [PMID: 35637473 PMCID: PMC9153155 DOI: 10.1186/s12915-022-01321-3] [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: 03/26/2022] [Accepted: 05/06/2022] [Indexed: 11/19/2022] Open
Abstract
Background In mammals, transitioning from sole milk uptake to the intake of solid feed results in dramatic developmental changes in intestinal function and immunological status. In fact, weaning stress is often accompanied by intestinal inflammatory processes. To develop effective intervention strategies, it is necessary to characterize the developmental pattern and immune response that occurs on weaning, as we have done in this study for piglets. Results To comprehensively delineate cell heterogeneity in ileum tissues and the underlying mechanisms in weaning-induced intestinal inflammation of piglets, we have analyzed the transcriptomes of 42,149 cells from ileum mucosa of normally suckling and post-weaned piglets. There were 31 cell subtypes including epithelial, stromal, and immune cells. A bifurcating trajectory was inferred to separate secretory and absorptive lineages. Integrated cross-species datasets showed well-conserved cellular architectures and transcription signatures between human and pig. Comparative analyses of cellular components, cell–cell communications, and molecular states revealed that T cell subpopulations were significantly altered in weaned piglets. We found that T helper (Th) 17 functional plasticity across changes in the cytokine milieu and the enrichment of granzyme B (GZMB)-expressing cytotoxic T cells potentially exacerbate mucosal inflammation via mitochondrial dysfunction in epithelial cells. Conclusions Our work has elucidated the single-cell molecular characteristics of the piglet ileum before and after weaning. We have provided an atlas that portrays the landscape of the intestinal pathophysiological inflammatory process associated with weaning, finding a level of conservation between human and pig that support the use of piglets as a model for human infants. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01321-3.
Collapse
Affiliation(s)
- Wenjie Tang
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Yifan Zhong
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Yusen Wei
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Zhaoxi Deng
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Jiangdi Mao
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Jingliang Liu
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Teresa G Valencak
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Jianxin Liu
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310000, Zhejiang, China
| | - Haifeng Wang
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310000, China.
| |
Collapse
|
38
|
Kain R, Nackenhorst MC. A View on Cathepsin C as a Target for Therapy in AAV. J Am Soc Nephrol 2022; 33:875-878. [PMID: 35396261 PMCID: PMC9063890 DOI: 10.1681/asn.2022030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
39
|
Fassan M, Collesei A, Angerilli V, Sbaraglia M, Fortarezza F, Pezzuto F, De Gaspari M, Businello G, Moni M, Rizzo S, Traverso G, Colosso V, Taschin E, Lunardi F, Valls AF, Schiavi F, Basso C, Calabrese F, Dei Tos AP. Multi-Design Differential Expression Profiling of COVID-19 Lung Autopsy Specimens Reveals Significantly Deregulated Inflammatory Pathways and SFTPC Impaired Transcription. Cells 2022; 11:cells11061011. [PMID: 35326463 PMCID: PMC8947344 DOI: 10.3390/cells11061011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
The transcriptomic profiling of lung damage associated with SARS-CoV-2 infection may lead to the development of effective therapies to prevent COVID-19-related deaths. We selected a series of 21 autoptic lung samples, 14 of which had positive nasopharyngeal swabs for SARS-CoV-2 and a clinical diagnosis of COVID-19-related death; their pulmonary viral load was quantified with a specific probe for SARS-CoV-2. The remaining seven cases had no documented respiratory disease and were used as controls. RNA from formalin-fixed paraffin-embedded (FFPE) tissue samples was extracted to perform gene expression profiling by means of targeted (Nanostring) and comprehensive RNA-Seq. Two differential expression designs were carried out leading to relevant results in terms of deregulation. SARS-CoV-2 positive specimens presented a significant overexpression in genes of the type I interferon signaling pathway (IFIT1, OAS1, ISG15 and RSAD2), complement activation (C2 and CFB), macrophage polarization (PKM, SIGLEC1, CD163 and MS4A4A) and Cathepsin C (CTSC). CD163, Siglec-1 and Cathepsin C overexpression was validated by immunohistochemistry. SFTPC, the encoding gene for pulmonary-associated surfactant protein C, emerged as a key identifier of COVID-19 patients with high viral load. This study successfully recognized SARS-CoV-2 specific immune signatures in lung samples and highlighted new potential therapeutic targets. A better understanding of the immunopathogenic mechanisms of SARS-CoV-2 induced lung damage is required to develop effective individualized pharmacological strategies.
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
- Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
- Correspondence: ; Tel.: +39-0498217931
| | - Antonio Collesei
- Familial Cancer Clinics, Veneto Institute of Oncology, IOV-IRCCS, 35127 Padua, Italy; (A.C.); (V.C.); (E.T.); (F.S.)
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy
| | - Valentina Angerilli
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
| | - Marta Sbaraglia
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
| | - Francesco Fortarezza
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
| | - Federica Pezzuto
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
| | - Monica De Gaspari
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
| | - Gianluca Businello
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
| | - Margherita Moni
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
- Cardiovascular Pathology Unit, Padua University Hospital, 35121 Padua, Italy
| | - Giulia Traverso
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
| | - Veronica Colosso
- Familial Cancer Clinics, Veneto Institute of Oncology, IOV-IRCCS, 35127 Padua, Italy; (A.C.); (V.C.); (E.T.); (F.S.)
| | - Elisa Taschin
- Familial Cancer Clinics, Veneto Institute of Oncology, IOV-IRCCS, 35127 Padua, Italy; (A.C.); (V.C.); (E.T.); (F.S.)
| | - Francesca Lunardi
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
| | - Aida Freire Valls
- NanoString Technologies, Inc., 530 Fairview Avenue N, Seattle, WA 98109, USA;
| | - Francesca Schiavi
- Familial Cancer Clinics, Veneto Institute of Oncology, IOV-IRCCS, 35127 Padua, Italy; (A.C.); (V.C.); (E.T.); (F.S.)
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
- Cardiovascular Pathology Unit, Padua University Hospital, 35121 Padua, Italy
| | - Fiorella Calabrese
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.D.G.); (S.R.); (C.B.)
| | - Angelo Paolo Dei Tos
- Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (V.A.); (M.S.); (G.B.); (M.M.); (G.T.); (F.L.); (A.P.D.T.)
- Surgical Pathology Unit, Padua University Hospital, 35121 Padua, Italy; (F.F.); (F.P.); (F.C.)
| |
Collapse
|
40
|
Jerke U, Eulenberg-Gustavus C, Rousselle A, Nicklin P, Kreideweiss S, Grundl MA, Eickholz P, Nickles K, Schreiber A, Korkmaz B, Kettritz R. Targeting Cathepsin C in PR3-ANCA Vasculitis. J Am Soc Nephrol 2022; 33:936-947. [PMID: 35292437 PMCID: PMC9063889 DOI: 10.1681/asn.2021081112] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/02/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The ANCA autoantigens proteinase 3 (PR3) and myeloperoxidase (MPO) are exclusively expressed by neutrophils and monocytes. ANCA-mediated activation of these cells is the key driver of the vascular injury process in ANCA-associated vasculitis (AAV), and neutrophil serine proteases (NSPs) are disease mediators. Cathepsin C (CatC) from zymogens activates the proteolytic function of NSPs, including PR3. Lack of NSP zymogen activation results in neutrophils with strongly reduced NSP proteins. METHODS To explore AAV-relevant consequences of blocking NSP zymogen activation by CatC, we used myeloid cells from patients with Papillon-Lefèvre syndrome, a genetic deficiency of CatC, to assess NSPs and NSP-mediated endothelial cell injury. We also examined pharmacologic CatC inhibition in neutrophil-differentiated human hematopoietic stem cells, primary human umbilical vein cells, and primary glomerular microvascular endothelial cells. RESULTS Patients with Papillon-Lefèvre syndrome showed strongly reduced NSPs in neutrophils and monocytes. Neutrophils from these patients produced a negative PR3-ANCA test, presented less PR3 on the surface of viable and apoptotic cells, and caused significantly less damage in human umbilical vein cells. These findings were recapitulated in human stem cells, in which a highly specific CatC inhibitor, but not prednisolone, reduced NSPs without affecting neutrophil differentiation, reduced membrane PR3, and diminished neutrophil activation upon PR3-ANCA but not MPO-ANCA stimulation. Compared with healthy controls, neutrophils from patients with Papillon-Lefèvre syndrome transferred less proteolytically active NSPs to glomerular microvascular endothelial cells, the cell type targeted in ANCA-induced necrotizing crescentic glomerulonephritis. Finally, both genetic CatC deficiency and pharmacologic inhibition, but not prednisolone, reduced neutrophil-induced glomerular microvascular endothelial cell damage. CONCLUSIONS These findings may offer encouragement for clinical studies of adjunctive CatC inhibitor in patients with PR3-AAV.
Collapse
Affiliation(s)
- Uwe Jerke
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anthony Rousselle
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Paul Nicklin
- Boehringer Ingelheim Pharma GmbH & Co., KG, Biberach, Germany
| | | | - Marc A Grundl
- Boehringer Ingelheim Pharma GmbH & Co., KG, Biberach, Germany
| | - Peter Eickholz
- Peridontology, Johann Wolfgang Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Katrin Nickles
- Peridontology, Johann Wolfgang Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ralph Kettritz
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany .,Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
41
|
Varda N, Novinec M. Evolutionary Analysis of Dipeptidyl Peptidase I. Int J Mol Sci 2022; 23:ijms23031852. [PMID: 35163774 PMCID: PMC8836896 DOI: 10.3390/ijms23031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/10/2022] Open
Abstract
Human dipeptidyl peptidase I (DPPI) belongs to the family of papain-like cysteine peptidases. Its distinctive features are the unique exclusion domain which enables the eponymous activity and homotetramerization of DPPI, and its dependence on chloride ions for enzymatic activity. The oligomeric state of DPPI is unique in this family of predominantly monomeric peptidases. However, a distant DPPI ortholog from Plasmodium falciparum has been shown to be monomeric, indicating that the oligomeric state of DPPI varies between lineages. The aim of this work was to study the evolution of DPPI, with particular attention to the structural features that determine its characteristic enzymatic activity and preferences, and to reconstruct the evolution of its oligomerization. We analyzed fifty-seven selected sequences of DPPI and confirmed its presence in three lineages, namely, Amorphea (including animals and Amoebozoa), Alveolates and the metamonad Giardia. The amino acid residues that bind the chloride ion are highly conserved in all species, indicating that the dependence on chloride ions for activity is an evolutionarily conserved feature of DPPI. The number of N-glycosylation sites is significantly increased in animals, particularly vertebrates. Analysis of homology models and subunit contacts suggests that oligomerization is likely restricted to DPPIs in the Amorphea group.
Collapse
|
42
|
Bahuguna A, Pal Khaket T, Bajpai VK, Shukla S, Park I, Na M, Suk Huh Y, Han YK, Chul Kang S, Kim M. N-acetyldopamine dimers from Oxya chinensis sinuosa attenuates lipopolysaccharides induced inflammation and inhibits cathepsin C activity. Comput Struct Biotechnol J 2022; 20:1177-1188. [PMID: 35317232 PMCID: PMC8908036 DOI: 10.1016/j.csbj.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 11/03/2022] Open
|
43
|
Antonelou M, Evans RDR, Henderson SR, Salama AD. Neutrophils are key mediators in crescentic glomerulonephritis and targets for new therapeutic approaches. Nephrol Dial Transplant 2022; 37:230-238. [PMID: 33057680 DOI: 10.1093/ndt/gfaa206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Indexed: 12/26/2022] Open
Abstract
Crescentic glomerulonephritis (CGN) results from a diverse set of diseases associated with immune dysregulation and the breakdown of self-tolerance to a wide range of autoantigens, some known and some that remain unknown. Experimental data demonstrate that neutrophils have an important role in the pathogenesis of CGN. Upon activation, neutrophils generate reactive oxygen species, release serine proteases and form neutrophil extracellular traps (NETs), all of which can induce direct tissue damage. In addition, serine proteases such as myeloperoxidase and proteinase 3, presented on NETs, can be processed and recognized as autoantigens, leading to the generation and maintenance of autoimmune responses in susceptible individuals. The basis of the specificity of autoimmune responses in different patients to NET proteins is unclear, but relates at least in part to differences in human leucocyte antigen expression. Conditions associated with CGN are often characterized by aberrant neutrophil activation and NETosis and, in some, impaired NET degradation. Targeting neutrophil degranulation and NETosis is now possible using a variety of novel compounds and may provide a promising therapeutic alternative to glucocorticoid use, which has been a mainstay of management in CGN for decades and is associated with significant adverse effects. In this review, we discuss the evidence supporting the role of neutrophils in the development of CGN and the pathways identified in neutrophil degranulation and NETosis that may translate to novel therapeutic applications.
Collapse
Affiliation(s)
- Marilina Antonelou
- University College London, Department of Renal Medicine, Royal Free Hospital, London, UK
| | - Rhys D R Evans
- University College London, Department of Renal Medicine, Royal Free Hospital, London, UK
| | - Scott R Henderson
- University College London, Department of Renal Medicine, Royal Free Hospital, London, UK
| | - Alan D Salama
- University College London, Department of Renal Medicine, Royal Free Hospital, London, UK
| |
Collapse
|
44
|
Lu F, Gong H, Lei H, Li J. Downregulation of cathepsin C alleviates endothelial cell dysfunction by suppressing p38 MAPK/NF-κB pathway in preeclampsia. Bioengineered 2022; 13:3019-3028. [PMID: 35037834 PMCID: PMC8974117 DOI: 10.1080/21655979.2021.2023994] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Endothelial cell dysfunction is an essential pathophysiological feature of preeclampsia (PE). It has been reported that cathepsin C is upregulated in the maternal vascular endothelium of PE patients. The excessive activation of p38 MAPK leads to various diseases, including PE. NF-κB pathway can promote uteroplacental dysfunction, endothelial stress and development of PE. Moreover, it has been verified that cathepsin C can activate p38 MAPK/NF-κB pathway. In the present work, hypoxia/reoxygenation (H/R) injury model of HUVECs was established to discuss the biological functions of cathepsin C in endothelial cell dysfunction and to elucidate the underlying molecular mechanism. The correlation between cathepsin C and p38 MAPK/NF-κB pathway in H/R-stimulated HUVECs as well as the effects of cathepsin C and p38 MAPK/NF-κB pathway on viability, apoptosis, invasion, in vitro angiogenesis of HUVECs and oxidative stress were assessed. The results revealed that H/R injury elevated cathepsin C expression and activated p38 MAPK/NF-κB pathway in HUVECs and cathepsin C knockdown inhibited the activity of p38 MAPK/NF-κB pathway in H/R-stimulated HUVECs. Downregulation of cathepsin C improved viability, inhibited apoptosis and enhanced invasion of H/R-stimulated HUVECs. In addition, downregulation of cathepsin C alleviated oxidative stress and induced stronger HUVEC angiogenesis in vitro. Furthermore, the protective effects of cathepsin C knockdown against endothelial cell dysfunction were reversed by p38 MAPK activator anisomycin. In other words, downregulation of cathepsin C could improve HUVEC viability and enhance anti-apoptotic capacity, anti-oxidative capability, invasive ability, as well as angiogenic potential of H/R-stimulated HUVECs by repressing p38 MAPK/NF-κB pathway.
Collapse
Affiliation(s)
- Fan Lu
- Department of Obstetrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Han Gong
- Department of Obstetrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Houkang Lei
- Department of Obstetrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Juan Li
- Department of Obstetrics, The Third People's Hospital of Yunnan Province, Kunming, Yunnan, Province, China
| |
Collapse
|
45
|
Shen XB, Chen X, Zhang ZY, Wu FF, Liu XH. Cathepsin C inhibitors as anti-inflammatory drug discovery: Challenges and opportunities. Eur J Med Chem 2021; 225:113818. [PMID: 34492551 DOI: 10.1016/j.ejmech.2021.113818] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022]
Abstract
Cathepsin C, an important lysosomal cysteine protease, mediates the maturation process of neutrophil serine proteases, and participates in the inflammation and immune regulation process associated with polymorphonuclear neutrophils. Therefore, cathepsin C is considered to be an attractive target for treating inflammatory diseases. With INS1007 (trade name: brensocatib) being granted a breakthrough drug designation by FDA for the treatment of Adult Non-cystic Fibrosis Bronchiectasis and Coronavirus Disease 2019, the development of cathepsin C inhibitor will attract attentions from medicinal chemists in the future soon. Here, we summarized the research results of cathepsin C as a therapeutic target, focusing on the development of cathepsin C inhibitor, and provided guidance and reference opinions for the upcoming development boom of cathepsin C inhibitor.
Collapse
Affiliation(s)
- Xiao Bao Shen
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, 236037, PR China
| | - Xing Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zhao Yan Zhang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Fu Fang Wu
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, 236037, PR China.
| | - Xin Hua Liu
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, 236037, PR China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
46
|
Korkmaz B, Lamort AS, Domain R, Beauvillain C, Gieldon A, Yildirim AÖ, Stathopoulos GT, Rhimi M, Jenne DE, Kettritz R. Cathepsin C inhibition as a potential treatment strategy in cancer. Biochem Pharmacol 2021; 194:114803. [PMID: 34678221 DOI: 10.1016/j.bcp.2021.114803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/08/2023]
Abstract
Epidemiological studies established an association between chronic inflammation and higher risk of cancer. Inhibition of proteolytic enzymes represents a potential treatment strategy for cancer and prevention of cancer metastasis. Cathepsin C (CatC) is a highly conserved lysosomal cysteine dipeptidyl aminopeptidase required for the activation of pro-inflammatory neutrophil serine proteases (NSPs, elastase, proteinase 3, cathepsin G and NSP-4). NSPs are locally released by activated neutrophils in response to pathogens and non-infectious danger signals. Activated neutrophils also release neutrophil extracellular traps (NETs) that are decorated with several neutrophil proteins, including NSPs. NSPs are not only NETs constituents but also play a role in NET formation and release. Although immune cells harbor large amounts of CatC, additional cell sources for this protease exists. Upregulation of CatC expression was observed in different tissues during carcinogenesis and correlated with metastasis and poor patient survival. Recent mechanistic studies indicated an important interaction of tumor-associated CatC, NSPs, and NETs in cancer development and metastasis and suggested CatC as a therapeutic target in a several cancer types. Cancer cell-derived CatC promotes neutrophil recruitment in the inflammatory tumor microenvironment. Because the clinical consequences of genetic CatC deficiency in humans resulting in the elimination of NSPs are mild, small molecule inhibitors of CatC are assumed as safe drugs to reduce the NSP burden. Brensocatib, a nitrile CatC inhibitor is currently tested in a phase 3 clinical trial as a novel anti-inflammatory therapy for patients with bronchiectasis. However, recently developed CatC inhibitors possibly have protective effects beyond inflammation. In this review, we describe the pathophysiological function of CatC and discuss molecular mechanisms substantiating pharmacological CatC inhibition as a potential strategy for cancer treatment.
Collapse
Affiliation(s)
- Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032 Tours, France.
| | - Anne-Sophie Lamort
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Roxane Domain
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032 Tours, France
| | - Céline Beauvillain
- University of Angers, University of Nantes, Angers University Hospital, INSERM UMR-1232, CRCINA, Innate Immunity and Immunotherapy, SFR ICAT, 49000 Angers, France
| | - Artur Gieldon
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Dieter E Jenne
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2); Max Planck Institute of Neurobiology, 82152 Planegg-Martinsried, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité und Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany; Nephrology and Intensive Care Medicine, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
47
|
Tubular lysosomes harbor active ion gradients and poise macrophages for phagocytosis. Proc Natl Acad Sci U S A 2021; 118:2113174118. [PMID: 34607961 PMCID: PMC8522270 DOI: 10.1073/pnas.2113174118] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Lysosomes are organelles that also act as cell-signaling hubs. They regulate functions ranging from antigen presentation to autophagy. Spherical lysosomes can spontaneously elongate into tubules in starving or inflamed immune cells. We describe a DNA-based reagent, denoted Tudor, that tubulates lysosomes in macrophages without triggering either an immune response or autophagy. Chemical imaging revealed that tubular lysosomes differ from vesicular ones in terms of their pH, calcium, and proteolytic activity. Tudor revealed a role for tubular lysosomes in that they enhance MMP9 secretion and phagocytosis in resting macrophages. The ability to tubulate lysosomes in resting immune cells without starving or inflaming them may help reveal new insights into how tubular lysosomes function. Lysosomes adopt dynamic, tubular states that regulate antigen presentation, phagosome resolution, and autophagy. Tubular lysosomes are studied either by inducing autophagy or by activating immune cells, both of which lead to cell states where lysosomal gene expression differs from the resting state. Therefore, it has been challenging to pinpoint the biochemical properties lysosomes acquire upon tubulation that could drive their functionality. Here we describe a DNA-based assembly that tubulates lysosomes in macrophages without activating them. Proteolytic activity maps at single-lysosome resolution revealed that tubular lysosomes were less degradative and showed proximal to distal luminal pH and Ca2+ gradients. Such gradients had been predicted but never previously observed. We identify a role for tubular lysosomes in promoting phagocytosis and activating MMP9. The ability to tubulate lysosomes without starving or activating immune cells may help reveal new roles for tubular lysosomes.
Collapse
|
48
|
D’Souza SS, Zhang Y, Bailey JT, Fung ITH, Kuentzel ML, Chittur SV, Yang Q. Type I Interferon signaling controls the accumulation and transcriptomes of monocytes in the aged lung. Aging Cell 2021; 20:e13470. [PMID: 34547174 PMCID: PMC8520712 DOI: 10.1111/acel.13470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/18/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022] Open
Abstract
Aging is paradoxically associated with a deteriorated immune defense (immunosenescence) and increased basal levels of tissue inflammation (inflammaging). The lung is particularly sensitive to the effects of aging. The immune cell mechanisms underlying physiological lung aging remain poorly understood. Here we reveal that aging leads to increased interferon signaling and elevated concentrations of chemokines in the lung, which is associated with infiltration of monocytes into the lung parenchyma. scRNA‐seq identified a novel Type‐1 interferon signaling dependent monocyte subset (MO‐ifn) that upregulated IFNAR1 expression and exhibited greater transcriptomal changes with aging than the other monocytes. Blockade of type‐1 interferon signaling by treatment with anti‐IFNAR1 neutralizing antibodies rapidly ablated MO‐ifn cells. Treatment with anti‐IFNAR1 antibodies also reduced airway chemokine concentrations and repressed the accumulation of the overall monocyte population in the parenchyma of the aged lung. Together, our work suggests that physiological aging is associated with increased basal level of airway monocyte infiltration and inflammation in part due to elevated type‐1 interferon signaling.
Collapse
Affiliation(s)
- Shanti S. D’Souza
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Yuanyue Zhang
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Jacob T. Bailey
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Ivan T. H. Fung
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Marcy L. Kuentzel
- Center for Functional Genomics University at Albany‐SUNY Rensselaer NY USA
| | - Sridar V. Chittur
- Center for Functional Genomics University at Albany‐SUNY Rensselaer NY USA
| | - Qi Yang
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| |
Collapse
|
49
|
Donadieu J, Frenz S, Merz L, Sicre De Fontbrune F, Rotulo GA, Beaupain B, Biosse-Duplan M, Audrain M, Croisille L, Ancliff P, Klein C, Bellanné-Chantelot C. Chronic neutropenia: how best to assess severity and approach management? Expert Rev Hematol 2021; 14:945-960. [PMID: 34486458 DOI: 10.1080/17474086.2021.1976634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Neutropenia is a relatively common finding in medical practice and the medical approach requires a gradual and pertinent diagnostic procedure as well as adapted management. AREAS COVERED The area of chronic neutropenia remains fragmented between diverse diseases or situations. Here physicians involved in different aspects of chronic neutropenia gather both the data from medical literature till the end of May 2021 and their experience to offer a global approach for the diagnosis of chronic neutropenia as well as their medical care. EXPERT OPINION In most cases, the neutropenia is transient, frequently related to a viral infection, and not harmful. However, neutropenia can be chronic (i.e. >3 months) and related to a number of etiologies, some clinically benign, such as so-called 'ethnic' neutropenia. Autoimmune neutropenia is the common form in young children, whereas idiopathic/immune neutropenia is a frequent etiology in young females. Inherited neutropenia (or congenital neutropenia) is exceptional, with approximately 30 new cases per 106 births and 30 known subtypes. Such patients have a high risk of invasive bacterial infections, and oral infections. Supportive therapy, which is primarily based on daily administration of an antibiotic prophylaxis and/or treatment with granulocyte-colony stimulating factor (G-CSF), contributes to avoiding recurrent infections.
Collapse
Affiliation(s)
- Jean Donadieu
- Centre De Référence Des Neutropénies Chroniques, Registre National Des Neutropénies Congénitales, Service d'Hémato-oncologie Pédiatrique, Hôpital Armand Trousseau Aphp, Paris, France
| | - Stephanie Frenz
- Dr. Von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lauren Merz
- Brigham and Women's Hospital, Department of Internal Medicine, Boston, MA, USA
| | | | - Gioacchino Andrea Rotulo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (Dinogmi), University of Genoa, Italy
| | - Blandine Beaupain
- Centre De Référence Des Neutropénies Chroniques, Registre National Des Neutropénies Congénitales, Service d'Hémato-oncologie Pédiatrique, Hôpital Armand Trousseau Aphp, Paris, France
| | | | - Marie Audrain
- Service d'Immunologie Laboratoire De Biologie Chu De Nantes 9 Quai Moncousu
| | | | - Phil Ancliff
- Pediatric Hematology, Great Ormond Street Hospital London, UK
| | - Christoph Klein
- Dr. Von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | |
Collapse
|
50
|
Banerjee A, Velagaleti R, Patil S, Pawar M, Yadav P, Kadam P, Qadri MM, Chakraborti S, Saini JS, Behera DB, Karanjai K, Iyer PS, Gharat LA, Das S. Development of potent and selective Cathepsin C inhibitors free of aortic binding liability by application of a conformational restriction strategy. Bioorg Med Chem Lett 2021; 47:128202. [PMID: 34139325 DOI: 10.1016/j.bmcl.2021.128202] [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: 02/25/2021] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
Cathepsin C plays a key role in the activation of several degradative enzymes linked to tissue destruction in chronic inflammatory and autoimmune diseases. Therefore, Cathepsin C inhibitors could potentially be effective therapeutics for the treatment of diseases such as chronic obstructive pulmonary disease (COPD) or acute respiratory distress syndrome (ARDS). In our efforts towards the development of a novel series of Cathepsin C inhibitors, we started working around AZD5248 (1), an α-amino acid based scaffold having potential liability of aortic binding. A novel series of amidoacetonitrile based Cathepsin C inhibitors were developed by the application of a conformational restriction strategy on 1. In particular, this work led to the development of a potent and selective Cathepsin C inhibitor 3p, free of aortic binding liability.
Collapse
Affiliation(s)
- Abhisek Banerjee
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Ranganadh Velagaleti
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Sandip Patil
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Mahesh Pawar
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Pravin Yadav
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Pradip Kadam
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Mohammad Mohsin Qadri
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Samitabh Chakraborti
- Pharmacology Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Jagmohan S Saini
- Computational Chemistry, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Dayanidhi B Behera
- Drug Metabolism and Pharmacokinetics, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Keya Karanjai
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Pravin S Iyer
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India.; Pharmacology Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India.; Computational Chemistry, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India.; Drug Metabolism and Pharmacokinetics, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Laxmikant A Gharat
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India
| | - Sanjib Das
- Medicinal Chemistry Division, Glenmark Research Centre, A-607, TTC Industrial Area, MIDC Mahape, Navi Mumbai 400 709, India..
| |
Collapse
|