1
|
Ferková S, Lepage M, Désilets A, Assouvie K, Lemieux G, Brochu I, Froehlich U, Gravel-Trudeau A, Vastra J, Jean F, Sarret P, Leduc R, Boudreault PL. Optimizing the pharmacokinetics and selectivity of TMPRSS2 inhibitors. Eur J Med Chem 2025; 294:117579. [PMID: 40382841 DOI: 10.1016/j.ejmech.2025.117579] [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: 02/18/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 05/20/2025]
Abstract
Since 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has undergone significant genomic mutations, contributing to resistance against existing 2019 coronavirus disease (COVID-19) treatments. In a previous study, we identified N-0385, a potent host-directed inhibitor of transmembrane serine protease 2 (TMPRSS2), which has therapeutic efficacy towards SARS-CoV-2 infection. However, in further evaluation of its preclinical druggability, N-0385 displayed unfavorable pharmacokinetic properties, including high bioavailability (99 %) following intranasal (IN) administration. This can lead to substantial systemic exposure and potential adverse effects due to off-target interactions. Here, we designed a library of peptidomimetic compounds with P3 site modifications on an optimized scaffold. We sought to maintain sub-nanomolar potency against TMPRSS2 (Kis < 2 nM), reduce pseudovirus infection, while addressing the lack of selectivity and excessive lung uptake. Notably, inhibitor 9, which contains Asp at the P3 position, achieved a two-fold increase in TMPRSS2 inhibitory potency (Ki = 0.13 ± 0.03 nM), a >700-fold selectivity over Factor Xa (FXa), and showed superior selectivity against other proteases (matriptase, transmembrane serine protease 6 (TMPRSS6), thrombin, furin, and tPA). Despite concerns about the role of FXa in the coagulation cascade, compound 9 had no impact on coagulation or thrombolysis 2 h after in vitro treatment. In the air-liquid interface (ALI) model of the lung epithelium, compound 9 displayed a 1.5-fold decrease in permeability compared to N-0385 and demonstrated sustained stability in lungs (11 h) and plasma (13 h). Taken together, our data demonstrate that continued optimization of this type of inhibitors will lead to improved therapeutics for the treatment of SARS-CoV-2 infection by IN administration.
Collapse
Affiliation(s)
- Sára Ferková
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthieu Lepage
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kevin Assouvie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Isabelle Brochu
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Ulrike Froehlich
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Alice Gravel-Trudeau
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jules Vastra
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Jean
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
2
|
Higashi T, Saito AC, Chiba H. Damage control of epithelial barrier function in dynamic environments. Eur J Cell Biol 2024; 103:151410. [PMID: 38579602 DOI: 10.1016/j.ejcb.2024.151410] [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: 12/30/2023] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024] Open
Abstract
Epithelial tissues cover the surfaces and lumens of the internal organs of multicellular animals and crucially contribute to internal environment homeostasis by delineating distinct compartments within the body. This vital role is known as epithelial barrier function. Epithelial cells are arranged like cobblestones and intricately bind together to form an epithelial sheet that upholds this barrier function. Central to the restriction of solute and fluid diffusion through intercellular spaces are occluding junctions, tight junctions in vertebrates and septate junctions in invertebrates. As part of epithelial tissues, cells undergo constant renewal, with older cells being replaced by new ones. Simultaneously, the epithelial tissue undergoes relative rearrangement, elongating, and shifting directionally as a whole. The movement or shape changes within the epithelial sheet necessitate significant deformation and reconnection of occluding junctions. Recent advancements have shed light on the intricate mechanisms through which epithelial cells sustain their barrier function in dynamic environments. This review aims to introduce these noteworthy findings and discuss some of the questions that remain unanswered.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Akira C Saito
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
3
|
Naidu AS, Wang CK, Rao P, Mancini F, Clemens RA, Wirakartakusumah A, Chiu HF, Yen CH, Porretta S, Mathai I, Naidu SAG. Precision nutrition to reset virus-induced human metabolic reprogramming and dysregulation (HMRD) in long-COVID. NPJ Sci Food 2024; 8:19. [PMID: 38555403 PMCID: PMC10981760 DOI: 10.1038/s41538-024-00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
SARS-CoV-2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus-host protein-protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia ('cytokine storm'), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25-70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new 'onset' clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Collapse
Affiliation(s)
- A Satyanarayan Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA.
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA.
| | - Chin-Kun Wang
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- School of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung, 40201, Taiwan
| | - Pingfan Rao
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- College of Food and Bioengineering, Fujian Polytechnic Normal University, No.1, Campus New Village, Longjiang Street, Fuqing City, Fujian, China
| | - Fabrizio Mancini
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President-Emeritus, Parker University, 2540 Walnut Hill Lane, Dallas, TX, 75229, USA
| | - Roger A Clemens
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- University of Southern California, Alfred E. Mann School of Pharmacy/D. K. Kim International Center for Regulatory & Quality Sciences, 1540 Alcazar St., CHP 140, Los Angeles, CA, 90089, USA
| | - Aman Wirakartakusumah
- International Union of Food Science and Technology (IUFoST), Guelph, ON, Canada
- IPMI International Business School Jakarta; South East Asian Food and Agriculture Science and Technology, IPB University, Bogor, Indonesia
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital, Ministry of Health & Well-being, Taichung, Taiwan
| | - Chi-Hua Yen
- Department of Family and Community Medicine, Chung Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Sebastiano Porretta
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President, Italian Association of Food Technology (AITA), Milan, Italy
- Experimental Station for the Food Preserving Industry, Department of Consumer Science, Viale Tanara 31/a, I-43121, Parma, Italy
| | - Issac Mathai
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- Soukya International Holistic Health Center, Whitefield, Bengaluru, India
| | - Sreus A G Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA
| |
Collapse
|
4
|
Nie X, Gao L, Zheng M, Wang S, Wang C, Li X, Liu O, Gou R, Liu J, Lin B. ST14 interacts with TMEFF1 and is a predictor of poor prognosis in ovarian cancer. BMC Cancer 2024; 24:330. [PMID: 38468232 DOI: 10.1186/s12885-024-11958-8] [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: 06/11/2022] [Accepted: 02/05/2024] [Indexed: 03/13/2024] Open
Abstract
TMEFF1 is a new protein involved in the physiological functions of the central nervous system, and we previously reported TMEFF1 can promote ovarian cancer. ST14 was determined to be involved in the processes of epidermal differentiation, epithelial cell integrity, and vascular endothelial cell migration, etc. The relationship between ST14 and TMEFF1 in the ovary remains unknown. In this study, we detected the expression of ST14 and TMEFF1 in 130 different ovarian cancer tissues through immunohistochemistry. We determined ST14 and TMEFF1 were highly expressed in ovarian cancer, indicating a higher degree of tumor malignancy and a worse prognosis. Tissues significantly expressing ST14 also highly expressed TMEFF1, and the expression of the two proteins was positively correlated. Consistently, immunofluorescence double staining demonstrated the co-localization of ST14 and TMEFF1 in the same region, and immunoprecipitation confirmed the interaction between ST14 and TMEFF1. TMEFF1 expression was also reduced after knocking down ST14 through Western blot. MTT, wound healing and Transwell assays results determined that knockdown of ST14 inhibited proliferation, migration and invasion of ovarian cancer cells in vitro, but the inhibitory effect was restored after adding TMEFF1 exogenous protein. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways analysis showed that ST14 and its related genes were enriched in the processes of epithelial formation, intercellular adhesion, protein localization, and mitosis regulation. We also clarified the kinase, microRNA, and transcription factor target networks and the impact of genetic mutations on prognosis. Overall, high expression of ST14 and TMEFF1 in ovarian cancer predicts higher tumor malignancy and a worse prognosis. ST14 and TMEFF1 co-localize and interact with each other in ovarian cancer. ST14 can regulate TMEFF1 expression to promote proliferation, migration and invasion of ovarian cancer cells. We speculate that the relationship between ST14 and TMEFF1 in ovarian cancer could become a potential target for anti-cancer therapy.
Collapse
Affiliation(s)
- Xin Nie
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Lingling Gao
- Union Hospital, Tongji Medical College, Department of Obstetrics and Gynecology, Huazhong University of Science and Technology, Wuhan, China
| | - Mingjun Zheng
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Shuang Wang
- Department of Gynecology and Obstetrics, Tianjin Central Gynecology and Obstetrics Hospital Affiliated to Nankai University, Tianjin, China
| | - Caixia Wang
- West China Second University Hospital, Department of Obstetrics and Gynecology, Sichuan University, Sichuan, China
| | - Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Ouxuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Rui Gou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Juanjuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Heping District, 110004, Shenyang, China.
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China.
| |
Collapse
|
5
|
Pawar NR, Buzza MS, Duru N, Strong AA, Antalis TM. Matriptase drives dissemination of ovarian cancer spheroids by a PAR-2/PI3K/Akt/MMP9 signaling axis. J Cell Biol 2023; 222:e202209114. [PMID: 37737895 PMCID: PMC10515437 DOI: 10.1083/jcb.202209114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 09/23/2023] Open
Abstract
The transmembrane serine protease matriptase is a key regulator of both barrier-disruptive and protective epithelial cell-cell interactions. Elevated matriptase is a consistent feature of epithelial ovarian cancers (OvCa), where multicellular spheroids shed from the primary tumor into the peritoneal cavity are critical drivers of metastasis. Dynamic cell-to-cell adhesive contacts are required for spheroid formation and maintenance. Here, we show that overactive matriptase, reflected in an increased ratio of matriptase to its inhibitor hepatocyte growth factor activator inhibitor 1 (HAI-1), disrupts cell-cell contacts to produce loose prometastatic spheroids that display increased mesothelial cell adhesion and submesothelial invasion. We show that these activities are dependent on the matriptase activation of a protease-activated receptor-2 (PAR-2) signaling pathway involving PI3K/Akt and MMP9-induced disruption of cell-cell adhesion by the release of the soluble E-cadherin ectodomain. These data reveal a novel pathological connection between matriptase activation of PAR-2 and disruption of cell-cell adhesion, and support the clinical investigation of this signaling axis as a therapeutic strategy for aggressive metastatic OvCa.
Collapse
Affiliation(s)
- Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, USA
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amando A. Strong
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, USA
| |
Collapse
|
6
|
Ehret E, Stroh S, Auberson M, Ino F, Jäger Y, Maillard M, Szabo R, Bugge TH, Frateschi S, Hummler E. Kidney-Specific Membrane-Bound Serine Proteases CAP1/Prss8 and CAP3/St14 Affect ENaC Subunit Abundances but Not Its Activity. Cells 2023; 12:2342. [PMID: 37830556 PMCID: PMC10572026 DOI: 10.3390/cells12192342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
The serine proteases CAP1/Prss8 and CAP3/St14 are identified as ENaC channel-activating proteases in vitro, highly suggesting that they are required for proteolytic activation of ENaC in vivo. The present study tested whether CAP3/St14 is relevant for renal proteolytic ENaC activation and affects ENaC-mediated Na+ absorption following Na+ deprivation conditions. CAP3/St14 knockout mice exhibit a significant decrease in CAP1/Prss8 protein expression with altered ENaC subunit and decreased pNCC protein abundances but overall maintain sodium balance. RNAscope-based analyses reveal co-expression of CAP3/St14 and CAP1/Prss8 with alpha ENaC in distal tubules of the cortex from wild-type mice. Double CAP1/Prss8; CAP3/St14-deficiency maintained Na+ and K+ balance on a Na+-deprived diet, restored ENaC subunit protein abundances but showed reduced NCC activity under Na+ deprivation. Overall, our data clearly show that CAP3/St14 is not required for direct proteolytic activation of ENaC but for its protein abundance. Our study reveals a complex regulation of ENaC by these serine proteases on the expression level rather than on its proteolytic activation.
Collapse
Affiliation(s)
- Elodie Ehret
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- National Center of Competence in Research “Kidney.CH”, 1011 Lausanne, Switzerland
| | - Sévan Stroh
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Muriel Auberson
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Frédérique Ino
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Yannick Jäger
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Marc Maillard
- Service of Nephrology, Department of Medicine, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Roman Szabo
- National Institutes of Health/NIDCR, Bethesda, MD 20892, USA
| | - Thomas H. Bugge
- National Institutes of Health/NIDCR, Bethesda, MD 20892, USA
| | - Simona Frateschi
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Edith Hummler
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- National Center of Competence in Research “Kidney.CH”, 1011 Lausanne, Switzerland
| |
Collapse
|
7
|
Szabo R, Kawaguchi M, Kataoka H, Bugge TH. Early-onset tufting enteropathy in HAI-2-deficient mice is independent of matriptase-mediated cleavage of EpCAM. Development 2023; 150:dev201801. [PMID: 37539662 PMCID: PMC10482385 DOI: 10.1242/dev.201801] [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: 03/22/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
Congenital tufting enteropathy (CTE) is a life-threatening intestinal disorder resulting from loss-of-function mutations in EPCAM and SPINT2. Mice deficient in Spint2, encoding the protease inhibitor HAI-2, develop CTE-like intestinal failure associated with a progressive loss of the EpCAM protein, which is caused by unchecked activity of the serine protease matriptase (ST14). Here, we show that loss of HAI-2 leads to increased proteolytic processing of EpCAM. Elimination of the reported matriptase cleavage site strongly suppressed proteolytic processing of EpCAM in vitro and in vivo. Unexpectedly, expression of cleavage-resistant EpCAM failed to prevent intestinal failure and postnatal lethality in Spint2-deficient mice. In addition, genetic inactivation of intestinal matriptase (St14) counteracted the effect of Spint2 deficiency in mice expressing cleavage-resistant EpCAM, indicating that matriptase does not drive intestinal dysfunction by excessive proteolysis of EpCAM. Interestingly, mice expressing cleavage-resistant EpCAM developed late-onset intestinal defects and exhibited a shortened lifespan even in the presence of HAI-2, suggesting that EpCAM cleavage is indispensable for EpCAM function. Our findings provide new insights into the role of EpCAM and the etiology of the enteropathies driven by Spint2 deficiency.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Frumenzio G, Chandramouli B, Besker N, Grottesi A, Talarico C, Frigerio F, Emerson A, Musiani F. Conformational response to ligand binding of TMPRSS2, a protease involved in SARS-CoV-2 infection: Insights through computational modeling. Proteins 2023; 91:1288-1297. [PMID: 37409524 DOI: 10.1002/prot.26548] [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: 04/12/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
Thanks to the considerable research which has been undertaken in the last few years to improve our understanding of the biology and mechanism of action of SARS-CoV-2, we know how the virus uses its surface spike protein to infect host cells. The transmembrane prosthesis, serine 2 (TMPRSS2) protein, located on the surface of human cells, recognizes the cleavage site in the spike protein, leading to the release of the fusion peptide and entry of the virus into the host cells. Because of its role, TMPRSS2 has been proposed as a drug target to prevent infection by the virus. In this study, we aim to increase our understanding of TMPRSS2 using long scale microsecond atomistic molecular dynamics simulations, focusing on the conformational changes over time. The comparison between simulations conducted on the protein in the native (apo) and inhibited form (holo), has shown that in the holo form the inhibitor stabilizes the catalytic site and induces rearrangements in the extracellular domain of the protein. In turn, it leads to the formation of a new cavity in the vicinity of the ligand binding pocket that is stable in the microsecond time scale. Given the low specificity of known protease inhibitors, these findings suggest a new potential drug target site that can be used to improve TMPRSS2 specific recognition by newly designed inhibitors.
Collapse
Affiliation(s)
- Giorgia Frumenzio
- Super Computing Applications and Innovation, Department HPC, CINECA, Casalecchio di Reno, Italy
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | - Andrew Emerson
- Super Computing Applications and Innovation, Department HPC, CINECA, Casalecchio di Reno, Italy
| | - Francesco Musiani
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Spinello A, D'Anna L, Bignon E, Miclot T, Grandemange S, Terenzi A, Barone G, Barbault F, Monari A. Mechanism of the Covalent Inhibition of Human Transmembrane Protease Serine 2 as an Original Antiviral Strategy. J Phys Chem B 2023. [PMID: 37428676 DOI: 10.1021/acs.jpcb.3c02910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The Transmembrane Protease Serine 2 (TMPRSS2) is a human enzyme which is involved in the maturation and post-translation of different proteins. In addition to being overexpressed in cancer cells, TMPRSS2 plays a further fundamental role in favoring viral infections by allowing the fusion of the virus envelope with the cellular membrane, notably in SARS-CoV-2. In this contribution, we resort to multiscale molecular modeling to unravel the structural and dynamical features of TMPRSS2 and its interaction with a model lipid bilayer. Furthermore, we shed light on the mechanism of action of a potential inhibitor (nafamostat), determining the free-energy profile associated with the inhibition reaction and showing the facile poisoning of the enzyme. Our study, while providing the first atomistically resolved mechanism of TMPRSS2 inhibition, is also fundamental in furnishing a solid framework for further rational design targeting transmembrane proteases in a host-directed antiviral strategy.
Collapse
Affiliation(s)
- Angelo Spinello
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Luisa D'Anna
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Emmanuelle Bignon
- Université de Lorraine and CNRS, UMR 7019 LPCT, F-54000 Nancy, France
| | - Tom Miclot
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
- Université de Lorraine and CNRS, UMR 7019 LPCT, F-54000 Nancy, France
| | | | - Alessio Terenzi
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Giampaolo Barone
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | | | - Antonio Monari
- Université Paris Cité and CNRS, ITODYS, F-75006 Paris, France
| |
Collapse
|
10
|
Wu Q, Li S, Zhang X, Dong N. Type II Transmembrane Serine Proteases as Modulators in Adipose Tissue Phenotype and Function. Biomedicines 2023; 11:1794. [PMID: 37509434 PMCID: PMC10376093 DOI: 10.3390/biomedicines11071794] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Adipose tissue is a crucial organ in energy metabolism and thermoregulation. Adipose tissue phenotype is controlled by various signaling mechanisms under pathophysiological conditions. Type II transmembrane serine proteases (TTSPs) are a group of trypsin-like enzymes anchoring on the cell surface. These proteases act in diverse tissues to regulate physiological processes, such as food digestion, salt-water balance, iron metabolism, epithelial integrity, and auditory nerve development. More recently, several members of the TTSP family, namely, hepsin, matriptase-2, and corin, have been shown to play a role in regulating lipid metabolism, adipose tissue phenotype, and thermogenesis, via direct growth factor activation or indirect hormonal mechanisms. In mice, hepsin deficiency increases adipose browning and protects from high-fat diet-induced hyperglycemia, hyperlipidemia, and obesity. Similarly, matriptase-2 deficiency increases fat lipolysis and reduces obesity and hepatic steatosis in high-fat diet-fed mice. In contrast, corin deficiency increases white adipose weights and cell sizes, suppresses adipocyte browning and thermogenic responses, and causes cold intolerance in mice. These findings highlight an important role of TTSPs in modifying cellular phenotype and function in adipose tissue. In this review, we provide a brief description about TTSPs and discuss recent findings regarding the role of hepsin, matriptase-2, and corin in regulating adipose tissue phenotype, energy metabolism, and thermogenic responses.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Shuo Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xianrui Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, Soochow University, Suzhou 215006, China
| |
Collapse
|
11
|
Gaymon DO, Barndt R, Stires H, Riggins RB, Johnson MD. ROS is a master regulator of in vitro matriptase activation. PLoS One 2023; 18:e0267492. [PMID: 36716335 PMCID: PMC9886240 DOI: 10.1371/journal.pone.0267492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Matriptase is a type II transmembrane serine protease that is widely expressed in normal epithelial cells and epithelial cancers. Studies have shown that regulation of matriptase expression and activation becomes deranged in several cancers and is associated with poor disease-free survival. Although the central mechanism of its activation has remained unknown, our lab has previously demonstrated that inflammatory conditions such as intracellular pH decrease strongly induces matriptase activation. In this investigation, we first demonstrate clear matriptase activation following Fulvestrant (ICI) and Tykerb (Lapatinib) treatment in HER2-amplified, estrogen receptor (ER)-positive BT474, MDA-MB-361 and ZR-75-30 or single ER-positive MCF7 cells, respectively. This activation modestly involved Phosphoinositide 3-kinase (PI3K) activation and occurred as quickly as six hours post treatment. We also demonstrate that matriptase activation is not a universal hallmark of stress, with Etoposide treated cells showing a larger degree of matriptase activation than Lapatinib and ICI-treated cells. While etoposide toxicity has been shown to be mediated through reactive oxygen species (ROS) and MAPK/ERK kinase (MEK) activity, MEK activity showed no correlation with matriptase activation. Novelly, we demonstrate that endogenous and exogenous matriptase activation are ROS-mediated in vitro and inhibited by N-acetylcysteine (NAC). Lastly, we demonstrate matriptase-directed NAC treatment results in apoptosis of several breast cancer cell lines either alone or in combination with clinically used therapeutics. These data demonstrate the contribution of ROS-mediated survival, its independence of kinase-mediated survival, and the plausibility of using matriptase activation to indicate the potential success of antioxidant therapy.
Collapse
Affiliation(s)
- Darius O. Gaymon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
- * E-mail:
| | - Robert Barndt
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Hillary Stires
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Rebecca B. Riggins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Michael. D. Johnson
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| |
Collapse
|
12
|
Higashi T, Saito AC, Fukazawa Y, Furuse M, Higashi AY, Ono M, Chiba H. EpCAM proteolysis and release of complexed claudin-7 repair and maintain the tight junction barrier. J Cell Biol 2022; 222:213688. [PMID: 36378161 PMCID: PMC9671161 DOI: 10.1083/jcb.202204079] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
TJs maintain the epithelial barrier by regulating paracellular permeability. Since TJs are under dynamically fluctuating intercellular tension, cells must continuously survey and repair any damage. However, the underlying mechanisms allowing cells to sense TJ damage and repair the barrier are not yet fully understood. Here, we showed that proteinases play an important role in the maintenance of the epithelial barrier. At TJ break sites, EpCAM-claudin-7 complexes on the basolateral membrane become accessible to apical membrane-anchored serine proteinases (MASPs) and the MASPs cleave EpCAM. Biochemical data and imaging analysis suggest that claudin-7 released from EpCAM contributes to the rapid repair of damaged TJs. Knockout (KO) of MASPs drastically reduced barrier function and live-imaging of TJ permeability showed that MASPs-KO cells exhibited increased size, duration, and frequency of leaks. Together, our results reveal a novel mechanism of TJ maintenance through the localized proteolysis of EpCAM at TJ leaks, and provide a better understanding of the dynamic regulation of epithelial permeability.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan,Correspondence to Tomohito Higashi:
| | - Akira C. Saito
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan,Department of Physiological Sciences, School of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Atsuko Y. Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Masahiro Ono
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
13
|
Lee SL, Lee MHH, Wu KJ, Chiang CW, Chang YX, Fang JD, Tung HH, Kuo LW, Wang Y. Post-ischemic protection of hepatocyte growth factor requires the type II transmembrane serine protease matriptase-A reciprocal regulation of the two for neuroprotection in stroke brain. FASEB J 2022; 36:e22494. [PMID: 35976173 DOI: 10.1096/fj.202200414r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 11/11/2022]
Abstract
In a rat middle cerebral artery occlusion (MACo) model of ischemic stroke, intracerebroventricular administration of human recombinant hepatocyte growth factor (HGF) mitigated motor impairment and cortical infarction. Recombinant HGF reduced MCAo-induced TNFα and IL1β expression, and alleviated perilesional reactivation of microglia and astrocyte. All of the aforementioned beneficial effects of HGF were antagonized by an inhibitor to the type II transmembrane serine protease matriptase (MTP). MCAo upregulated MTP mRNA and protein in the lesioned cortex. MTP protein, not the mRNA, was increased further by recombinant HGF but reduced when MTP inhibitor (MTPi) was added to the treatment. Changes of the endogenous active HGF by MCAo, HGF or MTPi paralleled with the changes of MTP protein under the same conditions whilst neither HGF mRNA nor the total endogenous HGF protein were altered. These data showed that the therapeutic effects of HGF in stroke brain is attributed to its proteolytic activation and that MTP is a main protease of the event. MCAo enhanced MTP mRNA and thus protein expression; the initial use of the recombinant active HGF stabilized MCAo-induced MTP protein and subsequent activation of endogenous latent HGF which in turn stabilized further MTP protein. A reciprocal regulation between MTP and HGF appears to be present where MTP promotes HGF activation and the active HGF prevents MTP protein turnover. This study, for the first time, shows that MTP can participate in neural protection in stroke brain through activation of HGF. The cycles of HGF-MTP regulation achieved preservation of the neurological activity.
Collapse
Affiliation(s)
- Sheau-Ling Lee
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, R.O.C
| | - Michelle Hui-Hsin Lee
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Chia-Wen Chiang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Yun-Xuan Chang
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Jung-Da Fang
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Hsiu-Hui Tung
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| |
Collapse
|
14
|
Structure and activity of human TMPRSS2 protease implicated in SARS-CoV-2 activation. Nat Chem Biol 2022; 18:963-971. [PMID: 35676539 DOI: 10.1038/s41589-022-01059-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 05/09/2022] [Indexed: 02/07/2023]
Abstract
Transmembrane protease, serine 2 (TMPRSS2) has been identified as key host cell factor for viral entry and pathogenesis of SARS-CoV-2. Specifically, TMPRSS2 proteolytically processes the SARS-CoV-2 Spike (S) protein, enabling virus-host membrane fusion and infection of the airways. We present here a recombinant production strategy for enzymatically active TMPRSS2 and characterization of its matured proteolytic activity, as well as its 1.95 Å X-ray cocrystal structure with the synthetic protease inhibitor nafamostat. Our study provides a structural basis for the potent but nonspecific inhibition by nafamostat and identifies distinguishing features of the TMPRSS2 substrate binding pocket that explain specificity. TMPRSS2 cleaved SARS-CoV-2 S protein at multiple sites, including the canonical S1/S2 cleavage site. We ranked the potency of clinical protease inhibitors with half-maximal inhibitory concentrations ranging from 1.4 nM to 120 µM and determined inhibitor mechanisms of action, providing the groundwork for drug development efforts to selectively inhibit TMPRSS2.
Collapse
|
15
|
Anand D, Hummler E, Rickman OJ. ENaC activation by proteases. Acta Physiol (Oxf) 2022; 235:e13811. [PMID: 35276025 PMCID: PMC9540061 DOI: 10.1111/apha.13811] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
Proteases are fundamental for a plethora of biological processes, including signalling and tissue remodelling, and dysregulated proteolytic activity can result in pathogenesis. In this review, we focus on a subclass of membrane‐bound and soluble proteases that are defined as channel‐activating proteases (CAPs), since they induce Na+ ion transport through an autocrine mechanism when co‐expressed with the highly amiloride‐sensitive epithelial sodium channel (ENaC) in Xenopus oocytes. These experiments first identified CAP1 (channel‐activating protease 1, prostasin) followed by CAP2 (channel‐activating protease 2, TMPRSS4) and CAP3 (channel‐activating protease 3, matriptase) as in vitro mediators of ENaC current. Since then, more serine‐, cysteine‐ and metalloproteases were confirmed as in vitro CAPs that potentially cleave and regulate ENaC, and thus this nomenclature was not further followed, but is accepted as functional term or alias. The precise mechanism of ENaC modulation by proteases has not been fully elucidated. Studies in organ‐specific protease knockout models revealed evidence for their role in increasing ENaC activity, although the proteases responsible for ENaC activation are yet to be identified. We summarize recent findings in animal models of these CAPs with respect to their implication in ENaC activation. We discuss the consequences of dysregulated CAPs underlying epithelial phenotypes in pathophysiological conditions, and the role of selected protease inhibitors. We believe that these proteases may present interesting therapeutic targets for diseases with aberrant sodium homoeostasis.
Collapse
Affiliation(s)
- Deepika Anand
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| | - Edith Hummler
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| | - Olivia J. Rickman
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| |
Collapse
|
16
|
Ernst J, Hiasat J, Alabek ML, Scanga HL, Motley W, Nischal KK. Expansion of the ophthalmic phenotype of SPINT2-related syndromic congenital sodium diarrhea. Am J Med Genet A 2021; 185:1270-1274. [PMID: 33547739 DOI: 10.1002/ajmg.a.62094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/08/2022]
Abstract
A 5-year-old girl presented with treatment-refractory dry eye and recurrent episodes of eye pain. She had been previously diagnosed with syndromic congenital sodium diarrhea (SCSD) caused by a pathogenic variant in SPINT2. Her local pediatric ophthalmologist had made the diagnosis of severe dry eye with corneal erosions, based on which, we arranged an eye exam under anesthesia (EUA) and punctal plug placement. Anterior segment optical coherence tomography (OCT) and corneal photographs were taken during the procedure. There are reports describing similar ophthalmic findings in this syndrome. However, to the best of our knowledge, this is the first case report to document OCT imaging and corneal photographs in a patient with SCSD, which we feel expands the ophthalmic phenotype of this rare genetic disorder.
Collapse
Affiliation(s)
- Julia Ernst
- Eye Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Division of Pediatric Ophthalmology, Strabismus and Adult Motility, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Medical University of Warsaw, Warsaw, Poland
| | - Jamila Hiasat
- Eye Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Division of Pediatric Ophthalmology, Strabismus and Adult Motility, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michelle L Alabek
- Eye Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Division of Pediatric Ophthalmology, Strabismus and Adult Motility, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hannah L Scanga
- Eye Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Division of Pediatric Ophthalmology, Strabismus and Adult Motility, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William Motley
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Ken K Nischal
- Eye Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Division of Pediatric Ophthalmology, Strabismus and Adult Motility, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Kavková M, Šulcová M, Dumková J, Zahradníček O, Kaiser J, Tucker AS, Zikmund T, Buchtová M. Coordinated labio-lingual asymmetries in dental and bone development create a symmetrical acrodont dentition. Sci Rep 2020; 10:22040. [PMID: 33328503 PMCID: PMC7745041 DOI: 10.1038/s41598-020-78939-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 11/26/2020] [Indexed: 11/09/2022] Open
Abstract
Organs throughout the body develop both asymmetrically and symmetrically. Here, we assess how symmetrical teeth in reptiles can be created from asymmetrical tooth germs. Teeth of lepidosaurian reptiles are mostly anchored to the jaw bones by pleurodont ankylosis, where the tooth is held in place on the labial side only. Pleurodont teeth are characterized by significantly asymmetrical development of the labial and lingual sides of the cervical loop, which later leads to uneven deposition of hard tissue. On the other hand, acrodont teeth found in lizards of the Acrodonta clade (i.e. agamas, chameleons) are symmetrically ankylosed to the jaw bone. Here, we have focused on the formation of the symmetrical acrodont dentition of the veiled chameleon (Chamaeleo calyptratus). Intriguingly, our results revealed distinct asymmetries in morphology of the labial and lingual sides of the cervical loop during early developmental stages, both at the gross and ultrastructural level, with specific patterns of cell proliferation and stem cell marker expression. Asymmetrical expression of ST14 was also observed, with a positive domain on the lingual side of the cervical loop overlapping with the SOX2 domain. In contrast, micro-CT analysis of hard tissues revealed that deposition of dentin and enamel was largely symmetrical at the mineralization stage, highlighting the difference between cervical loop morphology during early development and differentiation of odontoblasts throughout later odontogenesis. In conclusion, the early asymmetrical development of the enamel organ seems to be a plesiomorphic character for all squamate reptiles, while symmetrical and precisely orchestrated deposition of hard tissue during tooth formation in acrodont dentitions probably represents a novelty in the Acrodonta clade.
Collapse
Affiliation(s)
- M Kavková
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - M Šulcová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveri 97, 602 00, Brno, Czech Republic
| | - J Dumková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - O Zahradníček
- Department of Radiation Dosimetry, Nuclear Physics Institute, Czech Academy of Sciences, Prague, Czech Republic
| | - J Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - A S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27 Guy's Tower, Guy's Hospital, London Bridge, London, UK
| | - T Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - M Buchtová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic. .,Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveri 97, 602 00, Brno, Czech Republic.
| |
Collapse
|
18
|
Armistead J, Hatzold J, van Roye A, Fahle E, Hammerschmidt M. Entosis and apical cell extrusion constitute a tumor-suppressive mechanism downstream of Matriptase. J Cell Biol 2020; 219:132730. [PMID: 31819976 PMCID: PMC7041680 DOI: 10.1083/jcb.201905190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/02/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Armistead et al. show that in a bilayered epithelium in vivo, apical cell extrusion of basal cells is achieved via their engulfment by surface cells. In zebrafish hai1a mutants, this constitutes a tumor-suppressive mechanism, revealing a double face of Matriptase. The type II transmembrane serine protease Matriptase 1 (ST14) is commonly known as an oncogene, yet it also plays an understudied role in suppressing carcinogenesis. This double face is evident in the embryonic epidermis of zebrafish loss-of-function mutants in the cognate Matriptase inhibitor Hai1a (Spint1a). Mutant embryos display epidermal hyperplasia, but also apical cell extrusions, during which extruding outer keratinocytes carry out an entosis-like engulfment and entrainment of underlying basal cells, constituting a tumor-suppressive effect. These counteracting Matriptase effects depend on EGFR and the newly identified mediator phospholipase D (PLD), which promotes both mTORC1-dependent cell proliferation and sphingosine-1-phosphate (S1P)–dependent entosis and apical cell extrusion. Accordingly, hypomorphic hai1a mutants heal spontaneously, while otherwise lethal hai1a amorphs are efficiently rescued upon cotreatment with PLD inhibitors and S1P. Together, our data elucidate the mechanisms underlying the double face of Matriptase function in vivo and reveal the potential use of combinatorial carcinoma treatments when such double-face mechanisms are involved.
Collapse
Affiliation(s)
- Joy Armistead
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Anna van Roye
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Evelin Fahle
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Jia B, Thompson HA, Barndt RB, Chiu YL, Lee MJ, Lee SC, Wang JK, Tang HJ, Lin CY, Johnson MD. Mild acidity likely accelerates the physiological matriptase autoactivation process: a comparative study between spontaneous and acid-induced matriptase zymogen activation. Hum Cell 2020; 33:1068-1080. [PMID: 32779152 DOI: 10.1007/s13577-020-00410-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022]
Abstract
The pathophysiological functions of matriptase, a type 2 transmembrane serine protease, rely primarily on its enzymatic activity, which is under tight control through multiple mechanisms. Among those regulatory mechanisms, the control of zymogen activation is arguably the most important. Matriptase zymogen activation not only generates the mature active enzyme but also initiates suppressive mechanisms, such as rapid inhibition by HAI-1, and matriptase shedding. These tightly coupled events allow the potent matriptase tryptic activity to fulfill its biological functions at the same time as limiting undesired hazards. Matriptase is converted to the active enzyme via a process of autoactivation, in which the activational cleavage is thought to rely on the interactions of matriptase zymogen molecules and other as yet identified proteins. Matriptase autoactivation can occur spontaneously and is rapidly followed by the formation and then shedding of matriptase-HAI-1 complexes, resulting in the presence of relatively low levels of the complex on cells. Activation can also be induced by several non-protease factors, such as the exposure of cells to a mildly acidic buffer, which rapidly causes high-level matriptase zymogen activation in almost all cell lines tested. In the current study, the structural requirements for this acid-induced zymogen activation are compared with those required for spontaneous activation through a systematic analysis of the impact of 18 different mutations in various structural domains and motifs on matriptase zymogen activation. Our study reveals that both acid-induced matriptase activation and spontaneous activation depend on the maintenance of the structural integrity of the serine protease domain, non-catalytic domains, and posttranslational modifications. The common requirements of both modes of activation suggest that acid-induced matriptase activation may function as a physiological mechanism to induce pericellular proteolysis by accelerating matriptase autoactivation.
Collapse
Affiliation(s)
- Bailing Jia
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, 450003, China.,Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Hamishi A Thompson
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Robert B Barndt
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Yi-Lin Chiu
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA.,Department of Biochemistry National Defense Medical Center, Taipei, 114, Taiwan
| | - Mon-Juan Lee
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA.,Department of Bioscience Technology, Chang Jung Christian University, Tainan, 71101, Taiwan.,Department of Medical Science, Chang Jung Christian University, Tainan, 71101, Taiwan
| | - See-Chi Lee
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Jehng-Kang Wang
- Department of Biochemistry National Defense Medical Center, Taipei, 114, Taiwan
| | - Hung-Jen Tang
- Section of Infectious Diseases, Internal Medicine, Chi-Mei Medical Center, No.901, Chung-Hwa Rd. Yung-Kang Dist., Tainan City, 71004, Taiwan, ROC.
| | - Chen-Yong Lin
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA.
| | - Michael D Johnson
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, W412 Research Building 3970 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
20
|
Inhibition of an active zymogen protease: the zymogen form of matriptase is regulated by HAI-1 and HAI-2. Biochem J 2020; 477:1779-1794. [PMID: 32338287 DOI: 10.1042/bcj20200182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 11/17/2022]
Abstract
The membrane-bound serine protease matriptase belongs to a rare subset of serine proteases that display significant activity in the zymogen form. Matriptase is critically involved in epithelial differentiation and homeostasis, and insufficient regulation of its proteolytic activity directly causes onset and development of malignant cancer. There is strong evidence that the zymogen activity of matriptase is sufficient for its biological function(s). Activated matriptase is inhibited by the two Kunitz-type inhibitor domain-containing hepatocyte growth factor activator inhibitors 1 (HAI-1) and HAI-2, however, it remains unknown whether the activity of the matriptase zymogen is regulated. Using both purified proteins and a cell-based assay, we show that the catalytic activity of the matriptase zymogen towards a peptide-based substrate as well as the natural protein substrates, pro-HGF and pro-prostasin, can be inhibited by HAI-1 and HAI-2. Inhibition of zymogen matriptase by HAI-1 and HAI-2 appears similar to inhibition of activated matriptase and occurs at comparable inhibitor concentrations. This indicates that HAI-1 and HAI-2 interact with the active sites of zymogen and activated matriptase in a similar manner. Our results suggest that HAI-1 and HAI-2 regulate matriptase zymogen activity and thus may act as regulators of matriptase trans(auto)-activation. Due to the main localisation of HAI-2 in the ER and HAI-1 in the secretory pathway and on the cell surface, this regulation likely occurs both in the secretory pathway and on the plasma membrane. Regulation of an active zymogen form of a protease is a novel finding.
Collapse
|
21
|
Membrane-anchored serine proteases as regulators of epithelial function. Biochem Soc Trans 2020; 48:517-528. [PMID: 32196551 PMCID: PMC9869603 DOI: 10.1042/bst20190675] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
Cleavage of proteins in the extracellular milieu, including hormones, growth factors and their receptors, ion channels, and various cell adhesion and extracellular matrix molecules, plays a key role in the regulation of cell behavior. Among more than 500 proteolytic enzymes encoded by mammalian genomes, membrane-anchored serine proteases (MASPs), which are expressed on the surface of epithelial cells of all major organs, are excellently suited to mediate signal transduction across the epithelia and are increasingly being recognized as important regulators of epithelial development, function, and disease [ 1-3]. In this minireview, we summarize current knowledge of the in vivo roles of MASPs in acquisition and maintenance of some of the defining functions of epithelial tissues, such as barrier formation, ion transport, and sensory perception.
Collapse
|
22
|
Wu CJ, Lu M, Feng X, Nakato G, Udey MC. Matriptase Cleaves EpCAM and TROP2 in Keratinocytes, Destabilizing Both Proteins and Associated Claudins. Cells 2020; 9:cells9041027. [PMID: 32326212 PMCID: PMC7226414 DOI: 10.3390/cells9041027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023] Open
Abstract
The homologs EpCAM and TROP2, which both interact with claudin-1 and claudin-7, are frequently coexpressed in epithelia including skin. Intestine uniquely expresses high levels of EpCAM but not TROP2. We previously identified EpCAM as a substrate of the membrane-anchored protease matriptase and linked HAI-2, matriptase, EpCAM and claudin-7 in a pathway that is pivotal for intestinal epithelial cells (IEC) homeostasis. Herein, we reveal that TROP2 is also a matriptase substrate. Matriptase cleaved TROP2 when purified recombinant proteins were mixed in vitro. TROP2, like EpCAM, was also cleaved after co-transfection of matriptase in 293T cells. Neither EpCAM nor TROP2 cleavage was promoted by protease-disabled matriptase or matriptase that harbored the ichthyosis-associated G827R mutation. We confirmed that EpCAM and TROP2 are both expressed in skin and detected cleavage of these proteins in human keratinocytes (HaCaT cells) after the physiologic inhibition of matriptase by HAI proteins was relieved by siRNA knockdown. Knockdown of EpCAM or TROP2 individually had only small effects on claudin-1 and claudin-7 levels, whereas elimination of both markedly diminished claudin levels. HAI-1 knockdown promoted EpCAM and TROP2 cleavage accompanied by reductions in claudins, whereas HAI-2 knockdown had little impact. Double knockdown of HAI-1 and HAI-2 induced nearly complete cleavage of EpCAM and TROP2 and drastic reductions of claudins. These effects were eliminated by concurrent matriptase knockdown. Decreases in claudin levels were also diminished by the lysosomal inhibitor chloroquine and cleaved EpCAM/TROP2 fragments accumulated preferentially. We demonstrate that TROP2 and EpCAM exhibit redundancies with regard to regulation of claudin metabolism and that an HAI, matriptase, EpCAM and claudin pathway analogous to what we described in IECs exists in keratinocytes. This study may offer insights into the mechanistic basis for matriptase dysregulation-induced ichthyosis.
Collapse
Affiliation(s)
- Chuan-Jin Wu
- Laboratory of Immune Cell Biology, National Cancer Institute, Bethesda, MD 20892, USA
- Correspondence: (C.-J.W.); (M.C.U.); Tel.: +1-301-760-7452 (C.-J.W.); +1-314-454-8547 (M.C.U.)
| | - Michael Lu
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Xu Feng
- Retired from National Cancer Institute, Bethesda, MD 20892, USA;
| | - Gaku Nakato
- Kanagawa Institute of Industrial Science and Technology, Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-0821, Japan;
| | - Mark C. Udey
- Dermatology Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Correspondence: (C.-J.W.); (M.C.U.); Tel.: +1-301-760-7452 (C.-J.W.); +1-314-454-8547 (M.C.U.)
| |
Collapse
|
23
|
Kriaa A, Jablaoui A, Mkaouar H, Akermi N, Maguin E, Rhimi M. Serine proteases at the cutting edge of IBD: Focus on gastrointestinal inflammation. FASEB J 2020; 34:7270-7282. [PMID: 32307770 DOI: 10.1096/fj.202000031rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
Serine proteases have been long recognized to coordinate many physiological processes and play key roles in regulating the inflammatory response. Accordingly, their dysregulation has been regularly associated with several inflammatory disorders and suggested as a central mechanism in the pathophysiology of digestive inflammation. So far, studies addressing the proteolytic homeostasis in the gut have mainly focused on host serine proteases as candidates of interest, while largely ignoring the potential contribution of their bacterial counterparts. The human gut microbiota comprises a complex ecosystem that contributes to host health and disease. Yet, our understanding of microbially produced serine proteases and investigation of whether they are causally linked to IBD is still in its infancy. In this review, we highlight recent advances in the emerging roles of host and bacterial serine proteases in digestive inflammation. We also discuss the application of available tools in the gut to monitor disease-related serine proteases. An exhaustive representation and understanding of such functional potential would help in closing existing gaps in mechanistic knowledge.
Collapse
Affiliation(s)
- Aicha Kriaa
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Amin Jablaoui
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Héla Mkaouar
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Nizar Akermi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Emmanuelle Maguin
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| |
Collapse
|
24
|
Holt-Danborg L, Vodopiutz J, Nonboe AW, De Laffolie J, Skovbjerg S, Wolters VM, Müller T, Hetzer B, Querfurt A, Zimmer KP, Jensen JK, Entenmann A, Heinz-Erian P, Vogel LK, Janecke AR. SPINT2 (HAI-2) missense variants identified in congenital sodium diarrhea/tufting enteropathy affect the ability of HAI-2 to inhibit prostasin but not matriptase. Hum Mol Genet 2020; 28:828-841. [PMID: 30445423 DOI: 10.1093/hmg/ddy394] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
The syndromic form of congenital sodium diarrhea (SCSD) is caused by bi-allelic mutations in SPINT2, which encodes a Kunitz-type serine protease inhibitor (HAI-2). We report three novel SCSD patients, two novel SPINT2 mutations and review published cases. The most common findings in SCSD patients were choanal atresia (20/34) and keratitis of infantile onset (26/34). Characteristic epithelial tufts on intestinal histology were reported in 13/34 patients. Of 13 different SPINT2 variants identified in SCSD, 4 are missense variants and localize to the second Kunitz domain (KD2) of HAI-2. HAI-2 has been implicated in the regulation of the activities of several serine proteases including prostasin and matriptase, which are both important for epithelial barrier formation. No patient with bi-allelic stop mutations was identified, suggesting that at least one SPINT2 allele encoding a protein with residual HAI-2 function is necessary for survival. We show that the SCSD-associated HAI-2 variants p.Phe161Val, p.Tyr163Cys and p.Gly168Ser all display decreased ability to inhibit prostasin-catalyzed cleavage. However, the SCSD-associated HAI-2 variants inhibited matriptase as efficiently as the wild-type HAI-2. Homology modeling indicated limited solvent exposure of the mutated amino acids, suggesting that they induce misfolding of KD2. This suggests that prostasin needs to engage with an exosite motif located on KD2 in addition to the binding loop (Cys47/Arg48) located on the first Kunitz domain in order to inhibit prostasin. In conclusion our data suggests that SCSD is caused by lack of inhibition of prostasin or a similar protease in the secretory pathway or on the plasma membrane.
Collapse
Affiliation(s)
- Lasse Holt-Danborg
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Julia Vodopiutz
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna
| | - Annika W Nonboe
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Jan De Laffolie
- Abteilung Allgemeine Pädiatrie und Neonatologie, Zentrum für Kinderheilkunde und Jugendmedizin, Justus-Liebig-Universität, Gießen, Germany
| | - Signe Skovbjerg
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Victorien M Wolters
- Department of Pediatric Gastroenterology, WKZ/ UMC Utrecht, Utrecht, The Netherlands
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Hetzer
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Querfurt
- Gesundheit Nord gGmbH, Klinikverbund Bremen, Klinik für Kinder und Jugendmedizin, Professor-Hess-Kinderklinik, Klinikum Bremen-Mitte, Bremen, Germany
| | - Klaus-Peter Zimmer
- Abteilung Allgemeine Pädiatrie und Neonatologie, Zentrum für Kinderheilkunde und Jugendmedizin, Justus-Liebig-Universität, Gießen, Germany
| | - Jan K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Andreas Entenmann
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Heinz-Erian
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Lotte K Vogel
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Andreas R Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Abstract
Over the last two decades, a novel subgroup of serine proteases, the cell surface-anchored serine proteases, has emerged as an important component of the human degradome, and several members have garnered significant attention for their roles in cancer progression and metastasis. A large body of literature describes that cell surface-anchored serine proteases are deregulated in cancer and that they contribute to both tumor formation and metastasis through diverse molecular mechanisms. The loss of precise regulation of cell surface-anchored serine protease expression and/or catalytic activity may be contributing to the etiology of several cancer types. There is therefore a strong impetus to understand the events that lead to deregulation at the gene and protein levels, how these precipitate in various stages of tumorigenesis, and whether targeting of selected proteases can lead to novel cancer intervention strategies. This review summarizes current knowledge about cell surface-anchored serine proteases and their role in cancer based on biochemical characterization, cell culture-based studies, expression studies, and in vivo experiments. Efforts to develop inhibitors to target cell surface-anchored serine proteases in cancer therapy will also be summarized.
Collapse
|
26
|
Szabo R, Callies LK, Bugge TH. Matriptase drives early-onset intestinal failure in a mouse model of congenital tufting enteropathy. Development 2019; 146:dev183392. [PMID: 31628112 PMCID: PMC6899019 DOI: 10.1242/dev.183392] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
Syndromic congenital tufting enteropathy (CTE) is a life-threatening recessive human genetic disorder that is caused by mutations in SPINT2, encoding the protease inhibitor HAI-2, and is characterized by severe intestinal dysfunction. We recently reported the generation of a Spint2-deficient mouse model of CTE. Here, we show that the CTE-associated early-onset intestinal failure and lethality of Spint2-deficient mice is caused by unchecked activity of the serine protease matriptase. Macroscopic and histological defects observed in the absence of HAI-2, including villous atrophy, luminal bleeding, loss of mucin-producing goblet cells, loss of defined crypt architecture and the resulting acute inflammatory response in the large intestine, were all prevented by intestinal-specific inactivation of the St14 gene encoding matriptase. The CTE-associated loss of the cell junctional proteins EpCAM and claudin 7 was also prevented. As a result, inactivation of intestinal matriptase allowed Spint2-deficient mice to gain weight after birth and dramatically increased their lifespan. These data implicate matriptase as a causative agent in the development of CTE and may provide a new target for the treatment of CTE in individuals carrying SPINT2 mutations.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - LuLu K Callies
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Distinct Localization of Mature HGF from its Precursor Form in Developing and Repairing the Stomach. Int J Mol Sci 2019; 20:ijms20122955. [PMID: 31212972 PMCID: PMC6628191 DOI: 10.3390/ijms20122955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/06/2019] [Accepted: 06/13/2019] [Indexed: 01/23/2023] Open
Abstract
Hepatocyte growth factor (HGF) is secreted as an inactive single-chain HGF (scHGF); however, only proteolytically processed two-chain HGF (tcHGF) can activate the MET receptor. We investigated the localization of tcHGF and activated/phosphorylated MET (pMET) using a tcHGF-specific antibody. In day 16.5 mouse embryos, total HGF (scHGF + tcHGF) was mainly localized in smooth muscle cells close to, but separate from, MET-positive epithelial cells in endodermal organs, including the stomach. In the adult stomach, total HGF was localized in smooth muscle cells, and tcHGF was mainly localized in the glandular base region. Immunostaining for pMET and Lgr5-driven green fluorescent protein (GFP) indicated that pMET localization overlapped with Lgr5+ gastric stem cells. HGF promoted organoid formation similar to EGF, indicating the potential for HGF to promote the survival and growth of gastric stem cells. pMET and tcHGF localizations changed during regeneration following gastric injury. These results indicate that MET is constantly activated in gastric stem cells and that the localization of pMET differs from the primary localization of precursor HGF but has a close relationship to tcHGF. Our results suggest the importance of the microenvironmental generation of tcHGF in the regulation of development, regeneration, and stem cell behavior.
Collapse
|
28
|
Recent progress on inhibitors of the type II transmembrane serine proteases, hepsin, matriptase and matriptase-2. Future Med Chem 2019; 11:743-769. [DOI: 10.4155/fmc-2018-0446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Members of the type II transmembrane serine proteases (TTSP) family play a vital role in cell growth and development but many are also implicated in disease. Two of the well-studied TTSPs, matriptase and hepsin proteolytically process multiple protein substrates such as the inactive single-chain zymogens pro-HGF and pro-macrophage stimulating protein into the active heterodimeric forms, HGF and macrophage stimulating protein. These two proteases also have many other substrates which are associated with cancer and tumor progression. Another related TTSP, matriptase-2 is expressed in the liver and functions by regulating iron homoeostasis through the cleavage of hemojuvelin and thus is implicated in iron overload diseases. In the present review, we will discuss inhibitor design strategy and Structure activity relationships of TTSP inhibitors, which have been reported in the literature.
Collapse
|
29
|
Pawar NR, Buzza MS, Antalis TM. Membrane-Anchored Serine Proteases and Protease-Activated Receptor-2-Mediated Signaling: Co-Conspirators in Cancer Progression. Cancer Res 2019; 79:301-310. [PMID: 30610085 DOI: 10.1158/0008-5472.can-18-1745] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/12/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022]
Abstract
Pericellular proteolysis provides a significant advantage to developing tumors through the ability to remodel the extracellular matrix, promote cell invasion and migration, and facilitate angiogenesis. Recent advances demonstrate that pericellular proteases can also communicate directly to cells by activation of a unique group of transmembrane G-protein-coupled receptors (GPCR) known as protease-activated receptors (PAR). In this review, we discuss the specific roles of one of four mammalian PARs, namely PAR-2, which is overexpressed in advanced stage tumors and is activated by trypsin-like serine proteases that are highly expressed or otherwise dysregulated in many cancers. We highlight recent insights into the ability of different protease agonists to bias PAR-2 signaling and the newly emerging evidence for an interplay between PAR-2 and membrane-anchored serine proteases, which may co-conspire to promote tumor progression and metastasis. Interfering with these pathways might provide unique opportunities for the development of new mechanism-based strategies for the treatment of advanced and metastatic cancers.
Collapse
Affiliation(s)
- Nisha R Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Marguerite S Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Toni M Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland. .,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
30
|
Drees L, Königsmann T, Jaspers MHJ, Pflanz R, Riedel D, Schuh R. Conserved function of the matriptase-prostasin proteolytic cascade during epithelial morphogenesis. PLoS Genet 2019; 15:e1007882. [PMID: 30601807 PMCID: PMC6331135 DOI: 10.1371/journal.pgen.1007882] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/14/2019] [Accepted: 12/06/2018] [Indexed: 01/08/2023] Open
Abstract
Extracellular matrix (ECM) assembly and remodelling is critical during development and organ morphogenesis. Dysregulation of ECM is implicated in many pathogenic conditions, including cancer. The type II transmembrane serine protease matriptase and the serine protease prostasin are key factors in a proteolytic cascade that regulates epithelial ECM differentiation during development in vertebrates. Here, we show by rescue experiments that the Drosophila proteases Notopleural (Np) and Tracheal-prostasin (Tpr) are functional homologues of matriptase and prostasin, respectively. Np mediates morphogenesis and remodelling of apical ECM during tracheal system development and is essential for maintenance of the transepithelial barrier function. Both Np and Tpr degrade the zona pellucida-domain (ZP-domain) protein Dumpy, a component of the transient tracheal apical ECM. Furthermore, we demonstrate that Tpr zymogen and the ZP domain of the ECM protein Piopio are cleaved by Np and matriptase in vitro. Our data indicate that the evolutionarily conserved ZP domain, present in many ECM proteins of vertebrates and invertebrates, is a novel target of the conserved matriptase-prostasin proteolytic cascade. Epithelial tissue covers the outside of the animal body and lines internal organs. Its disorganization is the source of approximately 90% of all human cancers. Elaboration of the basic epithelial characteristics has led to an understanding of how complex structures such as the branched tubular networks of vertebrate lung or invertebrate tracheal system are organized. Aside from obvious morphological differences, specific compositions of the epithelial extracellular matrix (ECM) have been noted. For example, while the flexible ECM of the vertebrate skin mainly consists of collagen and elastic fibers, the rigid ECM of invertebrates is chitin-based to serve as an inflexible exoskeleton. We show that a central regulator of ECM differentiation and epithelial development in vertebrates, the matriptase-prostasin proteolytic cascade (MPPC), is conserved and essential for both Drosophila ECM morphogenesis and physiology. The functionally conserved components of the MPPC mediate cleavage of zona pellucida-domain (ZP-domain) proteins, which play crucial roles in organizing apical structures of the ECM in both vertebrates and invertebrates. Our data indicate that ZP-proteins are molecular targets of the conserved MPPC and that cleavage within the ZP-domains is a conserved mechanism of ECM development and differentiation.
Collapse
Affiliation(s)
- Leonard Drees
- Research Group Molecular Organogenesis, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Tatiana Königsmann
- Research Group Molecular Organogenesis, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Martin H. J. Jaspers
- Research Group Molecular Organogenesis, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ralf Pflanz
- Research Group Mass Spectrometry, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Dietmar Riedel
- Electron Microscopy Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Reinhard Schuh
- Research Group Molecular Organogenesis, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
- * E-mail:
| |
Collapse
|
31
|
Danielsen ET, Olsen AK, Coskun M, Nonboe AW, Larsen S, Dahlgaard K, Bennett EP, Mitchelmore C, Vogel LK, Troelsen JT. Intestinal regulation of suppression of tumorigenicity 14 (ST14) and serine peptidase inhibitor, Kunitz type -1 (SPINT1) by transcription factor CDX2. Sci Rep 2018; 8:11813. [PMID: 30087389 PMCID: PMC6081401 DOI: 10.1038/s41598-018-30216-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
The type II membrane-anchored serine protease, matriptase, encoded by suppression of tumorgenicity-14 (ST14) regulates the integrity of the intestinal epithelial barrier in concert with its inhibitor, HAI-1 encoded by serine peptidase inhibitor, Kunitz type -1 (SPINT1). The balance of the protease/inhibitor gene expression ratio is vital in preventing the oncogenic potential of matriptase. The intestinal cell lineage is regulated by a transcriptional regulatory network where the tumor suppressor, Caudal homeobox 2 (CDX2) is considered to be an intestinal master transcription factor. In this study, we show that CDX2 has a dual function in regulating both ST14 and SPINT1, gene expression in intestinal cells. We find that CDX2 is not required for the basal ST14 and SPINT1 gene expression; however changes in CDX2 expression affects the ST14/SPINT1 mRNA ratio. Exploring CDX2 ChIP-seq data from intestinal cell lines, we identified genomic CDX2-enriched enhancer elements for both ST14 and SPINT1, which regulate their corresponding gene promoter activity. We show that CDX2 displays both repressive and enhancing regulatory abilities in a cell specific manner. Together, these data reveal new insight into transcriptional mechanisms controlling the intestinal matriptase/inhibitor balance.
Collapse
Affiliation(s)
- E Thomas Danielsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark.,Institute of Cellular and Molecular Medicine, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Anders Krüger Olsen
- Institute of Cellular and Molecular Medicine, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Mehmet Coskun
- Department of Gastroenterology, University of Copenhagen, DK-2730, Herlev, Denmark
| | - Annika W Nonboe
- Institute of Cellular and Molecular Medicine, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sylvester Larsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark.,Department of Clinical Immunology, Naestved Hospital, Naestved, Region Zealand, Denmark
| | - Katja Dahlgaard
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Eric Paul Bennett
- Copenhagen Center for Glycomics, Department of Odontology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cathy Mitchelmore
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Lotte Katrine Vogel
- Institute of Cellular and Molecular Medicine, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
32
|
Szabo R, Bugge TH. Loss of HAI-2 in mice with decreased prostasin activity leads to an early-onset intestinal failure resembling congenital tufting enteropathy. PLoS One 2018; 13:e0194660. [PMID: 29617460 PMCID: PMC5884512 DOI: 10.1371/journal.pone.0194660] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/07/2018] [Indexed: 01/15/2023] Open
Abstract
Prostasin (CAP1/PRSS8) is a glycosylphosphatidylinositol (GPI)-anchored serine protease that is essential for epithelial development and overall survival in mice. Prostasin is regulated primarily by the transmembrane serine protease inhibitor, hepatocyte growth factor activator inhibitor (HAI)-2, and loss of HAI-2 function leads to early embryonic lethality in mice due to an unregulated prostasin activity. We have recently reported that critical in vivo functions of prostasin can be performed by proteolytically-inactive or zymogen-locked variants of the protease. Here we show that the zymogen form of prostasin does not bind to HAI-2 and, as a result, loss of HAI-2 does not affect prenatal development and survival of mice expressing only zymogen-locked variant of prostasin (Prss8 R44Q). Indeed, HAI-2-deficient mice homozygous for R44Q mutation (Spint2-/-;Prss8R44Q/R44Q) are born in the expected numbers and do not exhibit any obvious developmental abnormality at birth. However, postnatal growth in these mice is severely impaired and they all die within 4 to 7 days after birth due to a critical failure in the development of small and large intestines, characterized by a widespread villous atrophy, tufted villi, near-complete loss of mucin-producing goblet cells, loss of colonic crypt structure, and bleeding into the intestinal lumen. Intestines of Spint2-/-;Prss8R44Q/R44Q mice showed altered expression of epithelial junctional proteins, including reduced levels of EpCAM, E-cadherin, occludin, claudin-1 and -7, as well as an increased level of claudin-4, indicating that the loss of HAI-2 compromises intestinal epithelial barrier function. Our data indicate that the loss of HAI-2 in Prss8R44Q/R44Q mice leads to development of progressive intestinal failure that at both histological and molecular level bears a striking resemblance to human congenital tufting enteropathy, and may provide important clues for understanding and treating this debilitating human disease.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (RS); (THB)
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (RS); (THB)
| |
Collapse
|
33
|
Schepis A, Barker A, Srinivasan Y, Balouch E, Zheng Y, Lam I, Clay H, Hsiao CD, Coughlin SR. Protease signaling regulates apical cell extrusion, cell contacts, and proliferation in epithelia. J Cell Biol 2018; 217:1097-1112. [PMID: 29301867 PMCID: PMC5839797 DOI: 10.1083/jcb.201709118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/27/2017] [Accepted: 12/07/2017] [Indexed: 11/22/2022] Open
Abstract
Mechanisms that sense and regulate epithelial morphogenesis, integrity, and homeostasis are incompletely understood. Protease-activated receptor 2 (Par2), the Par2-activating membrane-tethered protease matriptase, and its inhibitor, hepatocyte activator inhibitor 1 (Hai1), are coexpressed in most epithelia and may make up a local signaling system that regulates epithelial behavior. We explored the role of Par2b in matriptase-dependent skin abnormalities in Hai1a-deficient zebrafish embryos. We show an unexpected role for Par2b in regulation of epithelial apical cell extrusion, roles in regulating proliferation that were opposite in distinct but adjacent epithelial monolayers, and roles in regulating cell-cell junctions, mobility, survival, and expression of genes involved in tissue remodeling and inflammation. The epidermal growth factor receptor Erbb2 and matrix metalloproteinases, the latter induced by Par2b, may contribute to some matriptase- and Par2b-dependent phenotypes and be permissive for others. Our results suggest that local protease-activated receptor signaling can coordinate cell behaviors known to contribute to epithelial morphogenesis and homeostasis.
Collapse
Affiliation(s)
- Antonino Schepis
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Adrian Barker
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Yoga Srinivasan
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Eaman Balouch
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Yaowu Zheng
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Ian Lam
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Hilary Clay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taiwan
| | - Shaun R Coughlin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
34
|
Rosa JB, Metzstein MM, Ghabrial AS. An Ichor-dependent apical extracellular matrix regulates seamless tube shape and integrity. PLoS Genet 2018; 14:e1007146. [PMID: 29309404 PMCID: PMC5774827 DOI: 10.1371/journal.pgen.1007146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/19/2018] [Accepted: 12/09/2017] [Indexed: 01/25/2023] Open
Abstract
During sprouting angiogenesis in the vertebrate vascular system, and primary branching in the Drosophila tracheal system, specialized tip cells direct branch outgrowth and network formation. When tip cells lumenize, they form subcellular (seamless) tubes. How these seamless tubes are made, shaped and maintained remains poorly understood. Here we characterize a Drosophila mutant called ichor (ich), and show that ich is essential for the integrity and shape of seamless tubes in tracheal terminal cells. We find that Ich regulates seamless tubulogenesis via its role in promoting the formation of a mature apical extracellular matrix (aECM) lining the lumen of the seamless tubes. We determined that ich encodes a zinc finger protein (CG11966) that acts, as a transcriptional activator required for the expression of multiple aECM factors, including a novel membrane-anchored trypsin protease (CG8213). Thus, the integrity and shape of seamless tubes are regulated by the aECM that lines their lumens.
Collapse
Affiliation(s)
- Jeffrey B. Rosa
- Department of Cell & Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mark M. Metzstein
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - Amin S. Ghabrial
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
35
|
Tseng CC, Jia B, Barndt R, Gu Y, Chen CY, Tseng IC, Su SF, Wang JK, Johnson MD, Lin CY. Matriptase shedding is closely coupled with matriptase zymogen activation and requires de novo proteolytic cleavage likely involving its own activity. PLoS One 2017; 12:e0183507. [PMID: 28829816 PMCID: PMC5567652 DOI: 10.1371/journal.pone.0183507] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/04/2017] [Indexed: 11/18/2022] Open
Abstract
The type 2 transmembrane serine protease matriptase is involved in many pathophysiological processes probably via its enzymatic activity, which depends on the dynamic relationship between zymogen activation and protease inhibition. Matriptase shedding can prolong the life of enzymatically active matriptase and increase accessibility to substrates. We show here that matriptase shedding occurs via a de novo proteolytic cleavage at sites located between the SEA domain and the CUB domain. Point or combined mutations at the four positively charged amino acid residues in the region following the SEA domain allowed Arg-186 to be identified as the primary cleavage site responsible for matriptase shedding. Kinetic studies further demonstrate that matriptase shedding is temporally coupled with matriptase zymogen activation. The onset of matriptase shedding lags one minute behind matriptase zymogen activation. Studies with active site triad Ser-805 point mutated matriptase, which no longer undergoes zymogen activation or shedding, further suggests that matriptase shedding depends on matriptase zymogen activation, and that matriptase proteolytic activity may be involved in its own shedding. Our studies uncover an autonomous mechanism coupling matriptase zymogen activation, proteolytic activity, and shedding such that a proportion of newly generated active matriptase escapes HAI-1-mediated rapid inhibition by shedding into the extracellular milieu.
Collapse
Affiliation(s)
- Chun-Che Tseng
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Bailing Jia
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- Department of Gastroenterology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Robert Barndt
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Yayun Gu
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Chien-Yu Chen
- Department of Biochemistry National Defense Medical Center, Taipei, Taiwan
- School of Medicine National Defense Medical Center, Taipei, Taiwan
| | - I-Chu Tseng
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Sheng-Fang Su
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Jehng-Kang Wang
- Department of Biochemistry National Defense Medical Center, Taipei, Taiwan
| | - Michael D. Johnson
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- * E-mail: (CYL); (MDJ)
| | - Chen-Yong Lin
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- * E-mail: (CYL); (MDJ)
| |
Collapse
|
36
|
Murray AS, Varela FA, List K. Type II transmembrane serine proteases as potential targets for cancer therapy. Biol Chem 2017; 397:815-26. [PMID: 27078673 DOI: 10.1515/hsz-2016-0131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022]
Abstract
Carcinogenesis is accompanied by increased protein and activity levels of extracellular cell-surface proteases that are capable of modifying the tumor microenvironment by directly cleaving the extracellular matrix, as well as activating growth factors and proinflammatory mediators involved in proliferation and invasion of cancer cells, and recruitment of inflammatory cells. These complex processes ultimately potentiate neoplastic progression leading to local tumor cell invasion, entry into the vasculature, and metastasis to distal sites. Several members of the type II transmembrane serine protease (TTSP) family have been shown to play critical roles in cancer progression. In this review the knowledge collected over the past two decades about the molecular mechanisms underlying the pro-cancerous properties of selected TTSPs will be summarized. Furthermore, we will discuss how these insights may facilitate the translation into clinical settings in the future by specifically targeting TTSPs as part of novel cancer treatment regimens.
Collapse
|
37
|
Lahey KA, Ronaghan NJ, Shang J, Dion SP, Désilets A, Leduc R, MacNaughton WK. Signaling pathways induced by serine proteases to increase intestinal epithelial barrier function. PLoS One 2017; 12:e0180259. [PMID: 28671992 PMCID: PMC5495298 DOI: 10.1371/journal.pone.0180259] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/13/2017] [Indexed: 12/30/2022] Open
Abstract
Changes in barrier function of the gastrointestinal tract are thought to contribute to the inflammatory bowel diseases Crohn's disease and ulcerative colitis. Previous work in our lab demonstrated that apical exposure of intestinal epithelial cell lines to serine proteases results in an increase in transepithelial electrical resistance (TER). However, the underlying mechanisms governing this response are unclear. We aimed to determine the requirement for proteolytic activity, epidermal growth factor receptor (EGFR) activation, and downstream intracellular signaling in initiating and maintaining enhanced barrier function following protease treatment using a canine intestinal epithelial cell line (SCBN). We also examined the role of phosphorylation of myosin regulatory light chain on the serine protease-induced increase in TER through. It was found that proteolytic activity of the serine proteases trypsin and matriptase is required to initiate and maintain the protease-mediated increase in TER. We also show that MMP-independent EGFR activation is essential to the sustained phase of the protease response, and that Src kinases may mediate EGFR transactivation. PI3-K and ERK1/2 signaling were important in reaching a maximal increase in TER following protease stimulation; however, their upstream activators are yet to be determined. CK2 inhibition prevented the increase in TER induced by serine proteases. The bradykinin B(2) receptor was not involved in the change in TER in response to serine proteases, and no change in phosphorylation of MLC was observed after trypsin or matriptase treatment. Taken together, our data show a requirement for ongoing proteolytic activity, EGFR transactivation, as well as downstream PI3-K, ERK1/2, and CK2 signaling in protease-mediated barrier enhancement of intestinal epithelial cells. The pathways mediating enhanced barrier function by proteases may be novel therapeutic targets for intestinal disorders characterized by disrupted epithelial barrier function.
Collapse
Affiliation(s)
- Kelcie A. Lahey
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Natalie J. Ronaghan
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Judie Shang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Sébastien P. Dion
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Leduc
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Wallace K. MacNaughton
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Friis S, Tadeo D, Le-Gall SM, Jürgensen HJ, Sales KU, Camerer E, Bugge TH. Matriptase zymogen supports epithelial development, homeostasis and regeneration. BMC Biol 2017; 15:46. [PMID: 28571576 PMCID: PMC5452369 DOI: 10.1186/s12915-017-0384-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/05/2017] [Indexed: 12/31/2022] Open
Abstract
Background Matriptase is a membrane serine protease essential for epithelial development, homeostasis, and regeneration, as well as a central orchestrator of pathogenic pericellular signaling in the context of inflammatory and proliferative diseases. Matriptase is an unusual protease in that its zymogen displays measurable enzymatic activity. Results Here, we used gain and loss of function genetics to investigate the possible biological functions of zymogen matriptase. Unexpectedly, transgenic mice mis-expressing a zymogen-locked version of matriptase in the epidermis displayed pathologies previously reported for transgenic mice mis-expressing wildtype epidermal matriptase. Equally surprising, mice engineered to express only zymogen-locked endogenous matriptase, unlike matriptase null mice, were viable, developed epithelial barrier function, and regenerated the injured epithelium. Compatible with these observations, wildtype and zymogen-locked matriptase were equipotent activators of PAR-2 inflammatory signaling. Conclusion The study demonstrates that the matriptase zymogen is biologically active and is capable of executing developmental and homeostatic functions of the protease. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0384-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stine Friis
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Section for Molecular Disease Biology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Tadeo
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Georgetown University School of Medicine, Washington, DC, 20057, USA
| | - Sylvain M Le-Gall
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Université Sorbonne Paris Cité, Paris, France
| | - Henrik Jessen Jürgensen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA
| | - Katiuchia Uzzun Sales
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Department of Cell and Molecular Biology, Ribierão Preto School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Université Sorbonne Paris Cité, Paris, France
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Buzza MS, Johnson TA, Conway GD, Martin EW, Mukhopadhyay S, Shea-Donohue T, Antalis TM. Inflammatory cytokines down-regulate the barrier-protective prostasin-matriptase proteolytic cascade early in experimental colitis. J Biol Chem 2017; 292:10801-10812. [PMID: 28490634 DOI: 10.1074/jbc.m116.771469] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/02/2017] [Indexed: 12/17/2022] Open
Abstract
Compromised gastrointestinal barrier function is strongly associated with the progressive and destructive pathologies of the two main forms of irritable bowel disease (IBD), ulcerative colitis (UC), and Crohn's disease (CD). Matriptase is a membrane-anchored serine protease encoded by suppression of tumorigenicity-14 (ST14) gene, which is critical for epithelial barrier development and homeostasis. Matriptase barrier-protective activity is linked with the glycosylphosphatidylinositol (GPI)-anchored serine protease prostasin, which is a co-factor for matriptase zymogen activation. Here we show that mRNA and protein expression of both matriptase and prostasin are rapidly down-regulated in the initiating inflammatory phases of dextran sulfate sodium (DSS)-induced experimental colitis in mice, and, significantly, the loss of these proteases precedes the appearance of clinical symptoms, suggesting their loss may contribute to disease susceptibility. We used heterozygous St14 hypomorphic mice expressing a promoter-linked β-gal reporter to show that inflammatory colitis suppresses the activity of the St14 gene promoter. Studies in colonic T84 cell monolayers revealed that barrier disruption by the colitis-associated Th2-type cytokines, IL-4 and IL-13, down-regulates matriptase as well as prostasin through phosphorylation of the transcriptional regulator STAT6 and that inhibition of STAT6 with suberoylanilide hydroxamic acid (SAHA) restores protease expression and reverses cytokine-induced barrier dysfunction. Both matriptase and prostasin are significantly down-regulated in colonic tissues from human subjects with active ulcerative colitis or Crohn's disease, implicating the loss of this barrier-protective protease pathway in the pathogenesis of irritable bowel disease.
Collapse
Affiliation(s)
- Marguerite S Buzza
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Tierra A Johnson
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Gregory D Conway
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Erik W Martin
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | | | - Terez Shea-Donohue
- the Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Toni M Antalis
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| |
Collapse
|
40
|
Nonboe AW, Krigslund O, Soendergaard C, Skovbjerg S, Friis S, Andersen MN, Ellis V, Kawaguchi M, Kataoka H, Bugge TH, Vogel LK. HAI-2 stabilizes, inhibits and regulates SEA-cleavage-dependent secretory transport of matriptase. Traffic 2017; 18:378-391. [PMID: 28371047 DOI: 10.1111/tra.12482] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 11/28/2022]
Abstract
It has recently been shown that hepatocyte growth factor activator inhibitor-2 (HAI-2) is able to suppress carcinogenesis induced by overexpression of matriptase, as well as cause regression of individual established tumors in a mouse model system. However, the role of HAI-2 is poorly understood. In this study, we describe 3 mutations in the binding loop of the HAI-2 Kunitz domain 1 (K42N, C47F and R48L) that cause a delay in the SEA domain cleavage of matriptase, leading to accumulation of non-SEA domain cleaved matriptase in the endoplasmic reticulum (ER). We suggest that, like other known SEA domains, the matriptase SEA domain auto-cleaves and reflects that correct oligomerization, maturation, and/or folding has been obtained. Our results suggest that the HAI-2 Kunitz domain 1 mutants influence the flux of matriptase to the plasma membrane by affecting the oligomerization, maturation and/or folding of matriptase, and as a result the SEA domain cleavage of matriptase. Two of the HAI-2 Kunitz domain 1 mutants investigated (C47F, R48L and C47F/R48L) also displayed a reduced ability to proteolytically silence matriptase. Hence, HAI-2 separately stabilizes matriptase, regulates the secretory transport, possibly via maturation/oligomerization and inhibits the proteolytic activity of matriptase in the ER, and possible throughout the secretory pathway.
Collapse
Affiliation(s)
- Annika W Nonboe
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark
| | - Oliver Krigslund
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark
| | - Christoffer Soendergaard
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark.,Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Signe Skovbjerg
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark
| | - Stine Friis
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark.,Department of Molecular Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen East, Denmark
| | - Martin N Andersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Vincent Ellis
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Lotte K Vogel
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen North, Denmark
| |
Collapse
|
41
|
Van Spaendonk H, Ceuleers H, Witters L, Patteet E, Joossens J, Augustyns K, Lambeir AM, De Meester I, De Man JG, De Winter BY. Regulation of intestinal permeability: The role of proteases. World J Gastroenterol 2017; 23:2106-2123. [PMID: 28405139 PMCID: PMC5374123 DOI: 10.3748/wjg.v23.i12.2106] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/20/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal barrier is - with approximately 400 m2 - the human body's largest surface separating the external environment from the internal milieu. This barrier serves a dual function: permitting the absorption of nutrients, water and electrolytes on the one hand, while limiting host contact with noxious luminal antigens on the other hand. To maintain this selective barrier, junction protein complexes seal the intercellular space between adjacent epithelial cells and regulate the paracellular transport. Increased intestinal permeability is associated with and suggested as a player in the pathophysiology of various gastrointestinal and extra-intestinal diseases such as inflammatory bowel disease, celiac disease and type 1 diabetes. The gastrointestinal tract is exposed to high levels of endogenous and exogenous proteases, both in the lumen and in the mucosa. There is increasing evidence to suggest that a dysregulation of the protease/antiprotease balance in the gut contributes to epithelial damage and increased permeability. Excessive proteolysis leads to direct cleavage of intercellular junction proteins, or to opening of the junction proteins via activation of protease activated receptors. In addition, proteases regulate the activity and availability of cytokines and growth factors, which are also known modulators of intestinal permeability. This review aims at outlining the mechanisms by which proteases alter the intestinal permeability. More knowledge on the role of proteases in mucosal homeostasis and gastrointestinal barrier function will definitely contribute to the identification of new therapeutic targets for permeability-related diseases.
Collapse
|
42
|
Modulating the selectivity of matriptase-2 inhibitors with unnatural amino acids. Eur J Med Chem 2017; 129:110-123. [DOI: 10.1016/j.ejmech.2017.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/19/2017] [Accepted: 02/05/2017] [Indexed: 12/12/2022]
|
43
|
Wu CJ, Feng X, Lu M, Morimura S, Udey MC. Matriptase-mediated cleavage of EpCAM destabilizes claudins and dysregulates intestinal epithelial homeostasis. J Clin Invest 2017; 127:623-634. [PMID: 28094766 DOI: 10.1172/jci88428] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Congenital tufting enteropathy (CTE) is a severe autosomal recessive human diarrheal disorder with characteristic intestinal epithelial dysplasia. CTE can be caused by mutations in genes encoding EpCAM, a putative adhesion molecule, and HAI-2, a cell surface protease inhibitor. A similar phenotype occurs in mice whose intestinal epithelial cells (IECs) fail to express the tight junction-associated protein claudin-7. EpCAM stabilizes claudin-7 in IECs, and HAI-2 regulates the cell surface serine protease matriptase, a known modifier of intestinal epithelial physiology. Therefore, we hypothesized that HAI-2, matriptase, EpCAM, and claudin-7 were functionally linked. Herein we have demonstrated that active matriptase cleaves EpCAM after Arg80 and that loss of HAI-2 in IECs led to unrestrained matriptase activity and efficient cleavage of EpCAM. Cleavage of EpCAM decreased its ability to associate with claudin-7 and targeted it for internalization and lysosomal degradation in conjunction with claudin-7. CTE-associated HAI-2 mutant proteins exhibited reduced ability to inhibit matriptase and also failed to efficiently stabilize claudin-7 in IECs. These results identify EpCAM as a substrate of matriptase and link HAI-2, matriptase, EpCAM, and claudin-7 in a functionally important pathway that causes disease when it is dysregulated.
Collapse
|
44
|
Tanabe LM, List K. The role of type II transmembrane serine protease-mediated signaling in cancer. FEBS J 2016; 284:1421-1436. [PMID: 27870503 DOI: 10.1111/febs.13971] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/29/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
Pericellular proteases have long been implicated in carcinogenesis. Previous research focused on these proteins, primarily as extracellular matrix (ECM) protein-degrading enzymes which allowed cancer cells to breach the basement membrane and invade surrounding tissue. However, recently, there has been a shift in the view of cell surface proteases, including serine proteases, as proteolytic modifiers of particular targets, including growth factors and protease-activated receptors, which are critical for the activation of oncogenic signaling pathways. Of the 176 human serine proteases currently identified, a subset of 17, known as type II transmembrane serine proteases (TTSPs). Many have been shown to be relevant to cancer progression since they were first identified as a family around the turn of the century. To this end, altered expression of TTSPs appeared as a trademark of several tumor types. However, the substrates and underlying signaling pathways remained unclear. Localization of these proteins to the cell surface places them in the unique position to mediate signal transduction between the cell and its surrounding environment. Many of the TTSPs have already been shown to play key roles in processes such as postnatal development, tissue homeostasis, and tumor progression, which share overlapping molecular mechanisms. In this review, we summarize the current knowledge regarding the role of the TTSP family in pro-oncogenic signaling.
Collapse
Affiliation(s)
- Lauren M Tanabe
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Karin List
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
45
|
Antalis TM, Conway GD, Peroutka RJ, Buzza MS. Membrane-anchored proteases in endothelial cell biology. Curr Opin Hematol 2016; 23:243-52. [PMID: 26906027 DOI: 10.1097/moh.0000000000000238] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW The endothelial cell plasma membrane is a metabolically active, dynamic, and fluid microenvironment where pericellular proteolysis plays a critical role. Membrane-anchored proteases may be expressed by endothelial cells as well as mural cells and leukocytes with distribution both inside and outside of the vascular system. Here, we will review the recent advances in our understanding of the direct and indirect roles of membrane-anchored proteases in vascular biology and the possible conservation of their extravascular functions in endothelial cell biology. RECENT FINDINGS Membrane-anchored proteases belonging to the serine or metalloprotease families contain amino-terminal or carboxy-terminal domains, which serve to tether their extracellular protease domains directly at the plasma membrane. This architecture enables protease function and substrate repertoire to be regulated through dynamic localization in distinct areas of the cell membrane. These proteases are proving to be key components of the cell machinery for regulating vascular permeability, generation of vasoactive peptides, receptor tyrosine kinase transactivation, extracellular matrix proteolysis, and angiogenesis. SUMMARY A complex picture of the interdependence between membrane-anchored protease localization and function is emerging that may provide a mechanism for precise coordination of extracellular signals and intracellular responses through communication with the cytoskeleton and with cellular signaling molecules.
Collapse
Affiliation(s)
- Toni M Antalis
- Center for Vascular and Inflammatory Diseases and the Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
46
|
Altered Prostasin (CAP1/Prss8) Expression Favors Inflammation and Tissue Remodeling in DSS-induced Colitis. Inflamm Bowel Dis 2016; 22:2824-2839. [PMID: 27755216 DOI: 10.1097/mib.0000000000000940] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) including ulcerative colitis and Crohn's disease are diseases with impaired epithelial barrier function. We aimed to investigate whether mutated prostasin and thus, reduced colonic epithelial sodium channel activity predisposes to develop an experimentally dextran sodium sulfate (DSS)-induced colitis. METHODS Wildtype, heterozygous (fr/+), and homozygous (fr/fr) prostasin-mutant rats were treated 7 days with DSS followed by 7 days of recovery and analyzed with respect to histology, clinicopathological parameters, inflammatory marker mRNA transcript expression, and sodium transporter protein expression. RESULTS In this study, a more detailed analysis on rat fr/fr colons revealed reduced numbers of crypt and goblet cells, and local angiodysplasia, as compared with heterozygous (fr/+) and wildtype littermates. Following 2% DSS treatment for 7 days followed by 7 days recovery, fr/fr animals lost body weight, and reached maximal diarrhea score and highest disease activity after only 3 days, and strongly increased cytokine levels. The histology score significantly increased in all groups, but fr/fr colons further displayed pronounced histological alterations with near absence of goblet cells, rearrangement of the lamina propria, and presence of neutrophils, eosinophils, and macrophages. Additionally, fr/fr colons showed ulcerations and edemas that were absent in fr/+ and wildtype littermates. Following recovery, fr/fr rats reached, although significantly delayed, near-normal diarrhea score and disease activity, but exhibited severe architectural remodeling, despite unchanged sodium transporter protein expression. CONCLUSIONS In summary, our results demonstrate a protective role of colonic prostasin expression against experimental colitis, and thus represent a susceptibility gene in the development of inflammatory bowel disease.
Collapse
|
47
|
Ronaghan NJ, Shang J, Iablokov V, Zaheer R, Colarusso P, Dion S, Désilets A, Leduc R, Turner JR, MacNaughton WK. The serine protease-mediated increase in intestinal epithelial barrier function is dependent on occludin and requires an intact tight junction. Am J Physiol Gastrointest Liver Physiol 2016; 311:G466-79. [PMID: 27492333 PMCID: PMC5076006 DOI: 10.1152/ajpgi.00441.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/28/2016] [Indexed: 01/31/2023]
Abstract
Barrier dysfunction is a characteristic of the inflammatory bowel diseases (IBD), Crohn's disease and ulcerative colitis. Understanding how the tight junction is modified to maintain barrier function may provide avenues for treatment of IBD. We have previously shown that the apical addition of serine proteases to intestinal epithelial cell lines causes a rapid and sustained increase in transepithelial electrical resistance (TER), but the mechanisms are unknown. We hypothesized that serine proteases increase barrier function through trafficking and insertion of tight junction proteins into the membrane, and this could enhance recovery of a disrupted monolayer after calcium switch or cytokine treatment. In the canine epithelial cell line, SCBN, we showed that matriptase, an endogenous serine protease, could potently increase TER. Using detergent solubility-based cell fractionation, we found that neither trypsin nor matriptase treatment changed levels of tight junction proteins at the membrane. In a fast calcium switch assay, serine proteases did not enhance the rate of recovery of the junction. In addition, serine proteases could not reverse barrier disruption induced by IFNγ and TNFα. We knocked down occludin in our cells using siRNA and found this prevented the serine protease-induced increase in TER. Using fluorescence recovery after photobleaching (FRAP), we found serine proteases induce a greater mobile fraction of occludin in the membrane. These data suggest that a functional tight junction is needed for serine proteases to have an effect on TER, and that occludin is a crucial tight junction protein in this mechanism.
Collapse
Affiliation(s)
- Natalie J. Ronaghan
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| | - Judie Shang
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| | - Vadim Iablokov
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| | - Raza Zaheer
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| | - Pina Colarusso
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| | - Sébastien Dion
- 2Département de Pharmacologie-Physiologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - Antoine Désilets
- 2Département de Pharmacologie-Physiologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - Richard Leduc
- 2Département de Pharmacologie-Physiologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - Jerrold R. Turner
- 3Departments of Pathology and Medicine (GI), Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wallace K. MacNaughton
- 1Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| |
Collapse
|
48
|
Szabo R, Lantsman T, Peters DE, Bugge TH. Delineation of proteolytic and non-proteolytic functions of the membrane-anchored serine protease prostasin. Development 2016; 143:2818-28. [PMID: 27385010 DOI: 10.1242/dev.137968] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/14/2016] [Indexed: 11/20/2022]
Abstract
The membrane-anchored serine proteases prostasin (PRSS8) and matriptase (ST14) initiate a cell surface proteolytic pathway essential for epithelial function. Mice expressing only catalytically inactive prostasin are viable, unlike prostasin null mice, indicating that at least some prostasin functions are non-proteolytic. Here we used knock-in mice expressing catalytically inactive prostasin (Prss8(Ki/Ki)) to show that the physiological and pathological functions of prostasin vary in their dependence on its catalytic activity. Whereas prostasin null mice exhibited partial embryonic and complete perinatal lethality, Prss8(Ki/Ki) mice displayed normal prenatal and postnatal survival. Unexpectedly, catalytically inactive prostasin caused embryonic lethality in mice lacking its cognate inhibitors HAI-1 (SPINT1) or HAI-2 (SPINT2). Proteolytically inactive prostasin, unlike the wild-type protease, was unable to activate matriptase during placentation. Surprisingly, all essential functions of prostasin in embryonic and postnatal development were compensated for by loss of HAI-1, indicating that prostasin is only required for mouse development and overall viability in the presence of this inhibitor. This study expands our knowledge of non-proteolytic functions of membrane-anchored serine proteases and provides unexpected new data on the mechanistic interactions between matriptase and prostasin in the context of epithelial development.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Taliya Lantsman
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diane E Peters
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA Program of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA 02110, USA
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Antalis TM. Coagulation signaling to epithelia. Blood 2016; 127:3114-6. [PMID: 27340252 PMCID: PMC4920017 DOI: 10.1182/blood-2016-05-715052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
50
|
Chen YW, Yin S, Lai YJJ, Johnson MD, Lin CY. Plasminogen-Dependent Matriptase Activation Accelerates Plasmin Generation by Differentiating Primary Human Keratinocytes. J Invest Dermatol 2016; 136:1210-1218. [DOI: 10.1016/j.jid.2016.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 01/19/2016] [Accepted: 01/25/2016] [Indexed: 11/25/2022]
|