1
|
Petrova E, López-Gay JM, Fahrner M, Leturcq F, de Villartay JP, Barbieux C, Gonschorek P, Tsoi LC, Gudjonsson JE, Schilling O, Hovnanian A. Comparative analyses of Netherton syndrome patients and Spink5 conditional knock-out mice uncover disease-relevant pathways. Commun Biol 2024; 7:152. [PMID: 38316920 PMCID: PMC10844249 DOI: 10.1038/s42003-024-05780-y] [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: 03/07/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Netherton syndrome (NS) is a rare skin disease caused by loss-of-function mutations in the serine peptidase inhibitor Kazal type 5 (SPINK5) gene. Disease severity and the lack of efficacious treatments call for a better understanding of NS mechanisms. Here we describe a novel and viable, Spink5 conditional knock-out (cKO) mouse model, allowing to study NS progression. By combining transcriptomics and proteomics, we determine a disease molecular profile common to mouse models and NS patients. Spink5 cKO mice and NS patients share skin barrier and inflammation signatures defined by up-regulation and increased activity of proteases, IL-17, IL-36, and IL-20 family cytokine signaling. Systemic inflammation in Spink5 cKO mice correlates with disease severity and is associated with thymic atrophy and enlargement of lymph nodes and spleen. This systemic inflammation phenotype is marked by neutrophils and IL-17/IL-22 signaling, does not involve primary T cell immunodeficiency and is independent of bacterial infection. By comparing skin transcriptomes and proteomes, we uncover several putative substrates of tissue kallikrein-related proteases (KLKs), demonstrating that KLKs can proteolytically regulate IL-36 pro-inflammatory cytokines. Our study thus provides a conserved molecular framework for NS and reveals a KLK/IL-36 signaling axis, adding new insights into the disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Evgeniya Petrova
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and University of Paris, Paris, France.
| | - Jesús María López-Gay
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Paris, F-75248, Cedex 05, France
- Sorbonne University, UPMC University Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005, Paris, France
| | - Matthias Fahrner
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| | - Florent Leturcq
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and University of Paris, Paris, France
| | - Jean-Pierre de Villartay
- Imagine Institute, Laboratory "Genome Dynamics in the Immune System", INSERM UMR 11635, Paris, France
| | - Claire Barbieux
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and University of Paris, Paris, France
| | - Patrick Gonschorek
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| | - Alain Hovnanian
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and University of Paris, Paris, France.
- Department of Genomic Medicine of rare diseases, Necker Hospital for Sick Children, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France.
- University of Paris Cité, Paris, France.
| |
Collapse
|
2
|
Lysophosphatidylserine Induces MUC5AC Production via the Feedforward Regulation of the TACE-EGFR-ERK Pathway in Airway Epithelial Cells in a Receptor-Independent Manner. Int J Mol Sci 2022; 23:ijms23073866. [PMID: 35409225 PMCID: PMC8999057 DOI: 10.3390/ijms23073866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 02/05/2023] Open
Abstract
Lysophosphatidylserine (LysoPS) is an amphipathic lysophospholipid that mediates a broad spectrum of inflammatory responses through a poorly characterized mechanism. Because LysoPS levels can rise in a variety of pathological conditions, we sought to investigate LysoPS's potential role in airway epithelial cells that actively participate in lung homeostasis. Here, we report a previously unappreciated function of LysoPS in production of a mucin component, MUC5AC, in the airway epithelial cells. LysoPS stimulated lung epithelial cells to produce MUC5AC via signaling pathways involving TACE, EGFR, and ERK. Specifically, LysoPS- dependent biphasic activation of ERK resulted in TGF-α secretion and strong EGFR phosphorylation leading to MUC5AC production. Collectively, LysoPS induces the expression of MUC5AC via a feedback loop composed of proligand synthesis and its proteolysis by TACE and following autocrine EGFR activation. To our surprise, we were not able to find a role of GPCRs and TLR2, known LyoPS receptors in LysoPS-induced MUC5AC production in airway epithelial cells, suggesting a potential receptor-independent action of LysoPS during inflammation. This study provides new insight into the potential function and mechanism of LysoPS as an emerging lipid mediator in airway inflammation.
Collapse
|
3
|
Fuentes-Prior P. Priming of SARS-CoV-2 S protein by several membrane-bound serine proteinases could explain enhanced viral infectivity and systemic COVID-19 infection. J Biol Chem 2020; 296:100135. [PMID: 33268377 PMCID: PMC7834812 DOI: 10.1074/jbc.rev120.015980] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
The ongoing COVID-19 pandemic has already caused over a million deaths worldwide, and this death toll will be much higher before effective treatments and vaccines are available. The causative agent of the disease, the coronavirus SARS-CoV-2, shows important similarities with the previously emerged SARS-CoV-1, but also striking differences. First, SARS-CoV-2 possesses a significantly higher transmission rate and infectivity than SARS-CoV-1 and has infected in a few months over 60 million people. Moreover, COVID-19 has a systemic character, as in addition to the lungs, it also affects the heart, liver, and kidneys among other organs of the patients and causes frequent thrombotic and neurological complications. In fact, the term "viral sepsis" has been recently coined to describe the clinical observations. Here I review current structure-function information on the viral spike proteins and the membrane fusion process to provide plausible explanations for these observations. I hypothesize that several membrane-associated serine proteinases (MASPs), in synergy with or in place of TMPRSS2, contribute to activate the SARS-CoV-2 spike protein. Relative concentrations of the attachment receptor, ACE2, MASPs, their endogenous inhibitors (the Kunitz-type transmembrane inhibitors, HAI-1/SPINT1 and HAI-2/SPINT2, as well as major circulating serpins) would determine the infection rate of host cells. The exclusive or predominant expression of major MASPs in specific human organs suggests a direct role of these proteinases in e.g., heart infection and myocardial injury, liver dysfunction, kidney damage, as well as neurological complications. Thorough consideration of these factors could have a positive impact on the control of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Pablo Fuentes-Prior
- Molecular Bases of Disease, Biomedical Research Institute (IIB) Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| |
Collapse
|
4
|
Flores-Sanchez F, Chavez-Dueñas L, Sanchez-Villamil J, Navarro-Garcia F. Pic Protein From Enteroaggregative E. coli Induces Different Mechanisms for Its Dual Activity as a Mucus Secretagogue and a Mucinase. Front Immunol 2020; 11:564953. [PMID: 33281812 PMCID: PMC7705071 DOI: 10.3389/fimmu.2020.564953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
A hallmark of enteroaggregative Escherichia coli (EAEC) infection is the formation of an intestinal biofilm, which comprises a mucus layer with immersed bacteria. Pic is an autotransporter secreted by EAEC, and other E. coli pathotypes, and has been involved in two apparently contradictory phenotypes, as a mucus secretagogue and as a mucinase. Here, we investigated this Pic dual activity, mucus secretagogue capability and mucinolytic activity, in human goblet cells that secrete MUC2 and MUC5AC. Pic induced mucus hypersecretion directly in the goblet cells, without other intestinal cell types involved. At the same time, Pic exhibited strong proteolytic activity on the secreted mucins. These activities were independent since a mutation in the serine protease motif (PicS258I) abolished mucin degradation while maintaining the mucus secretagogue activity intact. Furthermore, deoxycholic acid (DCA)-induced mucins were proteolytically degraded when goblet cells were co-incubated with DCA/Pic, while co-incubation with DCA/PicS258I induced a synergistic effect on mucus hypersecretion. Pic was more efficient degrading MUC5AC than MUC2, but no degradation was detected with Pic inactivated at the active site by mutation or pharmacological inhibition. Remarkably, Pic cleaved MUC2 and MUC5AC in the C-terminal domain, leaving N-terminal subproducts, impacting the feature of gel-forming mucins and allowing mucus layer penetration by EAEC. Astonishingly, Pic stimulated rapid mucin secretion in goblet-like cells by activating the intracellular calcium pathway resulting from the PLC signal transduction pathway, leading to the production of DAG and releasing IP3, a second messenger of calcium signaling. Therefore, the dual activity of Pic, as a mucus secretagogue and a mucinase, is relevant in the context of carbon source generation and mucus layer penetration, allowing EAEC to live within the layer of mucus but also access epithelial cells.
Collapse
Affiliation(s)
- Fernando Flores-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Lucia Chavez-Dueñas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Javier Sanchez-Villamil
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| |
Collapse
|
5
|
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread worldwide since its first incidence in Wuhan, China, in December 2019. Although the case fatality rate of COVID-19 appears to be lower than that of SARS and Middle East respiratory syndrome (MERS), the higher transmissibility of SARS-CoV-2 has caused the total fatality to surpass other viral diseases, reaching more than 1 million globally as of October 6, 2020. The rate at which the disease is spreading calls for a therapy that is useful for treating a large population. Multiple intersecting viral and host factor targets involved in the life cycle of the virus are being explored. Because of the frequent mutations, many coronaviruses gain zoonotic potential, which is dependent on the presence of cell receptors and proteases, and therefore the targeting of the viral proteins has some drawbacks, as strain-specific drug resistance can occur. Moreover, the limited number of proteins in a virus makes the number of available targets small. Although SARS-CoV and SARS-CoV-2 share common mechanisms of entry and replication, there are substantial differences in viral proteins such as the spike (S) protein. In contrast, targeting cellular factors may result in a broader range of therapies, reducing the chances of developing drug resistance. In this Review, we discuss the role of primary host factors such as the cell receptor angiotensin-converting enzyme 2 (ACE2), cellular proteases of S protein priming, post-translational modifiers, kinases, inflammatory cells, and their pharmacological intervention in the infection of SARS-CoV-2 and related viruses.
Collapse
Affiliation(s)
- Anil Mathew Tharappel
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Subodh Kumar Samrat
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Zhong Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY 12201, USA
| |
Collapse
|
6
|
Callies LK, Tadeo D, Simper J, Bugge TH, Szabo R. Iterative, multiplexed CRISPR-mediated gene editing for functional analysis of complex protease gene clusters. J Biol Chem 2019; 294:15987-15996. [PMID: 31501243 DOI: 10.1074/jbc.ra119.009773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Elucidation of gene function by reverse genetics in animal models frequently is complicated by the functional redundancy of homologous genes. This obstacle often is compounded by the tight clustering of homologous genes, which precludes the generation of multigene-deficient animals through standard interbreeding of single-deficient animals. Here, we describe an iterative, multiplexed CRISPR-based approach for simultaneous gene editing in the complex seven-member human airway trypsin-like protease/differentially expressed in a squamous cell carcinoma (HAT/DESC) cluster of membrane-anchored serine proteases. Through four cycles of targeting, we generated a library of 18 unique congenic mouse strains lacking combinations of HAT/DESC proteases, including a mouse strain deficient in all seven proteases. Using this library, we demonstrate that HAT/DESC proteases are dispensable for term development, postnatal health, and fertility and that the recently described function of the HAT-like 4 protease in epidermal barrier formation is unique among all HAT/DESC proteases. The study demonstrates the potential of iterative, multiplexed CRISPR-mediated gene editing for functional analysis of multigene clusters, and it provides a large array of new congenic mouse strains for the study of HAT/DESC proteases in physiological and in pathophysiological processes.
Collapse
Affiliation(s)
- LuLu K Callies
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel Tadeo
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Jan Simper
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Roman Szabo
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
7
|
Miki M, Yasuoka S, Tsutsumi R, Nakamura Y, Hajime M, Takeuchi Y, Miki K, Kitada S, Maekura R. Human airway trypsin-like protease enhances interleukin-8 synthesis in bronchial epithelial cells by activating protease-activated receptor 2. Arch Biochem Biophys 2019; 664:167-173. [PMID: 30677406 DOI: 10.1016/j.abb.2019.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/31/2018] [Accepted: 01/16/2019] [Indexed: 11/16/2022]
Abstract
Human airway trypsin-like protease (HAT) localizes at human bronchial epithelial cells (HBECs). HAT enhanced release of interleukin-8 (IL-8) from HBECs at 10-100 mU/mL and the enhanced release was almost completely abolished by 50 μM leupeptin, a serine protease inhibitor. Previous reports suggested that HAT displays its physiological functions via protease-activated receptor 2 (PAR2). In the present study, we examined the mechanism whereby HAT upregulates IL-8 synthesis in HBECs with a focus on PAR2. Northern blot analysis revealed that HAT enhanced IL-8 mRNA expression at concentrations of 10-100 mU/mL. PAR2 activating peptide (PAR2 AP) also enhanced IL-8 release and IL-8 mRNA expression in HBECs at 50-1,000 μM at similar levels as HAT. Knockdown of PAR2 mRNA by siRNA methods showed that PAR2 mRNA expression was significantly depressed in primary HBECs, and both HAT- and PAR2 AP-induced IL-8 mRNA elevation was significantly depressed in PAR2 siRNA-transfected HBECs. Additionally, HAT cleaved the PAR2 activating site (R36-S37 bond) of synthetic PAR2 N-terminal peptide. These results indicate that HAT stimulates IL-8 synthesis in airway epithelial cells via PAR2 and could help to amplify inflammation in chronic respiratory tract disease.
Collapse
Affiliation(s)
- Mari Miki
- Department of Respiratory Medicine, National Hospital Organization Toneyama National Hospital, Osaka, Japan.
| | - Susumu Yasuoka
- Department of Nutrition and Metabolism, University of Tokushima School of Medicine, Tokushima, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, University of Tokushima School of Medicine, Tokushima, Japan
| | - Yoichi Nakamura
- Medical Center for Allergic and Immune Diseases, Yokohama City Minato Red Cross Hospital, Kanagawa, Japan
| | - Maeda Hajime
- Department of Thoracic Surgery, National Hospital Organization Toneyama National Hospital, Osaka, Japan
| | - Yukiyasu Takeuchi
- Department of Thoracic Surgery, National Hospital Organization Toneyama National Hospital, Osaka, Japan
| | - Keisuke Miki
- Department of Respiratory Medicine, National Hospital Organization Toneyama National Hospital, Osaka, Japan
| | - Seigo Kitada
- Department of Respiratory Medicine, National Hospital Organization Toneyama National Hospital, Osaka, Japan
| | - Ryoji Maekura
- Graduate School of Health Care Sciences, Jikei Institute, Osaka, Japan
| |
Collapse
|
8
|
Muytjens CMJ, Yu Y, Diamandis EP. Functional proteomic profiling reveals KLK13 and TMPRSS11D as active proteases in the lower female reproductive tract. F1000Res 2019; 7:1666. [PMID: 30647911 PMCID: PMC6329257 DOI: 10.12688/f1000research.16255.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2018] [Indexed: 11/20/2022] Open
Abstract
Background: Cervical-vaginal fluid (CVF) hydrates the mucosa of the lower female reproductive tract and is known to contain numerous proteases. The low pH of CVF (4.5 or below in healthy women of reproductive age) is a uniquely human attribute and poses a challenge for the proteolytic functioning of the proteases identified in this complex biological fluid. Despite the abundance of certain proteases in CVF, the proteolytic activity and function of proteases in CVF is not well characterized. Methods: In the present study, we employed fluorogenic substrate screening to investigate the influence of pH and inhibitory compounds on the proteolytic activity in CVF. Activity-based probe (ABP) proteomics has evolved as a powerful tool to investigate active proteases within complex proteomes and a trypsin-specific ABP was used to identify active proteases in CVF. Results: Serine proteases are among the most abundant proteins in the CVF proteome. Labeling human CVF samples with the trypsin-specific ABP revealed serine proteases transmembrane protein serine 11D and kallikrein-related peptidase 13 as active proteases in CVF. Furthermore, we demonstrated that the proteolytic activity in CVF is highly pH-dependent with an almost absolute inhibition of trypsin-like proteolytic activity at physiological pH levels. Conclusions: These findings provide a framework to understand proteolytic activity in CVF. Furthermore, the present results provide clues for a novel regulatory mechanism in which fluctuations in CVF pH have the potential to control the catalytic activity in the lower female reproductive tract.
Collapse
Affiliation(s)
- Carla M J Muytjens
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada
| | - Yijing Yu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada.,Department of Clinical Biochemistry, University Health Network, Canada, Toronto, Canada
| |
Collapse
|
9
|
Wen Y, Li H, Zeng Y, Wen W, Pendleton KP, Lui VWY, Egloff AM, Grandis JR. MAPK1E322K mutation increases head and neck squamous cell carcinoma sensitivity to erlotinib through enhanced secretion of amphiregulin. Oncotarget 2018; 7:23300-11. [PMID: 27004400 PMCID: PMC5029627 DOI: 10.18632/oncotarget.8188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/06/2016] [Indexed: 01/19/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have not been effective in unselected head and neck squamous cell carcinoma (HNSCC) populations. We previously reported an exceptional response to a brief course of erlotinib in a patient with advanced HNSCC whose tumor harbored a MAPK1E322K somatic mutation. MAPK1E322Kwas associated with increased p-EGFR, increased EGFR downstream signaling and increased sensitivity to erlotinib. In this study, we investigated the mechanism of MAPK1E322K-mediated EGFR activation in the context of erlotinib sensitivity. We demonstrated increased AREG secretion in HNSCC cell lines harboring endogenous or exogenous MAPK1E322K compared to wild type MAPK1. We found inhibition or knockdown of MAPK1 with siRNA resulted in reduced secretion of AREG and decreased sensitivity to erlotinib in the setting of MAPK1E322K. MAPK1E322K was associated with increased AREG secretion leading to an autocrine feedback loop involving AREG, EGFR and downstream signaling. Knockdown of AREG in HNSCC cells harboring MAPK1E322K abrogated EGFR signaling and decreased sensitivity to erlotinib in vitro and in vivo. These cumulative findings implicate increased AREG secretion and EGFR activation as contributing to increased erlotinib sensitivity in MAPK1E322K HNSCC.
Collapse
Affiliation(s)
- Yihui Wen
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hua Li
- Department of Otolaryngology Head and Neck Surgery, University of California at San Francisco, San Francisco, California, USA
| | - Yan Zeng
- Department of Otolaryngology Head and Neck Surgery, University of California at San Francisco, San Francisco, California, USA
| | - Weiping Wen
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Kelsey P Pendleton
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vivian W Y Lui
- Department of Pharmacology and Pharmacy, School of Biomedical Sciences, Li-Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR
| | - Ann Marie Egloff
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Molecular and Cell Biology and Otolaryngology, Boston University, Boston, Massachusetts, USA
| | - Jennifer R Grandis
- Department of Otolaryngology Head and Neck Surgery, University of California at San Francisco, San Francisco, California, USA.,Clinical and Translational Science Institute, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
10
|
Menou A, Flajolet P, Duitman J, Justet A, Moog S, Jaillet M, Tabèze L, Solhonne B, Garnier M, Mal H, Mordant P, Castier Y, Cazes A, Sallenave J, A. Mailleux A, Crestani B. Human airway trypsin‐like protease exerts potent, antifibrotic action in pulmonary fibrosis. FASEB J 2018; 32:1250-1264. [DOI: 10.1096/fj.201700583r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Awen Menou
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Pauline Flajolet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - JanWillem Duitman
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Aurélien Justet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Sophie Moog
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Madeleine Jaillet
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Laure Tabèze
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Brigitte Solhonne
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Marc Garnier
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Departement d'Anesthésie et Réanimation, (AP‐HP) Hôpital Tenon Paris France
| | - Hervé Mal
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie et Transplantation Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Pierre Mordant
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Chirurgie Thoracique et Vasculaire Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Yves Castier
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Chirurgie Thoracique et Vasculaire Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Aurélie Cazes
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Departement d'Anatomie Pathologique Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| | - Jean‐Michel Sallenave
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Arnaud A. Mailleux
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
| | - Bruno Crestani
- INSERM, Unité 1552 Paris France
- Département Hospitalo‐Universitaire Fibrosis, Inflammation, and Remodeling in Renal and Respiratory Diseases (FIRE) Paris France
- Laboratoire d'Excellence Inflamex Paris France
- Université Paris Diderot, Sorbonne Paris Cité Paris France
- Service de Pneumologie A Assistance Publique‐Hôpitaux de Paris (AP‐HP), Hôpital Bichat Paris France
| |
Collapse
|
11
|
McMahon DB, Workman AD, Kohanski MA, Carey RM, Freund JR, Hariri BM, Chen B, Doghramji LJ, Adappa ND, Palmer JN, Kennedy DW, Lee RJ. Protease-activated receptor 2 activates airway apical membrane chloride permeability and increases ciliary beating. FASEB J 2017; 32:155-167. [PMID: 28874459 DOI: 10.1096/fj.201700114rrr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/21/2017] [Indexed: 12/15/2022]
Abstract
Mucociliary clearance, driven by the engine of ciliary beating, is the primary physical airway defense against inhaled pathogens and irritants. A better understanding of the regulation of ciliary beating and mucociliary transport is necessary for identifying new receptor targets to stimulate improved clearance in airway diseases, such as cystic fibrosis and chronic rhinosinusitis. In this study, we examined the protease-activated receptor (PAR)-2, a GPCR previously shown to regulate airway cell cytokine and mucus secretion, and transepithelial Cl- current. PAR-2 is activated by proteases secreted by airway neutrophils and pathogens. We cultured various airway cell lines, primary human and mouse sinonasal cells, and human bronchial cells at air-liquid interface and examined them using molecular biology, biochemistry, and live-cell imaging. We found that PAR-2 is expressed basolaterally, where it stimulates both intracellular Ca2+ release and Ca2+ influx, which activates low-level nitric oxide production, increases apical membrane Cl- permeability ∼3-5-fold, and increases ciliary beating ∼20-50%. No molecular or functional evidence of PAR-4 was observed. These data suggest a novel and previously overlooked role of PAR-2 in airway physiology, adding to our understanding of the role of this receptor in airway Ca2+ signaling and innate immunity.-McMahon, D. B., Workman, A. D., Kohanski, M. A., Carey, R. M., Freund, J. R., Hariri, B. M., Chen, B., Doghramji, L. J., Adappa, N. D., Palmer, J. N., Kennedy, D. W., Lee, R. J. Protease-activated receptor 2 activates airway apical membrane chloride permeability and increases ciliary beating.
Collapse
Affiliation(s)
- Derek B McMahon
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alan D Workman
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael A Kohanski
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ryan M Carey
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jenna R Freund
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Benjamin M Hariri
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bei Chen
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laurel J Doghramji
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nithin D Adappa
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James N Palmer
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David W Kennedy
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robert J Lee
- Department of Otorhinolaryngology-Head and Neck Surgery University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; .,Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Menou A, Duitman J, Flajolet P, Sallenave JM, Mailleux AA, Crestani B. Human airway trypsin-like protease, a serine protease involved in respiratory diseases. Am J Physiol Lung Cell Mol Physiol 2017; 312:L657-L668. [DOI: 10.1152/ajplung.00509.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/15/2017] [Accepted: 02/15/2017] [Indexed: 01/12/2023] Open
Abstract
More than 2% of all human genes are coding for a complex system of more than 700 proteases and protease inhibitors. Among them, serine proteases play extraordinary, diverse functions in different physiological and pathological processes. The human airway trypsin-like protease (HAT), also referred to as TMPRSS11D and serine 11D, belongs to the emerging family of cell surface proteolytic enzymes, the type II transmembrane serine proteases (TTSPs). Through the cleavage of its four major identified substrates, HAT triggers specific responses, notably in epithelial cells, within the pericellular and extracellular environment, including notably inflammatory cytokine production, inflammatory cell recruitment, or anticoagulant processes. This review summarizes the potential role of this recently described protease in mediating cell surface proteolytic events, to highlight the structural features, proteolytic activity, and regulation, including the expression profile of HAT, and discuss its possible roles in respiratory physiology and disease.
Collapse
Affiliation(s)
- Awen Menou
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
| | - JanWillem Duitman
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
| | - Pauline Flajolet
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
| | - Jean-Michel Sallenave
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
| | - Arnaud André Mailleux
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
| | - Bruno Crestani
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France; and
- APHP, Hôpital Bichat, Service de Pneumologie A, Paris, France
| |
Collapse
|
13
|
Asaduzzaman M, Nadeem A, Arizmendi N, Davidson C, Nichols HL, Abel M, Ionescu LI, Puttagunta L, Thebaud B, Gordon J, DeFea K, Hollenberg MD, Vliagoftis H. Functional inhibition of PAR2 alleviates allergen-induced airway hyperresponsiveness and inflammation. Clin Exp Allergy 2016; 45:1844-55. [PMID: 26312432 DOI: 10.1111/cea.12628] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 05/28/2015] [Accepted: 06/07/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Proteinase-activated receptor 2 (PAR2 ) is a G protein-coupled receptor activated by trypsin-like serine proteinases. PAR2 activation has been associated with inflammation including allergic airway inflammation. We have also shown that PAR2 activation in the airways leads to allergic sensitization. The exact contribution of PAR2 in the development of eosinophilic inflammation and airway hyperresponsiveness (AHR) in sensitized individuals is not clear. OBJECTIVE To investigate whether functional inhibition of PAR2 during allergen challenge of allergic mice would inhibit allergen-induced AHR and inflammation in mouse models of asthma. METHODS Mice were sensitized and challenged with ovalbumin (OVA) or cockroach extract (CE). To investigate the role of PAR2 in the development of AHR and airway inflammation, we administered blocking anti-PAR2 antibodies, or a cell permeable peptide inhibitor of PAR2 signalling, pepducin, i.n. before allergen challenges and then assessed AHR and airway inflammation. RESULTS Administration of anti-PAR2 antibodies significantly inhibited OVA- and CE-induced AHR and airway inflammation. In particular, two anti-PAR2 antibodies, the monoclonal SAM-11 and polyclonal B5, inhibited AHR, airway eosinophilia, the increase of cytokines in the lung tissue and antigen-specific T cell proliferation, but had no effect on antigen-specific IgG and IgE levels. Pepducin was also effective in inhibiting AHR and airway inflammation in an OVA model of allergic airway inflammation. CONCLUSIONS AND CLINICAL RELEVANCE Functional blockade of PAR2 in the airways during allergen challenge improves allergen-induced AHR and inflammation in mice. Therefore, topical PAR2 blockade in the airways, through anti-PAR2 antibodies or molecules that interrupt PAR2 signalling, has the potential to be used as a therapeutic option in allergic asthma.
Collapse
Affiliation(s)
- M Asaduzzaman
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - A Nadeem
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - N Arizmendi
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - C Davidson
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - H L Nichols
- Division of Biomedical Sciences and Cell, Molecular and Developmental Biology, University of California, Riverside, CA, USA
| | - M Abel
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - L I Ionescu
- Department of Physiology, Women and Children Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - L Puttagunta
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - B Thebaud
- Department of Physiology, Women and Children Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - J Gordon
- Immunology Research Group, University of Saskatchewan, Saskatoon, SK, Canada
| | - K DeFea
- Division of Biomedical Sciences and Cell, Molecular and Developmental Biology, University of California, Riverside, CA, USA
| | - M D Hollenberg
- Department of Pharmacology and Therapeutics, University of Calgary, Calgary, AB, Canada
| | - H Vliagoftis
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Duhaime MJ, Page KO, Varela FA, Murray AS, Silverman ME, Zoratti GL, List K. Cell Surface Human Airway Trypsin-Like Protease Is Lost During Squamous Cell Carcinogenesis. J Cell Physiol 2016; 231:1476-83. [PMID: 26297835 PMCID: PMC4933652 DOI: 10.1002/jcp.25173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/21/2015] [Indexed: 11/17/2022]
Abstract
Cancer progression is accompanied by increased levels of extracellular proteases that are capable of remodeling the extracellular matrix, as well as cleaving and activating growth factors and receptors that are involved in pro‐cancerous signaling pathways. Several members of the type II transmembrane serine protease (TTSP) family have been shown to play critical roles in cancer progression, however, the expression or function of the TTSP Human Airway Trypsin‐like protease (HAT) in carcinogenesis has not been examined. In the present study we aimed to determine the expression of HAT during squamous cell carcinogenesis. HAT transcript is present in several tissues containing stratified squamous epithelium and decreased expression is observed in carcinomas. We determined that HAT protein is consistently expressed on the cell surface in suprabasal/apical layers of squamous cells in healthy cervical and esophageal epithelia. To assess whether HAT protein is differentially expressed in normal tissue versus tissue in different stages of carcinogenesis, we performed a comprehensive immunohistochemical analysis of HAT protein expression levels and localization in arrays of paraffin embedded human cervical and esophageal carcinomas compared to the corresponding normal tissue. We found that HAT protein is expressed in the non‐proliferating, differentiated cellular strata and is lost during the dedifferentiation of epithelial cells, a hallmark of squamous cell carcinogenesis. Thus, HAT expression may potentially be useful as a marker for clinical grading and assessment of patient prognosis in squamous cell carcinomas. J. Cell. Physiol. 231: 1476–1483, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Duhaime
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Khaliph O Page
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Fausto A Varela
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Andrew S Murray
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.,Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.,Department of Cancer Biology Graduate Program, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Michael E Silverman
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Gina L Zoratti
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Karin List
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.,Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
15
|
Miller GS, Zoratti GL, Murray AS, Bergum C, Tanabe LM, List K. HATL5: a cell surface serine protease differentially expressed in epithelial cancers. PLoS One 2014; 9:e87675. [PMID: 24498351 PMCID: PMC3912027 DOI: 10.1371/journal.pone.0087675] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/28/2013] [Indexed: 01/15/2023] Open
Abstract
Over the last two decades, cell surface proteases belonging to the type II transmembrane serine protease (TTSP) family have emerged as important enzymes in the mammalian degradome, playing critical roles in epithelial biology, regulation of metabolic homeostasis, and cancer. Human airway trypsin-like protease 5 (HATL5) is one of the few family members that remains uncharacterized. Here we demonstrate that HATL5 is a catalytically active serine protease that is inhibited by the two Kunitz type serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 and 2, as well as by serpinA1. Full-length HATL5 is localized on the cell surface of cultured mammalian cells as demonstrated by confocal microscopy. HATL5 displays a relatively restricted tissue expression profile, with both transcript and protein present in the cervix, esophagus, and oral cavity. Immunohistochemical analysis revealed an expression pattern where HATL5 is localized on the cell surface of differentiated epithelial cells in the stratified squamous epithelia of all three of these tissues. Interestingly, HATL5 is significantly decreased in cervical, esophageal, and head and neck carcinomas as compared to normal tissue. Analysis of cervical and esophageal cancer tissue arrays demonstrated that the squamous epithelial cells lose their expression of HATL5 protein upon malignant transformation.
Collapse
Affiliation(s)
- Gregory S. Miller
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Gina L. Zoratti
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Andrew S. Murray
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Christopher Bergum
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Lauren M. Tanabe
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Karin List
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
- * E-mail:
| |
Collapse
|
16
|
Berasain C, Avila MA. Amphiregulin. Semin Cell Dev Biol 2014; 28:31-41. [PMID: 24463227 DOI: 10.1016/j.semcdb.2014.01.005] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 12/26/2022]
Abstract
Amphiregulin (AREG) is a ligand of the epidermal growth factor receptor (EGFR), a widely expressed transmembrane tyrosine kinase. AREG is synthesized as a membrane-anchored precursor protein that can engage in juxtacrine signaling on adjacent cells. Alternatively, after proteolytic processing by cell membrane proteases, mainly TACE/ADAM17, AREG is secreted and behaves as an autocrine or paracrine factor. AREG gene expression and release is induced by a plethora of stimuli including inflammatory lipids, cytokines, hormones, growth factors and xenobiotics. Through EGFR binding AREG activates major intracellular signaling cascades governing cell survival, proliferation and motility. Physiologically, AREG plays an important role in the development and maturation of mammary glands, bone tissue and oocytes. Chronic elevation of AREG expression is increasingly associated with different pathological conditions, mostly of inflammatory and/or neoplastic nature. Here we review the essential aspects of AREG structure, function and regulation, discuss the basis for its differential role within the EGFR family of ligands, and identify emerging aspects in AREG research with translational potential.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain; CIBERehd, Clinica Universidad de Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain.
| | - Matías A Avila
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain; CIBERehd, Clinica Universidad de Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain.
| |
Collapse
|
17
|
Jickling GC, Ander BP, Stamova B, Zhan X, Liu D, Rothstein L, Verro P, Khoury J, Jauch EC, Pancioli AM, Broderick JP, Sharp FR. RNA in blood is altered prior to hemorrhagic transformation in ischemic stroke. Ann Neurol 2013; 74:232-40. [PMID: 23468366 DOI: 10.1002/ana.23883] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 02/19/2013] [Accepted: 03/01/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Hemorrhagic transformation (HT) is a major complication of ischemic stroke that worsens outcomes and increases mortality. Disruption of the blood-brain barrier is a central feature of HT pathogenesis, and leukocytes may contribute to this process. We sought to determine whether ischemic strokes that develop HT have differences in RNA expression in blood within 3 hours of stroke onset prior to treatment with thrombolytic therapy. METHODS Stroke patient blood samples were obtained prior to treatment with thrombolysis, and leukocyte RNA was assessed by microarray analysis. Strokes that developed HT (n = 11) were compared to strokes without HT (n = 33) and controls (n = 14). Genes were identified (corrected p < 0.05, fold change ≥|1.2|), and functional analysis was performed. RNA prediction of HT in stroke was evaluated using cross-validation, and in a second stroke cohort (n = 52). RESULTS Ischemic strokes that developed HT had differential expression of 29 genes in circulating leukocytes prior to treatment with thrombolytic therapy. A panel of 6 genes could predict strokes that later developed HT with 80% sensitivity and 70.2% specificity. Key pathways involved in HT of human stroke are described, including amphiregulin, a growth factor that regulates matrix metalloproteinase-9; a shift in transforming growth factor-β signaling involving SMAD4, INPP5D, and IRAK3; and a disruption of coagulation factors V and VIII. INTERPRETATION Identified genes correspond to differences in inflammation and coagulation that may predispose to HT in ischemic stroke. Given the adverse impact of HT on stroke outcomes, further evaluation of the identified genes and pathways is warranted to determine their potential as therapeutic targets to reduce HT and as markers of HT risk.
Collapse
Affiliation(s)
- Glen C Jickling
- Department of Neurology and the MIND Institute, University of California, Davis, Sacramento, CA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Clarke LA, Sousa L, Barreto C, Amaral MD. Changes in transcriptome of native nasal epithelium expressing F508del-CFTR and intersecting data from comparable studies. Respir Res 2013; 14:38. [PMID: 23537407 PMCID: PMC3637641 DOI: 10.1186/1465-9921-14-38] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 03/07/2013] [Indexed: 01/06/2023] Open
Abstract
Background Microarray studies related to cystic fibrosis (CF) airway gene expression have gone some way in clarifying the complex molecular background of CF lung diseases, but have made little progress in defining a robust “molecular signature” associated with mutant CFTR expression. Disparate methodological and statistical analyses complicate comparisons between independent studies of the CF transcriptome, and although each study may be valid in isolation, the conclusions reached differ widely. Methods We carried out a small-scale whole genome microarray study of gene expression in human native nasal epithelial cells from F508del-CFTR homozygotes in comparison to non-CF controls. We performed superficial comparisons with other microarray datasets in an attempt to identify a subset of regulated genes that could act as a signature of F508del-CFTR expression in native airway tissue samples. Results Among the alterations detected in CF, up-regulation of genes involved in cell proliferation, and down-regulation of cilia genes were the most notable. Other changes involved gene expression changes in calcium and membrane pathways, inflammation, defence response, wound healing and the involvement of estrogen signalling. Comparison of our data set with previously published studies allowed us to assess the consistency of independent microarray data sets, and shed light on the limitations of such snapshot studies in measuring a system as subtle and dynamic as the transcriptome. Comparison of in-vivo studies nevertheless yielded a small molecular CF signature worthy of future investigation. Conclusions Despite the variability among the independent studies, the current CF transcriptome meta-analysis identified subsets of differentially expressed genes in native airway tissues which provide both interesting clues to CF pathogenesis and a possible CF biomarker.
Collapse
Affiliation(s)
- Luka A Clarke
- BioFIG-Centre for Biodiversity, Functional and Integrative Genomics, FCUL-Faculty of Sciences, University of Lisboa, Lisboa 1749-016, Portugal.
| | | | | | | |
Collapse
|
19
|
Antalis TM. DESC1 and HAT Peptidases. HANDBOOK OF PROTEOLYTIC ENZYMES 2013. [PMCID: PMC7150303 DOI: 10.1016/b978-0-12-382219-2.00654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
At the crossroads: EGFR and PTHrP signaling in cancer-mediated diseases of bone. Odontology 2012; 100:109-29. [PMID: 22684584 DOI: 10.1007/s10266-012-0070-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 01/01/2023]
Abstract
The epidermal growth factor receptor is a well-established cancer therapeutic target due to its stimulation of proliferation, motility, and resistance to apoptosis. Recently, additional roles for the receptor have been identified in growth of metastases. Similar to development, metastatic spread requires signaling interactions between epithelial-derived tumor cells and mesenchymal derivatives of the microenvironment. This necessitates reactivation of developmental signaling molecules, including the hypercalcemia factor parathyroid hormone-related protein. This review covers the variations of epidermal growth factor receptor signaling in cancers that produce bone metastases, regulation of parathyroid hormone-related protein, and evidence that the two molecules drive cancer-mediated diseases of bone.
Collapse
|
21
|
Kato M, Hashimoto T, Shimomura T, Kataoka H, Ohi H, Kitamura N. Hepatocyte growth factor activator inhibitor type 1 inhibits protease activity and proteolytic activation of human airway trypsin-like protease. J Biochem 2011; 151:179-87. [PMID: 22023801 DOI: 10.1093/jb/mvr131] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hepatocyte growth factor activator inhibitor type 1 (HAI-1) is a Kunitz-type transmembrane serine protease inhibitor initially identified as a potent inhibitor of hepatocyte growth factor activator (HGFA), a serine protease that converts pro-HGF to the active form. HAI-1 also has inhibitory activity against serine proteases such as matriptase, hepsin and prostasin. In this study, we examined effects of HAI-1 on the protease activity and proteolytic activation of human airway trypsin-like protease (HAT), a transmembrane serine protease that is expressed mainly in bronchial epithelial cells. A soluble form of HAI-1 inhibited the protease activity of HAT in vitro. HAT was proteolytically activated in cultured mammalian cells transfected with its expression vector, and a soluble form of active HAT was released into the conditioned medium. The proteolytic activation of HAT required its own serine protease activity. Co-expression of the transmembrane full-length HAI-1 inhibited the proteolytic activation of HAT. In addition, full-length HAI-1 associated with the transmembrane full-length HAT in co-expressing cells. Like other target proteases of HAI-1, HAT converted pro-HGF to the active form in vitro. These results suggest that HAI-1 functions as a physiological regulator of HAT by inhibiting its protease activity and proteolytic activation in airway epithelium.
Collapse
Affiliation(s)
- Minoru Kato
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan.
| | | | | | | | | | | |
Collapse
|
22
|
Choi YH, Lee SN, Aoyagi H, Yamasaki Y, Yoo JY, Park B, Shin DM, Yoon HG, Yoon JH. The extracellular signal-regulated kinase mitogen-activated protein kinase/ribosomal S6 protein kinase 1 cascade phosphorylates cAMP response element-binding protein to induce MUC5B gene expression via D-prostanoid receptor signaling. J Biol Chem 2011; 286:34199-214. [PMID: 21832046 DOI: 10.1074/jbc.m111.247684] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mucus hypersecretion is a prominent feature of respiratory diseases, and MUC5B is a major airway mucin. Mucin gene expression can be affected by inflammatory mediators, including prostaglandin (PG) D(2,) an inflammatory mediator synthesized by hematopoietic PGD synthase (H-PGDS). PGD(2) binds to either D-prostanoid receptor (DP1) or chemoattractant receptor homologous molecule expressed on T-helper type 2 cells (CRTH2). We investigated the mechanisms by which PGD(2) induces MUC5B gene expression in airway epithelial cells. Western blot analysis showed that H-PGDS was highly expressed in nasal polyps. Similar results were obtained for PGD(2) expression. In addition, we could clearly detect the expressions of both H-PGDS and DP1 in nasal epithelial cells but not CRTH2. We demonstrated that PGD(2) increased MUC5B gene expression in normal human nasal epithelial cells as well as in NCI-H292 cells in vitro. S5751, a DP1 antagonist, inhibited PGD(2)-induced MUC5B expression, whereas a CRTH2 antagonist (OC0459) did not. These data suggest that PGD(2) induced MUC5B expression via DP1. Pretreatment with extracellular signal-regulated kinase (ERK) inhibitor (PD98059) blocked both PGD(2)-induced ERK mitogen-activated protein kinase (MAPK) activation and MUC5B expression. Proximity ligation assays showed direct interaction between RSK1 and cAMP response element-binding protein (CREB). Stimulation with PGD(2) caused an increase in intracellular cAMP levels, whereas intracellular Ca(2+) did not have such an effect. PGD(2)-induced MUC5B mRNA levels were regulated by CREB via direct interaction with two cAMP-response element sites (-921/-914 and -900/-893). Finally, we demonstrated that PGD(2) can induce MUC5B overproduction via ERK MAPK/RSK1/CREB signaling and that DP1 receptor may have suppressive effects in controlling MUC5B overproduction in the airway.
Collapse
Affiliation(s)
- Yeon Ho Choi
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sales KU, Hobson JP, Wagenaar-Miller R, Szabo R, Rasmussen AL, Bey A, Shah MF, Molinolo AA, Bugge TH. Expression and genetic loss of function analysis of the HAT/DESC cluster proteases TMPRSS11A and HAT. PLoS One 2011; 6:e23261. [PMID: 21853097 PMCID: PMC3154331 DOI: 10.1371/journal.pone.0023261] [Citation(s) in RCA: 38] [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: 05/20/2011] [Accepted: 07/09/2011] [Indexed: 11/18/2022] Open
Abstract
Genome mining at the turn of the millennium uncovered a new family of type II transmembrane serine proteases (TTSPs) that comprises 17 members in humans and 19 in mice. TTSPs phylogenetically belong to one of four subfamilies: matriptase, hepsin/TMPRSS, corin and HAT/DESC. Whereas a wealth of information now has been gathered as to the physiological functions of members of the hepsin/TMPRSS, matriptase, and corin subfamilies of TTSPs, comparatively little is known about the functions of the HAT/DESC subfamily of proteases. Here we perform a combined expression and functional analysis of this TTSP subfamily. We show that the five human and seven murine HAT/DESC proteases are coordinately expressed, suggesting a level of functional redundancy. We also perform a comprehensive phenotypic analysis of mice deficient in two of the most widely expressed HAT/DESC proteases, TMPRSS11A and HAT, and show that the two proteases are dispensable for development, health, and long-term survival in the absence of external challenges or additional genetic deficits. Our comprehensive expression analysis and generation of TMPRSS11A- and HAT-deficient mutant mouse strains provide a valuable resource for the scientific community for further exploration of the HAT/DESC subfamily proteases in physiological and pathological processes.
Collapse
Affiliation(s)
- Katiuchia Uzzun Sales
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John P. Hobson
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rebecca Wagenaar-Miller
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Extramural Activities, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Roman Szabo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amber L. Rasmussen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexandra Bey
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Maham F. Shah
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alfredo A. Molinolo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas H. Bugge
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
24
|
Antalis TM, Bugge TH, Wu Q. Membrane-anchored serine proteases in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 99:1-50. [PMID: 21238933 PMCID: PMC3697097 DOI: 10.1016/b978-0-12-385504-6.00001-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Serine proteases of the trypsin-like family have long been recognized to be critical effectors of biological processes as diverse as digestion, blood coagulation, fibrinolysis, and immunity. In recent years, a subgroup of these enzymes has been identified that are anchored directly to plasma membranes, either by a carboxy-terminal transmembrane domain (Type I), an amino-terminal transmembrane domain with a cytoplasmic extension (Type II or TTSP), or through a glycosylphosphatidylinositol (GPI) linkage. Recent biochemical, cellular, and in vivo analyses have now established that membrane-anchored serine proteases are key pericellular contributors to processes vital for development and the maintenance of homeostasis. This chapter reviews our current knowledge of the biological and physiological functions of these proteases, their molecular substrates, and their contributions to disease.
Collapse
Affiliation(s)
- Toni M Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
25
|
Suen JY, Gardiner B, Grimmond S, Fairlie DP. Profiling gene expression induced by protease-activated receptor 2 (PAR2) activation in human kidney cells. PLoS One 2010; 5:e13809. [PMID: 21072196 PMCID: PMC2970545 DOI: 10.1371/journal.pone.0013809] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 10/04/2010] [Indexed: 12/28/2022] Open
Abstract
Protease-Activated Receptor-2 (PAR2) has been implicated through genetic knockout mice with cytokine regulation and arthritis development. Many studies have associated PAR2 with inflammatory conditions (arthritis, airways inflammation, IBD) and key events in tumor progression (angiogenesis, metastasis), but they have relied heavily on the use of single agonists to identify physiological roles for PAR2. However such probes are now known not to be highly selective for PAR2, and thus precisely what PAR2 does and what mechanisms of downstream regulation are truly affected remain obscure. Effects of PAR2 activation on gene expression in Human Embryonic Kidney cells (HEK293), a commonly studied cell line in PAR2 research, were investigated here by comparing 19,000 human genes for intersecting up- or down-regulation by both trypsin (an endogenous protease that activates PAR2) and a PAR2 activating hexapeptide (2f-LIGRLO-NH(2)). Among 2,500 human genes regulated similarly by both agonists, there were clear associations between PAR2 activation and cellular metabolism (1,000 genes), the cell cycle, the MAPK pathway, HDAC and sirtuin enzymes, inflammatory cytokines, and anti-complement function. PAR-2 activation up-regulated four genes more than 5 fold (DUSP6, WWOX, AREG, SERPINB2) and down-regulated another six genes more than 3 fold (TXNIP, RARG, ITGB4, CTSD, MSC and TM4SF15). Both PAR2 and PAR1 activation resulted in up-regulated expression of several genes (CD44, FOSL1, TNFRSF12A, RAB3A, COPEB, CORO1C, THBS1, SDC4) known to be important in cancer. This is the first widespread profiling of specific activation of PAR2 and provides a valuable platform for better understanding key mechanistic roles of PAR2 in human physiology. Results clearly support the development of both antagonists and agonists of human PAR2 as potential disease modifying therapeutic agents.
Collapse
Affiliation(s)
- Jacky Y. Suen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Brooke Gardiner
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Sean Grimmond
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P. Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
26
|
Lung protease/anti-protease network and modulation of mucus production and surfactant activity. Biochimie 2010; 92:1608-17. [DOI: 10.1016/j.biochi.2010.05.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 05/14/2010] [Indexed: 12/27/2022]
|
27
|
The cutting edge: membrane-anchored serine protease activities in the pericellular microenvironment. Biochem J 2010; 428:325-46. [PMID: 20507279 DOI: 10.1042/bj20100046] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The serine proteases of the trypsin-like (S1) family play critical roles in many key biological processes including digestion, blood coagulation, and immunity. Members of this family contain N- or C-terminal domains that serve to tether the serine protease catalytic domain directly to the plasma membrane. These membrane-anchored serine proteases are proving to be key components of the cell machinery for activation of precursor molecules in the pericellular microenvironment, playing vital functions in the maintenance of homoeostasis. Substrates activated by membrane-anchored serine proteases include peptide hormones, growth and differentiation factors, receptors, enzymes, adhesion molecules and viral coat proteins. In addition, new insights into our understanding of the physiological functions of these proteases and their involvement in human pathology have come from animal models and patient studies. The present review discusses emerging evidence for the diversity of this fascinating group of membrane serine proteases as potent modifiers of the pericellular microenvironment through proteolytic processing of diverse substrates. We also discuss the functional consequences of the activities of these proteases on mammalian physiology and disease.
Collapse
|
28
|
Cosgrove BD, Alexopoulos LG, Hang TC, Hendriks BS, Sorger PK, Griffith LG, Lauffenburger DA. Cytokine-associated drug toxicity in human hepatocytes is associated with signaling network dysregulation. MOLECULAR BIOSYSTEMS 2010; 6:1195-206. [PMID: 20361094 PMCID: PMC2943488 DOI: 10.1039/b926287c] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Idiosyncratic drug hepatotoxicity is a major problem in pharmaceutical development due to poor prediction capability of standard preclinical toxicity assessments and limited knowledge of its underlying mechanisms. Findings in animal models have shown that adverse effects of numerous drugs with idiosyncratic hepatotoxicity in humans can be reproduced in the presence of coincident inflammatory cytokine signaling. Following these observations, we have recently developed an in vitro drug/inflammatory cytokine co-treatment approach that can reproduce clinical drug hepatotoxicity signatures-particularly for idiosyncratic drugs-in cultured primary human hepatocytes. These observations have suggested that drug-induced stresses may interact with cytokine signaling to induce hepatic cytotoxicity, but the hepatocyte signaling mechanisms governing these interactions are poorly understood. Here, we collect high-throughput phosphoprotein signaling and cytotoxicity measurements in cultured hepatocytes, from multiple human donors, treated with combinations of hepatotoxic drugs (e.g. trovafloxacin, clarithromycin) and cytokines (tumor necrosis factor-alpha, interferon-gamma, interleukin-1 alpha, and interleukin-6). We demonstrate, through orthogonal partial least-squares regression (OPLSR) modeling of these signal-response data, that drug/cytokine hepatic cytotoxicity is integratively controlled by four key signaling pathways: Akt, p70 S6 kinase, MEK-ERK, and p38-HSP27. This modeling predicted, and experimental studies confirmed, that the MEK-ERK and p38-HSP27 pathways contribute pro-death signaling influences in drug/cytokine hepatic cytotoxicity synergy. Further, our four-pathway OPLSR model produced successful prediction of drug/cytokine hepatic cytotoxicities across different human donors, even though signaling and cytotoxicity responses were both highly donor-specific. Our findings highlight the critical role of kinase signaling in drug/cytokine hepatic cytotoxicity synergies and reveal that hepatic cytotoxicity responses are governed by multi-pathway signaling network balance.
Collapse
Affiliation(s)
- Benjamin D. Cosgrove
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Biotechnology Process Engineering Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Leonidas G. Alexopoulos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ta-chun Hang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Biotechnology Process Engineering Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Bart S. Hendriks
- Pfizer Research Technology Center, Cambridge, Massachusetts, USA
| | - Peter K. Sorger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Linda G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Biotechnology Process Engineering Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Biotechnology Process Engineering Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Cell Decision Processes Center, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
29
|
Wysocka M, Spichalska B, Lesner A, Jaros M, Brzozowski K, Łegowska A, Rolka K. Substrate specificity and inhibitory study of human airway trypsin-like protease. Bioorg Med Chem 2010; 18:5504-9. [PMID: 20620066 DOI: 10.1016/j.bmc.2010.06.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 06/14/2010] [Accepted: 06/16/2010] [Indexed: 11/19/2022]
Abstract
Human airway trypsin-like protease (HAT), also referred to as TMPRSS11D, is an important physiological enzyme with the main activity pronounced in an airway. In this work we have described the substrate specificity and selectivity study of the protease, performed by the combinatorial approach. Fluorogenic/chromogenic tetrapeptide library was used for this purpose. The most efficiently hydrolyzed substrates' sequences that we selected were ABZ-Arg-Gln-Asp-Arg(Lys)-ANB-NH(2). The most active inhibitor with C-terminal Arg residue underwent detectable proteolysis action in the presence of 35pM of HAT. Based on the selected sequences the two peptide aldehydes were synthesized and (Abz-Arg-Gln-Asp-Arg(Lys)-H) were found to be an effective HAT inhibitor, working in nanomolar range with inhibition constant 54nM and 112nM, respectively.
Collapse
Affiliation(s)
- M Wysocka
- Faculty of Chemistry Gdańsk University, ul Sobieskiego 18, 80-952 Gdańsk, Poland
| | | | | | | | | | | | | |
Collapse
|
30
|
Ewen D, Clarke S, Smith J, Berger C, Salmon G, Trevethick M, Shute J. The role of protease-activated receptors PAR-1 and PAR-2 in the repair of 16HBE 14o−epithelial cell monolayersin vitro. Clin Exp Allergy 2010; 40:435-49. [DOI: 10.1111/j.1365-2222.2010.03453.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Type II transmembrane serine proteases in cancer and viral infections. Trends Mol Med 2009; 15:303-12. [PMID: 19581128 DOI: 10.1016/j.molmed.2009.05.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 05/07/2009] [Accepted: 05/07/2009] [Indexed: 01/08/2023]
Abstract
Regulated proteolysis of cellular factors is pivotal to tissue development and homeostasis, whereas uncontrolled proteolytic activity is linked to disease. Type II transmembrane serine proteases (TTSPs) are expressed at the cell surface and are thus ideally located to regulate cell-cell and cell-matrix interactions. Increasing evidence demonstrates that aberrant expression of TTSPs is a hallmark of several cancers and recent studies have defined molecular mechanisms underlying TTSP-promoted carcinogenesis. In addition, new findings suggest that influenza and other respiratory viruses could exploit TTSPs to promote their spread, making these proteases potential targets for intervention in cancer and viral infections. Here, we review the role of TTSPs in tumorigenesis and viral infection and discuss potential approaches to therapy.
Collapse
|
32
|
Abstract
Analysis of genome and expressed sequence tag data bases at the turn of the millennium unveiled a new protease family named the type II transmembrane serine proteases (TTSPs) in a Journal of Biological Chemistry minireview (Hooper, J. D., Clements, J. A., Quigley, J. P., and Antalis, T. M. (2001) J. Biol. Chem. 276, 857-860). Since then, the number of known TTSPs has more than doubled, and more importantly, our understanding of the physiological functions of individual TTSPs and their contribution to human disease has greatly increased. Progress has also been made in identifying molecular substrates and endogenous inhibitors. This minireview summarizes the current knowledge of the rapidly advancing TTSP field.
Collapse
Affiliation(s)
- Thomas H Bugge
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
33
|
Heijink IH, Postma DS, Noordhoek JA, Broekema M, Kapus A. House dust mite-promoted epithelial-to-mesenchymal transition in human bronchial epithelium. Am J Respir Cell Mol Biol 2009; 42:69-79. [PMID: 19372245 DOI: 10.1165/rcmb.2008-0449oc] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The molecular basis of airway remodeling and loss of epithelial integrity in asthma is still undefined. We aimed to establish if exposure of human bronchial epithelium (16HBE cells) to asthma-related stimuli can induce epithelial-to-mesenchymal transition (EMT), a key process in tissue repair and remodeling associated with loss of intercellular contacts. We studied the effects of fibrogenic cytokine TGF-beta and protease-containing aeroallergen house dust mite (HDM) on mesenchymal and epithelial markers, cytoskeleton organization, and activation of beta-catenin-driven reporter TopFLASH. TGF-beta alone up-regulated vimentin and fibronectin, modestly down-regulated E-cadherin, but did not affect cytokeratin. HDM alone did not affect these markers, but promoted stress fibers. Importantly, when added to TGF-beta-primed epithelium, HDM induced E-cadherin internalization, enhanced beta-catenin-dependent transcription, and down-regulated cytokeratin. Regarding the underlying mechanisms, the stimuli together induced sustained myosin light chain phosphorylation, which was crucial for E-cadherin internalization and beta-catenin-dependent transcription. Previously, we showed that HDM signals through the epidermal growth factor receptor (EGFR). Accordingly, inhibition of EGFR prevented TGF-beta/HDM-induced mesenchymalization. TGF-beta facilitated uncoupling of EGFR from E-cadherin, its negative regulator, and prolonged EGFR signaling. Thus, we show that HDM promotes EMT in TGF-beta-primed epithelium. Analysis of primary epithelium appears consistent with this phenotypic change. We propose that TGF-beta secretion and dysregulated EGFR signaling may increase epithelial vulnerability to allergens and trigger the induction of EMT, a hitherto unrecognized contributor to airway remodeling in asthma.
Collapse
Affiliation(s)
- Irene H Heijink
- Department of Allergology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Willmarth NE, Ethier SP. Amphiregulin as a novel target for breast cancer therapy. J Mammary Gland Biol Neoplasia 2008; 13:171-9. [PMID: 18437539 DOI: 10.1007/s10911-008-9081-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Accepted: 04/03/2008] [Indexed: 10/22/2022] Open
Abstract
Amphiregulin, an EGF family growth factor, binds and activates the epidermal growth factor receptor (EGFR or ErbB1). Activation of the EGFR by amphiregulin can occur through autocrine, paracrine and juxtacrine mechanisms. Amphiregulin plays a role in several biological processes including nerve regeneration, blastocyst implantation, and bone formation. Amphiregulin also plays an important role in mammary duct formation as well as the outgrowth and branching of several other human tissues such as the lung, kidney and prostate. This effect is most likely due to the induction of genes involved in invasion and migration such as cytokines and matrix metalloproteases. Clinical studies have suggested that amphiregulin also plays a role in human breast cancer progression and its expression has been associated with aggressive disease. Therefore, amphiregulin may be a novel and effective target for the treatment of breast cancer and could represent an alternative to targeting the EGFR.
Collapse
Affiliation(s)
- Nicole E Willmarth
- Cancer Biology, Thomas Jefferson University, 233 S. 10th Street, Philadelphia, PA, USA
| | | |
Collapse
|
35
|
Adib-Conquy M, Pedron T, Petit-Bertron AF, Tabary O, Corvol H, Jacquot J, Clément A, Cavaillon JM. Neutrophils in cystic fibrosis display a distinct gene expression pattern. Mol Med 2008; 14:36-44. [PMID: 18026571 DOI: 10.2119/2007-00081.adib-conquy] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Accepted: 11/01/2007] [Indexed: 11/06/2022] Open
Abstract
We compared gene expression in blood neutrophils (polymorphonuclear leukocytes, or PMNs) collected from healthy subjects with those of cystic fibrosis (CF) patients devoid of bacterial colonization. Macroarray analysis of 1050 genes revealed upregulation of 62 genes and downregulation expression of 27 genes in CF blood PMNs. Among upregulated genes were those coding for vitronectin, some chemokines (particularly CCL17 and CCL18), some interleukin (IL) receptors (IL-3, IL-8, IL-10, IL-12), all three colony-stimulating factors (G-, M-, GM-CSF), numerous genes coding for molecules involved in signal transduction, and a few genes under the control of gamma-interferon. In contrast, none of the genes coding for adhesion molecules were modulated. The upregulation of six genes in CF PMNs (coding for thrombospondin-1, G-CSF, CXCL10, CCL17, IKKvarepsilon, IL-10Ra) was further confirmed by qPCR. In addition, the increased presence of G-CSF, CCL17, and CXCL10 was confirmed by ELISA in supernatants of neutrophils from CF patients. When comparison was performed between blood and airway PMNs of CF patients, there was a limited difference in terms of gene expression. Only the mRNA expression of amphiregulin and tumor necrosis factor (TNF) receptor p55 were significantly higher in airway PMNs. The presence of amphiregulin was confirmed by ELISA in the sputum of CF patients, suggesting for the first time a role of amphiregulin in cystic fibrosis. Altogether, this study clearly demonstrates that blood PMNs from CF patients display a profound modification of gene expression profile associated with the disease, suggesting a state of activation of these cells.
Collapse
|
36
|
Ostrowska E, Reiser G. Protease-activated receptor (PAR)-induced interleukin-8 production in airway epithelial cells requires activation of MAP kinases p44/42 and JNK. Biochem Biophys Res Commun 2008; 366:1030-5. [DOI: 10.1016/j.bbrc.2007.12.064] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2007] [Accepted: 12/10/2007] [Indexed: 11/17/2022]
|
37
|
Sokolova E, Reiser G. A novel therapeutic target in various lung diseases: Airway proteases and protease-activated receptors. Pharmacol Ther 2007; 115:70-83. [PMID: 17532472 DOI: 10.1016/j.pharmthera.2007.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 04/05/2007] [Indexed: 10/23/2022]
Abstract
Protease-activated receptors (PAR), which are G protein-coupled receptors, have 4 members, PAR-1 to PAR-4. PARs are activated by proteolysis of a peptide bond at the N-terminal domain of the receptor. PARs are widely distributed throughout the airways. Their activity is modulated by airway proteases of endogenous and exogenous origin, which can either activate or disable the receptors. The regulation of PAR activity by proteases is important under pathological conditions when the activity of proteases is increased. Moreover, various inflammatory mediators, such as cytokines, growth factors, or prostanoids, alter the PAR expression level. Elevated PAR levels are observed in various lung disorders, and their significance in the development of pathological situations in the lung is currently intensively investigated. Consequences of PAR activation can be either beneficial or deleterious, depending on the PAR subtype. PAR-1 has been shown to be an important player in the development of pulmonary fibrosis. Thus, PAR-1 represents an exciting target for clinical intervention in fibrotic diseases. PAR-2 contributes to allergic airway inflammation. However, the question whether the impact of PAR-2 is beneficial or deleterious is still under intensive discussion. Therefore, precise information concerning the participation of PAR-2 in various lesions is required. Moreover, it is necessary to generate selective PAR- and organ-targeted approaches for treating the diseases. A thorough understanding of PAR-induced cellular events and the consequences of receptor blockade may help in the development of novel therapeutic strategies targeted to prevent lung destruction and to avoid deterioration of conditions of patients with inflammatory or fibrotic lung diseases.
Collapse
Affiliation(s)
- Elena Sokolova
- Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Zentrum für Biochemie und Molekularbiologie, Institut für Neurobiochemie, Leipziger Strasse 44, D-39120, Magdeburg, Germany
| | | |
Collapse
|
38
|
Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol Rev 2007; 87:1047-82. [PMID: 17615396 DOI: 10.1152/physrev.00048.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD), comprised of pulmonary emphysema, chronic bronchitis, and structural and inflammatory changes of small airways, is a leading cause of morbidity and mortality in the world. A better understanding of the pathobiology of COPD is critical for the developing of novel therapies, as the majority of patients with the disease have little therapeutic options at the present time. The pathobiology of COPD encompasses multiple injurious processes including inflammation (excessive or inappropriate innate and adaptive immunity), cellular apoptosis, altered cellular and molecular alveolar maintenance program, abnormal cell repair, extracellular matrix destruction (protease and anti-protease imbalance), and oxidative stress (oxidant and antioxidant imbalance). These processes are triggered by urban and rural air pollutants and active and/or passive cigarette smoke and modified by cellular senescence and infection. A series of receptor-mediated signal transduction pathways are activated by reactive oxygen species and tobacco components, resulting in impairment of a variety of cell signaling and cytokine networks, subsequently leading to chronic airway responses with mucus production, airway remodeling, and alveolar destruction. The authors provide an updated insight into the molecular and cellular pathobiology of COPD based on human and/or animal data.
Collapse
Affiliation(s)
- Toshinori Yoshida
- Division of Cardiopulmonary Pathology, Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
39
|
Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation in human airway epithelial cells. Am J Respir Cell Mol Biol 2007; 37:169-85. [PMID: 17413031 DOI: 10.1165/rcmb.2006-0466oc] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
When cultured at an air-liquid interface (ALI) in the appropriate medium, primary human airway epithelial cells form a polarized, pseudostratified epithelium composed of ciliated and mucus-secreting cells. This culture system provides a useful tool for the in vitro study of airway epithelial biology and differentiation. We have performed microarray analysis on ALI cultures of human bronchial epithelial cells (HBECs) grown over a 28-d period to identify genes involved in mucociliary differentiation. We identified over 2,000 genes that displayed statistically significant 2-fold or greater changes in expression during the time course. Of the genes showing the largest increases, many are involved in processes associated with airway epithelial biology, such as cell adhesion, immunity, transport, and cilia formation; however, many novel genes were also identified. We compared our results with data from proteomic analyses of the ciliary axoneme and identified candidate genes that may have roles in cilia formation or function. Gene networks were generated using Ingenuity Pathways Analysis (Ingenuity Systems, Redwood City, CA) to identify signaling pathways involved in mucociliary cell differentiation or function. Networks containing genes involved in TGF-beta, WNT/beta-catenin, and epidermal growth factor receptor (EGFR) pathways were identified, suggesting potential roles for these families in airway epithelia. Microarray results were validated by real-time RT-PCR for a number of representative genes. This work has provided extensive information about gene expression changes during differentiation of airway epithelial cells, and will be a useful resource for researchers interested in respiratory function, pathology, and toxicology.
Collapse
Affiliation(s)
- Andrea J Ross
- Clinical Research Branch, Human Studies Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, University of North Carolina, Chapel Hill, North Carolina 27599-7315, USA
| | | | | | | |
Collapse
|
40
|
Wang H, Yi T, Zheng Y, He S. Induction of monocyte chemoattractant protein-1 release from A549 cells by agonists of protease-activated receptor-1 and -2. Eur J Cell Biol 2007; 86:233-42. [PMID: 17280738 DOI: 10.1016/j.ejcb.2006.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 10/27/2006] [Accepted: 12/04/2006] [Indexed: 02/05/2023] Open
Abstract
Hypersecretion of cytokines and serine proteases has been observed in asthma. However, the influence of proteases and protease-activated receptors (PARs) on monocyte chemoattractant protein-1 (MCP-1) release from airway epithelial cells remains largely unknown. In the present study, A549 cells were challenged with agonists of PARs, and levels of MCP-1 released in the supernatant and mRNA expression were examined by ELISA and real time polymerase chain reaction (PCR), respectively. The results show that thrombin, tryptase, elastase and trypsin induced an up to 6.5-, 1.8-, 1.6-, and 3.1-fold increase in MCP-1 release from A549 cells, respectively, following a 16-h incubation period. The protease-induced secretion of MCP-1 can be abolished by specific protease inhibitors. Agonist peptides of PAR-1 and PAR-2 stimulate MCP-1 secretion up to 15- and 12.7-fold, respectively. Real-time PCR showed that MCP-1 mRNA is up-regulated by the serine proteases tested and by agonist peptides of PAR-1 and PAR-2. In conclusion, serine proteases can stimulate MCP-1 release from A549 cells possibly through a PARs-related mechanism, suggesting that they are likely to contribute to MCP-1-related airway inflammatory disorders in man.
Collapse
Affiliation(s)
- Haiyan Wang
- Allergy and Inflammation Research Institute, The Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | | | | | | |
Collapse
|
41
|
Beaufort N, Leduc D, Eguchi H, Mengele K, Hellmann D, Masegi T, Kamimura T, Yasuoka S, Fend F, Chignard M, Pidard D. The human airway trypsin-like protease modulates the urokinase receptor (uPAR, CD87) structure and functions. Am J Physiol Lung Cell Mol Physiol 2007; 292:L1263-72. [PMID: 17237151 DOI: 10.1152/ajplung.00191.2006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human airway trypsin-like protease (HAT) is a respiratory epithelium-associated, type II transmembrane serine protease, which is also detected as an extracellular enzyme in lung fluids during airway inflammatory disorders. We have evaluated its capacity to affect the urokinase-type plasminogen activator receptor (uPAR), a membrane glycolipid-anchored, three-domain (D1D2D3) glycoprotein that plays a crucial role in innate immunity and inflammation by supporting cell migration and matrix degradation, with structure and biological properties that can be regulated via limited endoproteolysis. With the use of immunoblotting, flow immunocytometry, and ELISA analyses applied to a recombinant uPAR protein and to uPAR-expressing monocytic and human bronchial epithelial cells, it was shown that exposure of uPAR to soluble HAT in the range of 10-500 nM resulted in the proteolytic processing of the full-length (D1D2D3) into the truncated (D2D3) species, with cleavage occurring in the D1 to D2 linker sequence after arginine residues at position 83 and 89. Using immunohistochemistry, we found that both HAT and uPAR were expressed in the human bronchial epithelium. Moreover, transient cotransfection in epithelial cells showed that membrane coexpression of the two partners produced a constitutive and extensive shedding of the D1 domain, occurring for membrane-associated HAT concentrations in the nanomolar range. Because the truncated receptor was found to be unable to bind two of the major uPAR ligands, the adhesive matrix protein vitronectin and the serine protease urokinase, it thus appears that proteolytic regulation of uPAR by HAT is likely to modulate cell adherence and motility, as well as tissue remodeling during the inflammatory response in the airways.
Collapse
|
42
|
Theodoropoulos G, Carraway KL. Molecular signaling in the regulation of mucins. J Cell Biochem 2007; 102:1103-16. [DOI: 10.1002/jcb.21539] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|