10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The serine protease hepsin mediates urinary secretion and polymerisation of Zona Pellucida domain protein uromodulin

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Uromodulin is the most abundant protein in the urine. It is exclusively produced by renal epithelial cells and it plays key roles in kidney function and disease. Uromodulin mainly exerts its function as an extracellular matrix whose assembly depends on a conserved, specific proteolytic cleavage leading to conformational activation of a Zona Pellucida (ZP) polymerisation domain. Through a comprehensive approach, including extensive characterisation of uromodulin processing in cellular models and in specific knock-out mice, we demonstrate that the membrane-bound serine protease hepsin is the enzyme responsible for the physiological cleavage of uromodulin. Our findings define a key aspect of uromodulin biology and identify the first in vivo substrate of hepsin. The identification of hepsin as the first protease involved in the release of a ZP domain protein is likely relevant for other members of this protein family, including several extracellular proteins, as egg coat proteins and inner ear tectorins.

          DOI: http://dx.doi.org/10.7554/eLife.08887.001

          eLife digest

          Several proteins in humans and other animals contain a region called a 'zona pellucida domain'. This domain enables these proteins to associate with each other and form long filaments. Uromodulin is one such protein that was first identified more than fifty years ago. This protein is known to play a role in human diseases such as hypertension and kidney failure, but uromodulin’s biological purpose still remains elusive.

          Uromodulin is only made in the kidney and it is the most abundant protein in the urine of healthy individuals. Uromodulin also contains a so-called 'external hydrophobic patch' that must be removed before the zona pellucida domain can start to form filaments. This hydrophobic patch is removed when uromodulin is cut by an unknown enzyme; this cutting releases the rest of the uromodulin protein from the surface of the cells that line the kidney into the urine.

          Brunati et al. have now tested a panel of candidate enzymes and identified that one called hepsin is able to cut uromodulin. Hepsin is embedded in the cell membrane of the cells that line the kidney. When the level of hepsin was artificially reduced in cells grown in the laboratory, uromodulin remained anchored to the cell surface, its processing was altered and it did not form filaments.

          Brunati et al. next analysed mice in which the gene encoding hepsin had been deleted. While these animals did not have any major defects in their internal organs, they had much lower levels of uromodulin in their urine. Furthermore, this residual urinary protein was not cut properly and it did not assemble into filaments. Thus, these findings reveal that hepsin is the enzyme that is responsible for releasing uromodulin in the urine. This discovery could be exploited to alter the levels of uromodulin release, and further studies using mice lacking hepsin may also help to understand uromodulin’s biological role. Finally, it will be important to understand if hepsin, or a similar enzyme, is also responsible for the release of other proteins containing the zona pellucida domain.

          DOI: http://dx.doi.org/10.7554/eLife.08887.002

          Related collections

          Most cited references57

          • Record: found
          • Abstract: found
          • Article: not found

          NIH Image to ImageJ: 25 years of image analysis.

          For the past 25 years NIH Image and ImageJ software have been pioneers as open tools for the analysis of scientific images. We discuss the origins, challenges and solutions of these two programs, and how their history can serve to advise and inform other software projects.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Dipeptidyl-peptidase IV (CD26)--role in the inactivation of regulatory peptides.

            Dipeptidyl-peptidase IV (DPP IV/CD26) has a dual function as a regulatory protease and as a binding protein. Its role in the inactivation of bioactive peptides was recognized 20 years ago due to its unique ability to liberate Xaa-Pro or Xaa-Ala dipeptides from the N-terminus of regulatory peptides, but further examples are now emerging from in vitro and vivo experiments. Despite the minimal N-terminal truncation by DPP IV, many mammalian regulatory peptides are inactivated--either totally or only differentially--for certain receptor subtypes. Important DPP IV substrates include neuropeptides like neuropeptide Y or endomorphin, circulating peptide hormones like peptide YY, growth hormone-releasing hormone, glucagon-like peptides(GLP)-1 and -2, gastric inhibitory polypeptide as well as paracrine chemokines like RANTES (regulated on activation normal T cell expressed and secreted), stromal cell-derived factor, eotaxin and macrophage-derived chemokine. Based on these findings the potential clinical uses of selective DPP IV inhibitors or DPP IV-resistant analogues, especially for the insulinotropic hormone GLP-1, have been tested to enhance insulin secretion and to improve glucose tolerance in diabetic animals. Thus, DPP IV appears to be a major physiological regulator for some regulatory peptides, neuropeptides, circulating hormones and chemokines.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Multiple loci associated with indices of renal function and chronic kidney disease.

              Chronic kidney disease (CKD) has a heritable component and is an important global public health problem because of its high prevalence and morbidity. We conducted genome-wide association studies (GWAS) to identify susceptibility loci for glomerular filtration rate, estimated by serum creatinine (eGFRcrea) and cystatin C (eGFRcys), and CKD (eGFRcrea < 60 ml/min/1.73 m(2)) in European-ancestry participants of four population-based cohorts (ARIC, CHS, FHS, RS; n = 19,877; 2,388 CKD cases), and tested for replication in 21,466 participants (1,932 CKD cases). We identified significant SNP associations (P < 5 × 10(-8)) with CKD at the UMOD locus, with eGFRcrea at UMOD, SHROOM3 and GATM-SPATA5L1, and with eGFRcys at CST and STC1. UMOD encodes the most common protein in human urine, Tamm-Horsfall protein, and rare mutations in UMOD cause mendelian forms of kidney disease. Our findings provide new insights into CKD pathogenesis and underscore the importance of common genetic variants influencing renal function and disease.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                17 December 2015
                2015
                : 4
                : e08887
                Affiliations
                [1 ]deptDivision of Genetics and Cell Biology , San Raffaele Scientific Institute , Milan, Italy
                [2 ]deptDepartment of Biosciences and Nutrition & Center for Innovative Medicine , Karolinska Institutet , Huddinge, Sweden
                [3 ]deptFunctional Proteomics , FIRC Institute of Molecular Oncology , Milan, Italy
                [4 ]deptProtein Microsequencing Facility , San Raffaele Scientific Institute , Milan, Italy
                [5 ]deptInstitute of Physiology, Zurich Center for Integrative Human Physiology , University of Zurich , Zurich, Switzerland
                [6 ]deptDepartment of Molecular Cardiology , Lerner Research Institute , Cleveland, United States
                [7 ]deptDepartment of Pharmacology and Toxicology , University of Lausanne , Lausanne, Switzerland
                [8]Salk Institute , United States
                [9]Salk Institute , United States
                Author notes
                Article
                08887
                10.7554/eLife.08887
                4755741
                26673890
                e14e3e94-2c73-4915-aad7-35eb3c4d9d69
                © 2015, Brunati et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 24 May 2015
                : 02 November 2015
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100002426, Fondazione Telethon;
                Award ID: TCR08006
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002426, Fondazione Telethon;
                Award ID: GGP14263
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100003196, Ministero della Salute;
                Award ID: RF-2010-2319394
                Award Recipient :
                Funded by: Swiss National Science Foundation;
                Award ID: 31003A-144198/1
                Award Recipient :
                Funded by: National Center of Competence in Research;
                Award ID: Kidney.Ch
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001866, Fonds National de la Recherche Luxembourg;
                Award ID: 6903109
                Award Recipient :
                Funded by: European Commission Seventh Framework Programme (FP7/2007-2013);
                Award ID: 246539 (Marie Curie Actions Programme), 305608 (EURenOmics)
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002661, Fonds De La Recherche Scientifique - FNRS;
                Award Recipient :
                Funded by: Fonds De La Recherche Scientifique Medicale;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100003357, Gebert Rüf Stiftung;
                Award ID: GRS-038/12
                Award Recipient :
                Funded by: Swiss National Science Foundation;
                Award ID: 310030-146490
                Award Recipient :
                Funded by: Swiss National Science Foundation;
                Award ID: 32003B-149309
                Award Recipient :
                Funded by: Swedish Research Council;
                Award ID: 2012-5093
                Award Recipient :
                Funded by: Goran Gustafsson Foundation for Research in Natural Sciences and Medicine;
                Award Recipient :
                Funded by: European Research Council, under the European Union's Seventh Framework Programme (FP7/2007-2013);
                Award ID: ERC 260759
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Biochemistry
                Cell Biology
                Research Article
                Custom metadata
                2.5
                Hepsin is the first protease identified to modulate specific cleavage and secretion of a Zona Pellucida domain protein, uromodulin.

                Life sciences
                zona pellucida domain,uromodulin,hepsin,serine protease,mouse
                Life sciences
                zona pellucida domain, uromodulin, hepsin, serine protease, mouse

                Comments

                Comment on this article