1
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 PMCID: PMC12110317 DOI: 10.3390/cancers17101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| |
Collapse
|
2
|
Modenbach JM, Möller C, Asgarbeik S, Geist N, Rimkus N, Dörr M, Wolfgramm H, Steil L, Susemihl A, Graf L, Schmöker O, Böttcher D, Hammer E, Glaubitz J, Lammers M, Delcea M, Völker U, Aghdassi AA, Lerch MM, Weiss FU, Bornscheuer UT, Sendler M. Biochemical analyses of cystatin-C dimers and cathepsin-B reveals a trypsin-driven feedback mechanism in acute pancreatitis. Nat Commun 2025; 16:1702. [PMID: 39962054 PMCID: PMC11833081 DOI: 10.1038/s41467-025-56875-x] [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: 07/31/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Acute pancreatitis (AP) is characterised by self-digestion of the pancreas by its own proteases. This pathophysiological initiating event in AP occurs inside pancreatic acinar cells where intrapancreatic trypsinogen becomes prematurely activated by cathepsin B (CTSB), and induces the digestive protease cascade, while cathepsin L (CTSL) degrades trypsin and trypsinogen and therefore prevents the development of AP. These proteases are located in the secretory compartment of acinar cells together with cystatin C (CST3), an endogenous inhibitor of CTSB and CTSL. The results are based on detailed biochemical analysis, site-directed mutagenesis and molecular dynamics simulations in combination with an experimental disease model of AP using CST3 deficient mice. This identifies that CST3 is a critical regulator of CTSB and CTSL activity during AP. CST3 deficient mice show a higher intracellular CTSB activity resulting in elevated trypsinogen activation accompanied by an increased disease severity. This reveals that CST3 can be cleaved by trypsin disabling the inhibition of CTSB, but not of CTSL. Furthermore, dimerised CST3 enhances the CTSB activity by binding to an allosteric pocket specific to the CTSB structure. CST3 shifts from an inhibitor to an activator of CTSB and therefore fuels the intrapancreatic protease cascade during the onset of AP.
Collapse
Affiliation(s)
| | - Christina Möller
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Saeedeh Asgarbeik
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Norman Geist
- Department of Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Niklas Rimkus
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Mark Dörr
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Hannes Wolfgramm
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Leif Steil
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Anne Susemihl
- Department of Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
- Department of Medicine C, University Medicine Greifswald, Greifswald, Germany
| | - Leonie Graf
- Department of Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Ole Schmöker
- Department of Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Dominique Böttcher
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Elke Hammer
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Juliane Glaubitz
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Michael Lammers
- Department of Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Mihaela Delcea
- Department of Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Uwe T Bornscheuer
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Greifswald, Germany.
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
3
|
Sehgal ANA, Safran J, Kratzer B, Gattinger P, Stieger RB, Musiejovsky L, Trapin D, Ettel P, Körmöczi U, Rottal A, Borochova K, Dorofeeva Y, Tulaeva I, Weber M, Grabmeier-Pfistershammer K, Perkmann T, Wiedermann U, Valenta R, Pickl WF. Flow Cytometry-Based Measurement of Antibodies Specific for Cell Surface-Expressed Folded SARS-CoV-2 Receptor-Binding Domains. Vaccines (Basel) 2024; 12:377. [PMID: 38675759 PMCID: PMC11053794 DOI: 10.3390/vaccines12040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has now become endemic and is currently one of the important respiratory virus infections regularly affecting mankind. The assessment of immunity against SARS-CoV-2 and its variants is important for guiding active and passive immunization and SARS-CoV-2-specific treatment strategies. METHODS We here devised a novel flow cytometry-based diagnostic platform for the assessment of immunity against cell-bound virus antigens. This platform is based on a collection of HEK-293T cell lines which, as exemplified in our study, stably express the receptor-binding domains (RBDs) of the SARS-CoV-2 S-proteins of eight major SARS-CoV-2 variants, ranging from Wuhan-Hu-1 to Omicron. RESULTS RBD-expressing cell lines stably display comparable levels of RBD on the surface of HEK-293T cells, as shown with anti-FLAG-tag antibodies directed against a N-terminally introduced 3x-FLAG sequence while the functionality of RBD was proven by ACE2 binding. We exemplify the usefulness and specificity of the cell-based test by direct binding of IgG and IgA antibodies of SARS-CoV-2-exposed and/or vaccinated individuals in which the assay shows a wide linear performance range both at very low and very high serum antibody concentrations. In another application, i.e., antibody adsorption studies, the test proved to be a powerful tool for measuring the ratios of individual variant-specific antibodies. CONCLUSION We have established a toolbox for measuring SARS-CoV-2-specific immunity against cell-bound virus antigens, which may be considered as an important addition to the armamentarium of SARS-CoV-2-specific diagnostic tests, allowing flexible and quick adaptation to new variants of concern.
Collapse
Affiliation(s)
- Al Nasar Ahmed Sehgal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Jera Safran
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Pia Gattinger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Robert B. Stieger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Laszlo Musiejovsky
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Paul Ettel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Ulrike Körmöczi
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Arno Rottal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Kristina Borochova
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Yulia Dorofeeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Inna Tulaeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Milena Weber
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| |
Collapse
|
4
|
Khan E, Chakrabarty S, Shariff S, Bardhan M. Genetics and Genomics of Chronic Pancreatitis with a Focus on Disease Biology and Molecular Pathogenesis. Glob Med Genet 2023; 10:324-334. [PMID: 38025192 PMCID: PMC10665123 DOI: 10.1055/s-0043-1776981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Chronic pancreatitis is a long-term fibroinflammatory condition of the pancreas with varying incidences across countries. The recent increase in its occurrence implies the involvement of genetic, hereditary, and unconventional risk factors. However, there is a lack of updated literature on recent advances in genetic polymorphisms of chronic pancreatitis. Therefore, this review aims to present recent findings on the genetic implications of chronic pancreatitis based on individual gene mechanisms and to discuss epigenetics and epistasis involved in the disease. Four mechanisms have been implicated in the pathogenesis of chronic pancreatitis, including premature activation of proteases, endoplasmic reticulum stress, ductal pathway dysfunction, and inflammatory pathway dysfunction. These mechanisms involve genes such as PRSS1, PRSS2, SPINK, CEL, PNLIP, PNLIPRP2, CFTR, CaSR, CLDN2, Alpha 1 antitrypsin, and GGT1 . Studying genetic polymorphisms on the basis of altered genes and their products may aid clinicians in identifying predispositions in patients with and without common risk factors. Further research may also identify associations between genetic predispositions and disease staging or prognosis, leading to personalized treatment protocols and precision medicine.
Collapse
Affiliation(s)
- Erum Khan
- Department of Neurology, Alzheimer's Disease Research Center, The university of Alabama at Birmingham, Birmingham, United States
| | - Soura Chakrabarty
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Mainak Bardhan
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, United States
| |
Collapse
|
5
|
Masson E, Zou WB, Pu N, Rebours V, Génin E, Wu H, Lin JH, Wang YC, Abrantes A, Aguilera Munoz L, Albouys J, Alric L, Amiot X, Archambeaud I, Audiau S, Bastide L, Baudon J, Bellaiche G, Bellon S, Bertrand V, Bideau K, Billiemaz K, Billioud C, Bonnefoy S, Borderon C, Bournet B, Breton E, Brugel M, Buscail L, Cadiot G, Camus M, Causse X, Chamouard P, Chaput U, Cholet F, Ciocan DM, Clavel C, Coffin B, Coimet-Berger L, Creveaux I, Culetto A, Daboussi O, Mestier LDE, Degand T, D'Engremont C, Denis B, Dermine S, Desgrippes R, D'Aubigny AD, Enaud R, Fabre A, Gargot D, Gelsi E, Gentilcore E, Gincul R, Ginglinger-Favre E, Giovannini M, Gomercic C, Gondran H, Grainville T, Grandval P, Grasset D, Grimaldi S, Grimbert S, Hagege H, Heissat S, Hentic O, Herber-Mayne A, Hervouet M, Hoibian S, Jacques J, Jais B, Kaassis M, Koch S, Lacaze E, Lacroute J, Lamireau T, Laurent L, Guillou XLE, Rhun MLE, Leblanc S, Levy P, Lievre A, Lorenzo D, Maire F, Marcel K, Matias C, Mauillon J, Morgant S, Moussata D, Muller N, Nambot S, Napoleon B, Olivier A, Pagenault M, Pelletier AL, Pennec O, Pinard F, Pioche M, Prost B, et alMasson E, Zou WB, Pu N, Rebours V, Génin E, Wu H, Lin JH, Wang YC, Abrantes A, Aguilera Munoz L, Albouys J, Alric L, Amiot X, Archambeaud I, Audiau S, Bastide L, Baudon J, Bellaiche G, Bellon S, Bertrand V, Bideau K, Billiemaz K, Billioud C, Bonnefoy S, Borderon C, Bournet B, Breton E, Brugel M, Buscail L, Cadiot G, Camus M, Causse X, Chamouard P, Chaput U, Cholet F, Ciocan DM, Clavel C, Coffin B, Coimet-Berger L, Creveaux I, Culetto A, Daboussi O, Mestier LDE, Degand T, D'Engremont C, Denis B, Dermine S, Desgrippes R, D'Aubigny AD, Enaud R, Fabre A, Gargot D, Gelsi E, Gentilcore E, Gincul R, Ginglinger-Favre E, Giovannini M, Gomercic C, Gondran H, Grainville T, Grandval P, Grasset D, Grimaldi S, Grimbert S, Hagege H, Heissat S, Hentic O, Herber-Mayne A, Hervouet M, Hoibian S, Jacques J, Jais B, Kaassis M, Koch S, Lacaze E, Lacroute J, Lamireau T, Laurent L, Guillou XLE, Rhun MLE, Leblanc S, Levy P, Lievre A, Lorenzo D, Maire F, Marcel K, Matias C, Mauillon J, Morgant S, Moussata D, Muller N, Nambot S, Napoleon B, Olivier A, Pagenault M, Pelletier AL, Pennec O, Pinard F, Pioche M, Prost B, Queneherve L, Rebours V, Reboux N, Rekik S, Riachi G, Rohmer B, Roquelaure B, Hezode IR, Rostain F, Saurin JC, Servais L, Stan-Iuga R, Subtil C, Texier C, Thomassin L, Tougeron D, Tsakiris L, Valats JC, Vuitton L, Wallenhorst T, Wangerme M, Zanaldi H, Zerbib F. Classification of PRSS1 variants responsible for chronic pancreatitis: An expert perspective from the Franco-Chinese GREPAN study group. Pancreatology 2023; 23:491-506. [PMID: 37581535 DOI: 10.1016/j.pan.2023.04.004] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND PRSS1 was the first reported chronic pancreatitis (CP) gene. The existence of both gain-of-function (GoF) and gain-of-proteotoxicity (GoP) pathological PRSS1 variants, together with the fact that PRSS1 variants have been identified in CP subtypes spanning the range from monogenic to multifactorial, has made the classification of PRSS1 variants very challenging. METHODS All currently reported PRSS1 variants (derived primarily from two databases) were manually reviewed with respect to their clinical genetics, functional analysis and population allele frequency. They were classified by variant type and pathological mechanism within the framework of our recently proposed ACMG/AMP guidelines-based seven-category system. RESULTS The total number of distinct germline PRSS1 variants included for analysis was 100, comprising 3 copy number variants (CNVs), 12 5' and 3' variants, 19 intronic variants, 5 nonsense variants, 1 frameshift deletion variant, 6 synonymous variants, 1 in-frame duplication, 3 gene conversions and 50 missense variants. Based upon a combination of clinical genetic and functional analysis, population data and in silico analysis, we classified 26 variants (all 3 CNVs, the in-frame duplication, all 3 gene conversions and 19 missense) as "pathogenic", 3 variants (missense) as "likely pathogenic", 5 variants (four missense and one promoter) as "predisposing", 13 variants (all missense) as "unknown significance", 2 variants (missense) as "likely benign", and all remaining 51 variants as "benign". CONCLUSIONS We describe an expert classification of the 100 PRSS1 variants reported to date. The results have immediate implications for reclassifying many ClinVar-registered PRSS1 variants as well as providing optimal guidelines/standards for reporting PRSS1 variants.
Collapse
Affiliation(s)
- Emmanuelle Masson
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; Service de Génétique Médicale et de Biologie de la Reproduction, CHRU Brest, F-29200, Brest, France
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Na Pu
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; Department of Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Vinciane Rebours
- Pancreatology and Digestive Oncology Department, Beaujon Hospital, APHP - Clichy, Université Paris Cité, Paris, France
| | - Emmanuelle Génin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; Service de Génétique Médicale et de Biologie de la Reproduction, CHRU Brest, F-29200, Brest, France
| | - Hao Wu
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jin-Huan Lin
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Yuan-Chen Wang
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Marc Hervouet
- Hôpital d'instruction des armées Percy, Clamart, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Nagel F, Susemihl A, Geist N, Möhlis K, Palm GJ, Lammers M, Delcea M. Structural Basis of the Pancreatitis-Associated Autoproteolytic Failsafe Mechanism in Human Anionic Trypsin. J Inflamm Res 2022; 15:3633-3642. [PMID: 35775010 PMCID: PMC9239388 DOI: 10.2147/jir.s367699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
Affiliation(s)
- Felix Nagel
- Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Anne Susemihl
- Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
- Department of Hematology and Oncology, Internal Medicine C, University of Greifswald, Greifswald, Germany
| | - Norman Geist
- Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Kevin Möhlis
- Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Gottfried J Palm
- Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Michael Lammers
- Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Mihaela Delcea
- Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
- Correspondence: Mihaela Delcea, Biophysical Chemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany, Tel +49 3834 420 4423, Fax +49 3834 420 4377, Email
| |
Collapse
|
7
|
Beyer G, Hoffmeister A, Michl P, Gress TM, Huber W, Algül H, Neesse A, Meining A, Seufferlein TW, Rosendahl J, Kahl S, Keller J, Werner J, Friess H, Bufler P, Löhr MJ, Schneider A, Lynen Jansen P, Esposito I, Grenacher L, Mössner J, Lerch MM, Mayerle J. S3-Leitlinie Pankreatitis – Leitlinie der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) – September 2021 – AWMF Registernummer 021-003. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:419-521. [PMID: 35263785 DOI: 10.1055/a-1735-3864] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Georg Beyer
- Medizinische Klinik und Poliklinik II, LMU Klinikum, Ludwig-Maximilians-Universität München, Deutschland
| | - Albrecht Hoffmeister
- Bereich Gastroenterologie, Klinik und Poliklinik für Onkologie, Gastroenterologie, Hepatologie Pneumologie und Infektiologie, Universitätsklinikum Leipzig, Deutschland
| | - Patrick Michl
- Universitätsklinik u. Poliklinik Innere Medizin I mit Schwerpunkt Gastroenterologie, Universitätsklinikum Halle, Deutschland
| | - Thomas Mathias Gress
- Klinik für Gastroenterologie und Endokrinologie, Universitätsklinikum Gießen und Marburg, Deutschland
| | - Wolfgang Huber
- Comprehensive Cancer Center München TUM, II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - Hana Algül
- Comprehensive Cancer Center München TUM, II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - Albrecht Neesse
- Klinik für Gastroenterologie, gastrointestinale Onkologie und Endokrinologie, Universitätsmedizin Göttingen, Deutschland
| | - Alexander Meining
- Medizinische Klinik und Poliklinik II Gastroenterologie und Hepatologie, Universitätsklinikum Würzburg, Deutschland
| | | | - Jonas Rosendahl
- Universitätsklinik u. Poliklinik Innere Medizin I mit Schwerpunkt Gastroenterologie, Universitätsklinikum Halle, Deutschland
| | - Stefan Kahl
- Klinik für Innere Medizin m. Schwerpkt. Gastro./Hämat./Onko./Nephro., DRK Kliniken Berlin Köpenick, Deutschland
| | - Jutta Keller
- Medizinische Klinik, Israelitisches Krankenhaus, Hamburg, Deutschland
| | - Jens Werner
- Klinik für Allgemeine, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Universitätsklinikum München, Deutschland
| | - Helmut Friess
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, München, Deutschland
| | - Philip Bufler
- Klinik für Pädiatrie m. S. Gastroenterologie, Nephrologie und Stoffwechselmedizin, Charité Campus Virchow-Klinikum - Universitätsmedizin Berlin, Deutschland
| | - Matthias J Löhr
- Department of Gastroenterology, Karolinska, Universitetssjukhuset, Stockholm, Schweden
| | - Alexander Schneider
- Klinik für Gastroenterologie und Hepatologie, Klinikum Bad Hersfeld, Deutschland
| | - Petra Lynen Jansen
- Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
| | - Irene Esposito
- Pathologisches Institut, Heinrich-Heine-Universität und Universitätsklinikum Duesseldorf, Duesseldorf, Deutschland
| | - Lars Grenacher
- Conradia Radiologie München Schwabing, München, Deutschland
| | - Joachim Mössner
- Bereich Gastroenterologie, Klinik und Poliklinik für Onkologie, Gastroenterologie, Hepatologie Pneumologie und Infektiologie, Universitätsklinikum Leipzig, Deutschland
| | - Markus M Lerch
- Klinik für Innere Medizin A, Universitätsmedizin Greifswald, Deutschland.,Klinikum der Ludwig-Maximilians-Universität (LMU) München, Deutschland
| | - Julia Mayerle
- Medizinische Klinik und Poliklinik II, LMU Klinikum, Ludwig-Maximilians-Universität München, Deutschland
| | | |
Collapse
|
8
|
Kasuga A, Okamoto T, Udagawa S, Mori C, Mie T, Furukawa T, Yamada Y, Takeda T, Matsuyama M, Sasaki T, Ozaka M, Ueki A, Sasahira N. Molecular Features and Clinical Management of Hereditary Pancreatic Cancer Syndromes and Familial Pancreatic Cancer. Int J Mol Sci 2022; 23:1205. [PMID: 35163129 PMCID: PMC8835700 DOI: 10.3390/ijms23031205] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Hereditary pancreatic cancers are caused by several inherited genes. Familial pancreatic cancer is defined as pancreatic cancer arising in a patient with at least two first-degree relatives with pancreatic cancer in the absence of an identified genetic cause. Hereditary pancreatic cancer syndromes and familial pancreatic cancers account for about 10% of pancreatic cancer cases. Germline mutations in BRCA1, BRCA2, ATM, PALB2, CDKN2A, STK11, and TP53 and mismatch repair genes (MLH1, MSH2, MSH6, PMS2, and EPCAM) are among the well-known inherited susceptibility genes. Currently available targeted medications include poly (ADP-ribose) polymerase inhibitors (PARP) for cases with mutant BRCA and immune checkpoint inhibitors for cases with mismatch repair deficiency. Loss of heterozygosity of hereditary pancreatic cancer susceptibility genes such as BRCA1/2 plays a key role in carcinogenesis and sensitivity to PARP inhibitors. Signature 3 identified by whole genome sequencing is also associated with homologous recombination deficiency and sensitivity to targeted therapies. In this review, we summarize molecular features and treatments of hereditary pancreatic cancer syndromes and surveillance procedures for unaffected high-risk cases. We also review transgenic murine models to gain a better understanding of carcinogenesis in hereditary pancreatic cancer.
Collapse
Affiliation(s)
- Akiyoshi Kasuga
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takeshi Okamoto
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Shohei Udagawa
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan;
| | - Chinatsu Mori
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takafumi Mie
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Yuto Yamada
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Masato Matsuyama
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Arisa Ueki
- Department of Clinical Genetics, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan;
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| |
Collapse
|
9
|
A high prevalence of genetic polymorphisms in idiopathic and alcohol-associated chronic pancreatitis patients in Ireland. HPB (Oxford) 2021; 23:231-237. [PMID: 32669225 DOI: 10.1016/j.hpb.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Individual genetic architecture is considered central to susceptibility and progression of disease in chronic pancreatitis. The study aimed to evaluate the presence of common pancreatic gene mutations in a defined cohort of idiopathic and alcohol-induced chronic pancreatitis patients in Ireland. METHODS The study comprised patients with idiopathic and alcohol-induced chronic pancreatitis and historic controls. Variants in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, cationic trypsinogen (PRSS1) gene and serine protease inhibitor kazal type-1 (SPINK1) gene, were assessed by Taqman© genotyping assay. RESULTS Of n = 126 patients and n = 167 controls, mutations were detected in 23 (20%) and in 10 (6%) respectively (P < 0.001). The majority of mutations found were in the SPINK1 gene variant N34S (13%) which increased disease risk almost six-fold (OR 5.9). Neither CFTR severe mutation (F508del) (P = 0.649) nor mild variant (R117H) (P = 0.327) were over-represented amongst patients compared to control subjects. PRSS1 variants were not detected in either patient or control subjects. CONCLUSION There was a significant prevalence of chronic pancreatitis-associated gene mutations in this well-phenotyped cohort. In patients with alcohol-related or idiopathic chronic pancreatitis, the possibility of genetic mutations in the SPINK 1 gene should be considered as a contributing aetiology factor.
Collapse
|
10
|
Signal peptide of HIV-1 envelope modulates glycosylation impacting exposure of V1V2 and other epitopes. PLoS Pathog 2020; 16:e1009185. [PMID: 33370382 PMCID: PMC7793277 DOI: 10.1371/journal.ppat.1009185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/08/2021] [Accepted: 11/24/2020] [Indexed: 11/19/2022] Open
Abstract
HIV-1 envelope (Env) is a trimer of gp120-gp41 heterodimers, synthesized from a precursor gp160 that contains an ER-targeting signal peptide (SP) at its amino-terminus. Each trimer is swathed by ~90 N-linked glycans, comprising complex-type and oligomannose-type glycans, which play an important role in determining virus sensitivity to neutralizing antibodies. We previously examined the effects of single point SP mutations on Env properties and functions. Here, we aimed to understand the impact of the SP diversity on glycosylation of virus-derived Env and virus neutralization by swapping SPs. Analyses of site-specific glycans revealed that SP swapping altered Env glycan content and occupancy on multiple N-linked glycosites, including conserved N156 and N160 glycans in the V1V2 region at the Env trimer apex and N88 at the trimer base. Virus neutralization was also affected, especially by antibodies against V1V2, V3, and gp41. Likewise, SP swaps affected the recognition of soluble and cell-associated Env by antibodies targeting distinct V1V2 configurations, V3 crown, and gp41 epitopes. These data highlight the contribution of SP sequence diversity in shaping the Env glycan content and its impact on the configuration and accessibility of V1V2 and other Env epitopes. HIV-1 Env glycoprotein is produced by a precursor gp160 that has a signal peptide at its N-terminus. The SP is highly diverse among the HIV-1 isolates. This study presents site-specific analyses of N-linked glycosylation on HIV-1 envelope glycoproteins from infectious viruses produced with different envelope signal peptides. We show that signal peptide swapping alters the envelope glycan shield, including the conserved N156 and N160 glycans located in the V1V2 region on the trimer apex, to impact Env recognition and virus neutralization by antibodies. The data offer crucial insights into the role of signal peptide in the interplay between HIV-1 and antibodies and its potential utility to control Env glycosylation in the development of Env-based HIV-1 vaccine.
Collapse
|
11
|
Weiss FU, Laemmerhirt F, Lerch MM. Next generation sequencing pitfalls in diagnosing trypsinogen (PRSS1) mutations in chronic pancreatitis. Gut 2020; 70:gutjnl-2020-322864. [PMID: 32989020 DOI: 10.1136/gutjnl-2020-322864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 02/04/2023]
Affiliation(s)
- Frank U Weiss
- Medicine A, University Medicine Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Felix Laemmerhirt
- Medicine A, University Medicine Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Markus M Lerch
- Medicine A, University Medicine Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| |
Collapse
|
12
|
Boon L, Ugarte-Berzal E, Vandooren J, Opdenakker G. Protease propeptide structures, mechanisms of activation, and functions. Crit Rev Biochem Mol Biol 2020; 55:111-165. [PMID: 32290726 DOI: 10.1080/10409238.2020.1742090] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteases are a diverse group of hydrolytic enzymes, ranging from single-domain catalytic molecules to sophisticated multi-functional macromolecules. Human proteases are divided into five mechanistic classes: aspartate, cysteine, metallo, serine and threonine proteases, based on the catalytic mechanism of hydrolysis. As a protective mechanism against uncontrolled proteolysis, proteases are often produced and secreted as inactive precursors, called zymogens, containing inhibitory N-terminal propeptides. Protease propeptide structures vary considerably in length, ranging from dipeptides and propeptides of about 10 amino acids to complex multifunctional prodomains with hundreds of residues. Interestingly, sequence analysis of the different protease domains has demonstrated that propeptide sequences present higher heterogeneity compared with their catalytic domains. Therefore, we suggest that protease inhibition targeting propeptides might be more specific and have less off-target effects than classical inhibitors. The roles of propeptides, besides keeping protease latency, include correct folding of proteases, compartmentalization, liganding, and functional modulation. Changes in the propeptide sequence, thus, have a tremendous impact on the cognate enzymes. Small modifications of the propeptide sequences modulate the activity of the enzymes, which may be useful as a therapeutic strategy. This review provides an overview of known human proteases, with a focus on the role of their propeptides. We review propeptide functions, activation mechanisms, and possible therapeutic applications.
Collapse
Affiliation(s)
- Lise Boon
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Jennifer Vandooren
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Role of endoplasmic reticulum stress and protein misfolding in disorders of the liver and pancreas. Adv Med Sci 2019; 64:315-323. [PMID: 30978662 DOI: 10.1016/j.advms.2019.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/17/2018] [Accepted: 03/21/2019] [Indexed: 12/24/2022]
Abstract
The endoplasmic reticulum (ER) is the site of synthesis and folding of membrane and secretory proteins. The fraction of protein passing through the ER represents a large proportion of the total protein in the cell. Protein folding, glycosylation, sorting and transport are essential tasks of the ER and a compromised ER folding network has been recognized to be a key component in the disease pathogenicity of common neurodegenerative, metabolic and malignant diseases. On the other hand, the ER protein folding machinery also holds significant potential for therapeutic interventions. Many causes can lead to ER stress. A disturbed calcium homeostasis, the generation of reactive oxygen species (ROS) and a persistent overload of misfolded proteins within the ER can drive the course of adisease. In this review the role of ER-stress in diseases of the liver and pancreas will be examined using pancreatitis and Wilson´s disease as examples. Potential therapeutic targets in ER-stress pathways will also be discussed.
Collapse
|
14
|
Silencing of Aberrant Secretory Protein Expression by Disease-Associated Mutations. J Mol Biol 2019; 431:2567-2580. [PMID: 31100385 DOI: 10.1016/j.jmb.2019.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 11/23/2022]
Abstract
Signal recognition particle (SRP) recognizes signal sequences of secretory proteins and targets them to the endoplasmic reticulum membrane for translocation. Many human diseases are connected with defects in signal sequences. The current dogma states that the molecular basis of the disease-associated mutations in the secretory proteins is connected with defects in their transport. Here, we demonstrate for several secretory proteins with disease-associated mutations that the molecular mechanism is different from the dogma. Positively charged or helix-breaking mutations in the signal sequence hydrophobic core prevent synthesis of the aberrant proteins and lead to degradation of their mRNAs. The degree of mRNA depletion depends on the location and severity of the mutation in the signal sequence and correlates with inhibition of SRP interaction. Thus, SRP protects secretory protein mRNAs from degradation. The data demonstrate that if disease-associated mutations obstruct SRP interaction, they lead to silencing of the mutated protein expression.
Collapse
|
15
|
Wierbowski SD, Fragoza R, Liang S, Yu H. Extracting Complementary Insights from Molecular Phenotypes for Prioritization of Disease-Associated Mutations. CURRENT OPINION IN SYSTEMS BIOLOGY 2018; 11:107-116. [PMID: 31086831 PMCID: PMC6510504 DOI: 10.1016/j.coisb.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rapid advances in next-generation sequencing technology have resulted in an explosion of whole-exome/genome sequencing data, providing an unprecedented opportunity to identify disease- and trait-associated variants in humans on a large scale. To date, the long-standing paradigm has leveraged fitness-based approximations to translate this ever-expanding sequencing data into causal insights in disease. However, while this approach robustly identifies variants under evolutionary constraint, it fails to provide molecular insights. Moreover, complex disease phenomena often violate standard assumptions of a direct organismal phenotype to overall fitness effect relationship. Here we discuss the potential of a molecular phenotype-oriented paradigm to uniquely identify candidate disease-causing mutations from the human genetic background. By providing a direct connection between single nucleotide mutations and observable organismal and cellular phenotypes associated with disease, we suggest that molecular phenotypes can readily incorporate alongside established fitness-based methodologies to provide complementary insights to the functional impact of human mutations. Lastly, we discuss how integrated approaches between molecular phenotypes and fitness-based perspectives facilitate new insights into the molecular mechanisms underlying disease-associated mutations while also providing a platform for improved interpretation of epistasis in human disease.
Collapse
Affiliation(s)
- Shayne D. Wierbowski
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Robert Fragoza
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Siqi Liang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Haiyuan Yu
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
16
|
PRSS1 (R122H) mutation in an Indian family with low penetrance is associated with pancreatitis phenotype. Indian J Gastroenterol 2018; 37:67-69. [PMID: 29476405 DOI: 10.1007/s12664-018-0828-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/10/2018] [Indexed: 02/04/2023]
Abstract
Mutations in PRSS1 gene namely R122H and N29I cause hereditary pancreatitis. They are autosomal dominant with a high penetrance (80%) reported in North American, North-east Asian, and North European ethnicities. However, the mutations are reportedly absent in Indian, African, and South American ethnicities. We report here for the first time a family from India that is positive for R122H mutation in the PRSS1 gene. The proband is symptomatic with chronic pancreatitis, however, the father although heterozygous for R122H is asymptomatic.
Collapse
|
17
|
Xiao Y, Yuan W, Yu B, Guo Y, Xu X, Wang X, Yu Y, Yu Y, Gong B, Xu C. Targeted Gene Next-Generation Sequencing in Chinese Children with Chronic Pancreatitis and Acute Recurrent Pancreatitis. J Pediatr 2017; 191:158-163.e3. [PMID: 29173301 DOI: 10.1016/j.jpeds.2017.08.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/24/2017] [Accepted: 08/23/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To identify causal mutations in certain genes in children with acute recurrent pancreatitis (ARP) or chronic pancreatitis (CP). STUDY DESIGN After patients were enrolled (CP, 55; ARP, 14) and their clinical characteristics were investigated, we performed next-generation sequencing to detect nucleotide variations among the following 10 genes: cationic trypsinogen protease serine 1 (PRSS1), serine protease inhibitor, Kazal type 1 (SPINK1), cystic fibrosis transmembrane conductance regulator gene (CFTR), chymotrypsin C (CTRC), calcium-sensing receptor (CASR), cathepsin B (CTSB), keratin 8 (KRT8), CLAUDIN 2 (CLDN2), carboxypeptidase A1 (CPA1), and ATPase type 8B member 1 (ATP8B1). Mutations were searched against online databases to obtain information on the cause of the diseases. Certain novel mutations were analyzed using the SIFT2 and Polyphen-2 to predict the effect on protein function. RESULTS There were 45 patients with CP and 10 patients with ARP who harbored 1 or more mutations in these genes; 45 patients had at least 1 mutation related to pancreatitis. Mutations were observed in the PRSS1, SPINK1, and CFTR genes in 17 patients, the CASR gene in 5 patients, and the CTSB, CTRC, and KRT8 genes in 1 patient. Mutations were not found in the CLDN, CPA1, or ATP8B1 genes. We found that mutations in SPINK1 may increase the risk of pancreatic duct stones (OR, 11.07; P = .003). The patients with CFTR mutations had a higher level of serum amylase (316.0 U/L vs 92.5 U/L; P = .026). CONCLUSION Mutations, especially those in PRSS1, SPINK1, and CFTR, accounted for the major etiologies in Chinese children with CP or ARP. Children presenting mutations in the SPINK1 gene may have a higher risk of developing pancreatic duct stones.
Collapse
Affiliation(s)
- Yuan Xiao
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Wentao Yuan
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Bo Yu
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yan Guo
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xu Xu
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinqiong Wang
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yi Yu
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yi Yu
- Pediatric Department, Ruijin Hospital North, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Biao Gong
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Chundi Xu
- Pediatric Department, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; Pediatric Department, Ruijin Hospital North, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Abstract
Genetic investigations have provided unique insight into the mechanism of chronic pancreatitis in humans and firmly established that uncontrolled trypsin activity is a central pathogenic factor. Mutations in the PRSS1, SPINK1, and CTRC genes promote increased activation of trypsinogen to trypsin by stimulation of autoactivation or by impairing protective trypsinogen degradation and/or trypsin inhibition. Here we review key genetic and biochemical features of the trypsin-dependent pathological pathway in chronic pancreatitis.
Collapse
Affiliation(s)
- Eszter Hegyi
- Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA, 02118, USA
| | - Miklós Sahin-Tóth
- Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA, 02118, USA.
| |
Collapse
|
19
|
Polonikov AV, Samgina TA, Nazarenko PM, Bushueva OY, Ivanov VP. Alcohol Consumption and Cigarette Smoking are Important Modifiers of the Association Between Acute Pancreatitis and the PRSS1-PRSS2 Locus in Men. Pancreas 2017; 46:230-236. [PMID: 27846138 DOI: 10.1097/mpa.0000000000000729] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The present study was designed to investigate whether the susceptibility to acute pancreatitis (AP) attributable to polymorphism rs10273639 at the PRSS1-PRSS2 locus is dependent on alcohol consumption and cigarette smoking. METHODS A total of 603 unrelated Russian individuals including 304 patients with physician-diagnosed AP and 299 sex- and age-matched healthy controls have been recruited for the study. A polymorphism rs10273639 (-408C>T) of PRSS1-PRSS2 was genotyped by TaqMan-based assay. RESULTS A variant allele -408T (P = 0.003) and genotypes -408CT plus TT (P = 0.002) were associated with decreased AP risk only in men. The odds ratios for AP in the CC homozygotes versus the variant genotypes were 1.95 [95% confidence interval (CI), 0.65-5.85; P = 0.23], 1.72 (95% CI, 0.93-3.20; P = 0.08), and 2.37 (95% CI, 1.09-5.13; P = 0.03) for men who consumed up to 28, 29 to 59, and more than 60 alcohol drinks a week, respectively. Cigarette smokers with the -408CC genotype had an increased risk of AP (odds ratio, 2.07; 95% CI, 1.25-3.42; P = 0.004), whereas nonsmoker carriers did not have a disease risk (odds ratio, 1.48; 95% CI, 0.58-3.82; P = 0.42). CONCLUSIONS We confirmed a robust association of polymorphism rs10273639 at PRSS1-PRSS2 with AP in the Russian population. The present study is the first to show that relationship between the locus and disease is significantly modified by alcohol consumption and cigarette smoking.
Collapse
Affiliation(s)
- Alexey V Polonikov
- From the *Department of Biology, Medical Genetics and Ecology, †Research Institute for Genetic and Molecular Epidemiology, ‡Department of Surgical Diseases №2, Kursk State Medical University, Kursk, Russian Federation; and §Laboratory of Genetics, Kursk State University, Kursk, Russian Federation
| | | | | | | | | |
Collapse
|
20
|
Sathiyasekaran M, Biradar V, Ramaswamy G, Srinivas S, Ashish B, Sumathi B, Nirmala D, Geetha M. Pancreatitis in Children. Indian J Pediatr 2016; 83:1459-1472. [PMID: 27271883 DOI: 10.1007/s12098-016-2115-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 04/13/2016] [Indexed: 12/12/2022]
Abstract
Pancreatic disease in children has a wide clinical spectrum and may present as Acute pancreatitis (AP), Acute recurrent pancreatitis (ARP), Chronic pancreatitis (CP) and Pancreatic disease without pancreatitis. This article highlights the etiopathogenesis and management of pancreatitis in children along with clinical data from five tertiary care hospitals in south India [Chennai (3), Cochin and Pune].
Collapse
Affiliation(s)
- Malathi Sathiyasekaran
- Department of Pediatric Gastroenterology, Kanchi Kamakoti CHILDS Trust Hospital, Apollo & SMF Hospitals, Chennai, India
| | - Vishnu Biradar
- Department of Pediatric Gastroenterology, Deenanath Mangeshkar Hospital, Pune, Maharashtra, India
| | - Ganesh Ramaswamy
- Department of Pediatrics, Kanchi Kamakoti CHILDS Trust Hospital & CHILDS Trust Medical Research Foundation, Chennai-34, India.
| | - S Srinivas
- Department of Pediatric Gastroenterology, Kanchi Kamakoti CHILDS Trust Hospital, Apollo & SMF Hospitals, Chennai, India
| | - B Ashish
- Department of Pediatrics, Kanchi Kamakoti CHILDS Trust Hospital & CHILDS Trust Medical Research Foundation, Chennai-34, India
| | - B Sumathi
- Department of Pediatric Gastroenterology, Institute of Child Health & Hospital for Children, Egmore, Chennai, India
| | - D Nirmala
- Department of Pediatric Gastroenterology, Institute of Child Health & Hospital for Children, Egmore, Chennai, India
| | - M Geetha
- Department of Pediatric Gastroenterology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| |
Collapse
|
21
|
Abstract
OBJECTIVES Causes of acute recurrent pancreatitis (ARP) or chronic pancreatitis (CP) are sometimes difficult to determine in children. In such patients, genetic analysis may prove helpful. The present study analyzed mutations of cationic trypsinogen (PRSS1), serine protease inhibitor Kazal type 1 (SPINK1), chymotrypsin C (CTRC), and carboxypeptidase A1 (CPA1) and investigated the clinical features of children with these mutations. METHODS Genetic analyses of mutations in these 4 genes were conducted in 128 patients with ARP or CP. Characteristics of the patients showing mutations were investigated using medical records. RESULTS Fifty of the 128 (39.1%) subjects had at least 1 mutation (median age at onset, 7.6 years). Abdominal pain was the presenting symptom of pancreatitis in 48 of the 50 patients (96%). Fifteen of those 50 patients (30.0%) had a family history of pancreatitis. Gene mutations were present in PRSS1 in 26 patients, SPINK1 in 23, CTRC in 3, and CPA1 in 5. In the 31 patients with mutations in SPINK1, CTRC, or CPA1, 16 (51.6%) had homozygous or heterozygous mutations with other mutations. Three patients underwent surgery and another 4 patients underwent endoscopy to manage ARP or CP. Although 3 of the 7 patients complained of mild abdominal pain, none of those 7 patients experienced any obvious episode of ARP after treatment. CONCLUSIONS In pediatric patients with idiopathic ARP and CP, genetic analysis is useful for identifying the cause of pancreatitis. Early endoscopic or surgical treatment prevents ARP by extending the interval between episodes of pancreatitis in this population.
Collapse
|
22
|
Abstract
Hereditary pancreatitis (HP) is a rare cause of acute, recurrent acute, and chronic pancreatitis. It may present similarly to other causes of acute and chronic pancreatitis, and often there has been a protracted evaluation prior to the diagnosis of HP. Since it was first described in 1952, multiple genetic defects that affect the action of digestive enzymes in the pancreas have been implicated. The most common mutations involve the PRSS1, CFTR, SPINK1, and CTRC genes. New mutations in these genes and previously unrecognized mutations in other genes are being discovered due to the increasing use of next-generation genomic sequencing. While the inheritance pathways of these genetic mutations may be variable and complex, sometimes involving coinheritance of other mutations, the clinical presentation of patients tends to be similar. Interactions with environmental triggers often play a role. Patients tend to present at an early age (prior to the second decade of life) and have a significantly increased risk for the development of pancreatic adenocarcinoma. Patients with HP may develop sequelae of chronic pancreatitis such as strictures and fluid collections as well as exocrine and endocrine insufficiency. Management of patients with HP involves avoidance of environmental triggers, surveillance for pancreatic adenocarcinoma, medical therapy for endocrine and exocrine insufficiency, pain management, and endoscopic or surgical treatment for complications. Care for affected patients should be individualized, with an emphasis on early diagnosis and multidisciplinary involvement to develop a comprehensive treatment strategy.
Collapse
Affiliation(s)
- Kara L Raphael
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Field F Willingham
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
23
|
Common Variants in CLDN2 and MORC4 Genes Confer Disease Susceptibility in Patients with Chronic Pancreatitis. PLoS One 2016; 11:e0147345. [PMID: 26820620 PMCID: PMC4731142 DOI: 10.1371/journal.pone.0147345] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/31/2015] [Indexed: 01/09/2023] Open
Abstract
A recent genome-wide association study (GWAS) identified association with variants in X-linked CLDN2 and MORC4, and PRSS1-PRSS2 loci with chronic pancreatitis (CP) in North American patients of European ancestry. We selected 9 variants from the reported GWAS and replicated the association with CP in Indian patients by genotyping 1807 unrelated Indians of Indo-European ethnicity, including 519 patients with CP and 1288 controls. The etiology of CP was idiopathic in 83.62% and alcoholic in 16.38% of 519 patients. Our study confirmed a significant association of 2 variants in CLDN2 gene (rs4409525-OR 1.71, P = 1.38 x 10-09; rs12008279-OR 1.56, P = 1.53 x 10-04) and 2 variants in MORC4 gene (rs12688220-OR 1.72, P = 9.20 x 10-09; rs6622126-OR 1.75, P = 4.04x10-05) in Indian patients with CP. We also found significant association at PRSS1-PRSS2 locus (OR 0.60; P = 9.92 x 10-06) and SAMD12-TNFRSF11B (OR 0.49, 95% CI [0.31-0.78], P = 0.0027). A variant in the gene MORC4 (rs12688220) showed significant interaction with alcohol (OR for homozygous and heterozygous risk allele -14.62 and 1.51 respectively, P = 0.0068) suggesting gene-environment interaction. A combined analysis of the genes CLDN2 and MORC4 based on an effective risk allele score revealed a higher percentage of individuals homozygous for the risk allele in CP cases with 5.09 fold enhanced risk in individuals with 7 or more effective risk alleles compared with individuals with 3 or less risk alleles (P = 1.88 x 10-14). Genetic variants in CLDN2 and MORC4 genes were associated with CP in Indian patients.
Collapse
|
24
|
High Penetrance of the PRSS1 A16V Mutation in a Kindred With SPINK1 N34S and CFTR TG11-5T Co-mutations. Pancreas 2016; 45:e2-4. [PMID: 26658045 DOI: 10.1097/mpa.0000000000000445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
25
|
Genetic susceptibility factors for alcohol-induced chronic pancreatitis. Pancreatology 2015; 15:S23-31. [PMID: 26149858 DOI: 10.1016/j.pan.2015.05.476] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/10/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Abstract
Chronic pancreatitis is a progressive inflammatory disease of the pancreas and frequently associated with immoderate alcohol consumption. Since only a small proportion of alcoholics eventually develop chronic pancreatitis genetic susceptibility factors have long been suspected to contribute to the pathogenesis of the disease. Smaller studies in ethnically defined populations have found that not only polymorphism in proteins involved in the metabolism of ethanol, such as Alcohol Dehydrogenase and Aldehyde Dehydrogenase, can confer a risk for developing chronic pancreatitis but also mutations that had previously been reported in association with idiopathic pancreatitis, such as SPINK1 mutations. In a much broader approach employing genome wide search strategies the NAPS study found that polymorphisms in the Trypsin locus (PRSS1 rs10273639), and the Claudin 2 locus (CLDN2-RIPPLY1-MORC4 locus rs7057398 and rs12688220) confer an increased risk of developing alcohol-induced pancreatitis. These results from North America have now been confirmed by a European consortium. In another genome wide approach polymorphisms in the genes encoding Fucosyltransferase 2 (FUT2) non-secretor status and blood group B were not only found in association with higher serum lipase levels in healthy volunteers but also to more than double the risk for developing alcohol-associated chronic pancreatitis. These novel genetic associations will allow to investigate the pathophysiological and biochemical basis of alcohol-induced chronic pancreatitis on a cellular level and in much more detail than previously possible.
Collapse
|
26
|
Koziel D, Gluszek S, Kowalik A, Chlopek M, Pieciak L. Genetic mutations in SPINK1, CFTR, CTRC genes in acute pancreatitis. BMC Gastroenterol 2015; 15:70. [PMID: 26100556 PMCID: PMC4476205 DOI: 10.1186/s12876-015-0302-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 06/16/2015] [Indexed: 12/29/2022] Open
Abstract
Background Explanation of the ultimate causes of acute and chronic pancreatitis is challenging. Hence, it is necessary to seek various etiological factors, including genetic mutations that may be of importance in triggering recurrence and progression of acute to chronic pancreatitis. The aim of this study was to determine the frequency of genetic mutations in patients with acute pancreatitis and to investigate their relationship with the etiology and clinical course. Methods The study included 221 patients treated for acute pancreatitis and 345 healthy subjects as a control group. Peripheral blood samples were collected from each study participant and genomic DNA was isolated. Genotyping of common mutations in the SPINK1 (p.N34S and p.P55S) and CTRC (p.I259V, p.V235I, p.K247_R254del, p.E225A) genes was performed using the high-resolution melting method. Mutations in the CFTR p.F508del (delF508_CTT) were genotyped using allele-specific amplification polymerase chain reaction. All detected mutations were confirmed with direct capillary DNA sequencing. Results Mutations in SPINK 1, CFTR and CTRC were detected in 6.3 %, 2.3 % and 1.8 % of patients with acute pancreatitis versus 3.2 %, 3.8 % and 1.2 % of volunteers in the control group. No relationship was found between the detected mutations and severity of acute pancreatitis: mild acute pancreatitis, mutation of CFTR in 4 (2.8 %) and CTRC in 2 (1.4 %) patients; severe acute pancreatitis, mutation of CFTR and CTRC in 1 (2.6 %) case each. The SPINK1 mutation was significantly more frequent in 8 (10.4 %) severe cases than in 6 (4.2 %) mild cases (P < 0.05), and was observed in 5/70 (7.1 %) patients with alcohol-related AP, 5/81 (6.2 %) with biliary AP, and 4/63 (6.3 %) in those without any established cause of the disease. Conclusions Mutation p.N34S in SPINK1 may predispose patients to acute pancreatitis, especially in those abusing alcohol, and may promote a more severe course of the disease.
Collapse
Affiliation(s)
- Dorota Koziel
- Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland.
| | - Stanislaw Gluszek
- Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland. .,Clinic General Oncological and Endocrinological Surgery, Regional Hospital, Kielce, Poland.
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland.
| | - Malgorzata Chlopek
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland.
| | - Liliana Pieciak
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland.
| |
Collapse
|
27
|
Evaluating Adults With Idiopathic Pancreatitis for Genetic Predisposition: Higher Prevalence of Abnormal Results With Use of Complete Gene Sequencing. Pancreas 2015; 44:116-21. [PMID: 25251442 PMCID: PMC4262640 DOI: 10.1097/mpa.0000000000000225] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES In adults with unexplained pancreatitis, the yield of complete gene versus select exosome sequencing on mutation detection and distinguishing clinical characteristics associated with mutations requires clarification. We sought to (1) compare frequency of mutations identified using different techniques and (2) compare clinical characteristics between adults with and without mutations. METHODS This is a cohort study of adults with unexplained pancreatitis who underwent genetic testing between January 2008 and December 2012. We compare probabilities of having a positive mutation with complete gene sequencing versus alternatives and describe differences in characteristics among patients with and without mutations. RESULTS Of the 370 patients, 67 (18%) had a genetic mutation; 24 (6%) were of high risk. Mutations were significantly more prevalent with use of complete sequencing (42%) versus other approaches (8%, P < 0.0001). Most (44/67, 66%) with a mutation had no family history. Those with high-risk mutations were more likely to have a family history of chronic pancreatitis (21% vs 4%, P = 0.002). Patients with pancreas divisum were more likely to have mutations (27% vs 14%, P = 0.0007). CONCLUSION Among individuals with adult-onset pancreatic disease, the probability of finding any mutation, including high risk, is significantly higher using complete gene sequencing. The impact on patients and providers requires further investigation.
Collapse
|
28
|
Németh BC, Sahin-Tóth M. Human cationic trypsinogen (PRSS1) variants and chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2014; 306:G466-73. [PMID: 24458023 PMCID: PMC3949028 DOI: 10.1152/ajpgi.00419.2013] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Variations in the serine protease 1 (PRSS1) gene encoding human cationic trypsinogen have been conclusively associated with autosomal dominant hereditary pancreatitis and sporadic nonalcoholic chronic pancreatitis. Most high-penetrance PRSS1 variants increase intrapancreatic trypsin activity by stimulating trypsinogen autoactivation and/or by inhibiting chymotrypsin C-dependent trypsinogen degradation. Alternatively, some PRSS1 variants can cause trypsinogen misfolding, which results in intracellular retention and degradation with consequent endoplasmic reticulum stress. However, not all PRSS1 variants are pathogenic, and clinical relevance of rare variants is often difficult to ascertain. Here we review the PRSS1 variants published since 1996 and discuss their functional properties and role in chronic pancreatitis.
Collapse
Affiliation(s)
- Balázs Csaba Németh
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts
| |
Collapse
|
29
|
Brock C, Nielsen LM, Lelic D, Drewes AM. Pathophysiology of chronic pancreatitis. World J Gastroenterol 2013; 19:7231-7240. [PMID: 24259953 PMCID: PMC3831204 DOI: 10.3748/wjg.v19.i42.7231] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/09/2013] [Accepted: 08/29/2013] [Indexed: 02/06/2023] Open
Abstract
Chronic pancreatitis (CP) is an inflammatory disease of the pancreas characterized by progressive fibrotic destruction of the pancreatic secretory parenchyma. Despite the heterogeneity in pathogenesis and involved risk factors, processes such as necrosis/apoptosis, inflammation or duct obstruction are involved. This fibrosing process ultimately leads to progressive loss of the lobular morphology and structure of the pancreas, deformation of the large ducts and severe changes in the arrangement and composition of the islets. These conditions lead to irreversible morphological and structural changes resulting in impairment of both exocrine and endocrine functions. The prevalence of the disease is largely dependent on culture and geography. The etiological risk-factors associated with CP are multiple and involve both genetic and environmental factors. Throughout this review the M-ANNHEIM classification system will be used, comprising a detailed description of risk factors such as: alcohol-consumption, nicotine-consumption, nutritional factors, hereditary factors, efferent duct factors, immunological factors and miscellaneous and rare metabolic factors. Increased knowledge of the different etiological factors may encourage the use of further advanced diagnostic tools, which potentially will help clinicians to diagnose CP at an earlier stage. However, in view of the multi factorial disease and the complex clinical picture, it is not surprising that treatment of patients with CP is challenging and often unsuccessful.
Collapse
|
30
|
Witt H, Beer S, Rosendahl J, Chen JM, Chandak GR, Masamune A, Bence M, Szmola R, Oracz G, Macek M, Bhatia E, Steigenberger S, Lasher D, Bühler F, Delaporte C, Tebbing J, Ludwig M, Pilsak C, Saum K, Bugert P, Masson E, Paliwal S, Bhaskar S, Sobczynska-Tomaszewska A, Bak D, Balascak I, Choudhuri G, Reddy DN, Rao GV, Thomas V, Kume K, Nakano E, Kakuta Y, Shimosegawa T, Durko L, Szabó A, Schnúr A, Hegyi P, Rakonczay Z, Pfützer R, Schneider A, Groneberg DA, Braun M, Schmidt H, Witt U, Friess H, Algül H, Landt O, Schuelke M, Krüger R, Wiedenmann B, Schmidt F, Zimmer KP, Kovacs P, Stumvoll M, Blüher M, Müller T, Janecke A, Teich N, Grützmann R, Schulz HU, Mössner J, Keim V, Löhr M, Férec C, Sahin-Tóth M. Variants in CPA1 are strongly associated with early onset chronic pancreatitis. Nat Genet 2013; 45:1216-1220. [PMID: 23955596 PMCID: PMC3909499 DOI: 10.1038/ng.2730] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/23/2013] [Indexed: 12/12/2022]
Abstract
Chronic pancreatitis is an inflammatory disorder of the pancreas. We analyzed CPA1, encoding carboxypeptidase A1, in subjects with nonalcoholic chronic pancreatitis (cases) and controls in a German discovery set and three replication sets. Functionally impaired variants were present in 29/944 (3.1%) German cases and 5/3,938 (0.1%) controls (odds ratio (OR) = 24.9, P = 1.5 × 10(-16)). The association was strongest in subjects aged ≤ 10 years (9.7%; OR = 84.0, P = 4.1 × 10(-24)). In the replication sets, defective CPA1 variants were present in 8/600 (1.3%) cases and 9/2,432 (0.4%) controls from Europe (P = 0.01), 5/230 (2.2%) cases and 0/264 controls from India (P = 0.02) and 5/247 (2.0%) cases and 0/341 controls from Japan (P = 0.013). The mechanism by which CPA1 variants confer increased pancreatitis risk may involve misfolding-induced endoplasmic reticulum stress rather than elevated trypsin activity, as is seen with other genetic risk factors for this disease.
Collapse
Affiliation(s)
- Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Department of Pediatrics, Klinikum rechts der Isar (MRI), Technische Universität München (TUM), Parzivalstr. 16, 80804 Munich, Germany
| | - Sebastian Beer
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Jonas Rosendahl
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Giriraj Ratan Chandak
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Melinda Bence
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Richárd Szmola
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
- 2nd Department of Medicine, Semmelweis University, Szentkirályi u. 46, H-1088, Budapest, Hungary
| | - Grzegorz Oracz
- Department of Gastroenterology, Hepatology and Immunology, The Children’s Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Milan Macek
- Department of Biology and Medical Genetics, University Hospital Motol & 2nd Faculty of Medicine of Charles University Prague, V Uvalu 84, Prague, CZ 150 06, Czech Republic
| | - Eesh Bhatia
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Sandra Steigenberger
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Denise Lasher
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Florence Bühler
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Catherine Delaporte
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Johanna Tebbing
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Maren Ludwig
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Claudia Pilsak
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Karolin Saum
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg - Hessen, Friedrich-Ebert-Str. 107, 68167 Mannheim, Germany
| | - Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Sumit Paliwal
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Seema Bhaskar
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Agnieszka Sobczynska-Tomaszewska
- MedGen Health Care Centre, Orzycka 27, 02-695 Warsaw, Poland
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Daniel Bak
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Ivan Balascak
- Clinic of Obstetrics and Gynecology, Neonatology Department, University Hospital Motol & 2 Medical School, Charles University Prague, V Uvalu 84, 103 Prague 5, Czech Republic
| | - Gourdas Choudhuri
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - D. Nageshwar Reddy
- Asian Institute of Gastroenterology, 6-3-661, Somajiguda, Hyderabad 500 082, India
| | - G. Venkat Rao
- Asian Institute of Gastroenterology, 6-3-661, Somajiguda, Hyderabad 500 082, India
| | - Varghese Thomas
- Department of Gastroenterology, Medical College Hospital, Calicut 673008, India
| | - Kiyoshi Kume
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Eriko Nakano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Yoichi Kakuta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Lukasz Durko
- Department of Digestive Tract Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| | - András Szabó
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Andrea Schnúr
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Péter Hegyi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Zoltán Rakonczay
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Roland Pfützer
- Department of Internal Medicine, Klinikum Döbeln, Sörmitzer Str. 10, 04720 Döbeln, Germany
| | - Alexander Schneider
- Department of Medicine II, Universitätsmedizin Mannheim, Ruprecht-Karls-Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - David Alexander Groneberg
- Institute for Occupational Medicine, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, Haus 9B, 60590 Frankfurt, Germany
| | - Markus Braun
- Institute for Occupational Medicine, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, Haus 9B, 60590 Frankfurt, Germany
| | - Hartmut Schmidt
- Department for Transplant Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Ulrike Witt
- Department of Surgery, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Hana Algül
- Department of Gastroenterology, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Olfert Landt
- TIB MOLBIOL, Eresburgstr. 22-23, 12103 Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
- NeuroCure Clinical Research Center, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Renate Krüger
- Department of Pediatrics, Division of Pediatric Pulmonology, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Bertram Wiedenmann
- Department of Internal Medicine, Division of Hepatology and Gastroenterology, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Frank Schmidt
- Department of Pediatrics, University of Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle/Saale, Germany
| | - Klaus-Peter Zimmer
- Department of Pediatrics, Justus-Liebig-Universität, Feulgenstr. 12, 35385 Gießen, Germany
| | - Peter Kovacs
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Michael Stumvoll
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Matthias Blüher
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Thomas Müller
- Universitätsklinik für Pädiatrie I, Department für Kinder-und Jugendheilkunde, Medizinische Universität Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Andreas Janecke
- Universitätsklinik für Pädiatrie I, Department für Kinder-und Jugendheilkunde, Medizinische Universität Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
- Sektion für Humangenetik, Medizinische Universität Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Niels Teich
- Practice for Digestive and Metabolic Diseases, Nordstr. 21, 04105 Leipzig, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Joachim Mössner
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Volker Keim
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| |
Collapse
|
31
|
Wang W, Sun XT, Weng XL, Zhou DZ, Sun C, Xia T, Hu LH, Lai XW, Ye B, Liu MY, Jiang F, Gao J, Bo LM, Liu Y, Liao Z, Li ZS. Comprehensive screening for PRSS1, SPINK1, CFTR, CTRC and CLDN2 gene mutations in Chinese paediatric patients with idiopathic chronic pancreatitis: a cohort study. BMJ Open 2013; 3:e003150. [PMID: 24002981 PMCID: PMC3773632 DOI: 10.1136/bmjopen-2013-003150] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/24/2013] [Accepted: 07/26/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Genetic alterations may contribute to chronic pancreatitis (CP) in Chinese young patients. This study was designed to investigate mutations of cationic trypsinogen (PRSS1), pancreatic secretory trypsin inhibitor or serine protease inhibitor Kazal type 1 (SPINK1), cystic fibrosis transmembrane conductance regulator (CFTR), chymotrypsin C (CTRC) and CLDN2 genes and the copy number variations (CNVs) of PRSS1 and asses associations with the development of idiopathic CP (ICP) in Chinese children. DESIGN Retrospective. SETTING A single center. PARTICIPANTS 75 ICP Chinese children (40 boys and 35 girls). PRIMARY AND SECONDARY OUTCOME MEASURES Mutations of PRSS1, SPINK1, CFTR, CTRC and CLDN2 genes and CNVs. RESULTS 7 patients had heterozygous mutations in PRSS1, that is, N29I (n=1), R122H or R122C (n=6). The CNVs of PRSS1 in five patients had abnormal copies (1 copy (n=4), five copies (n=1)). 43 patients had IVS3+2T>C (rs148954387) (10 homozygous and 33 heterozygous) in SPINK1. None of the PRSS1 mutation patients carried a SPINK1 mutation. Frequency of PRSS1 and SPINK1 mutations was 9.3% and 57.3%, respectively, with an overall frequency of 66.6% (50/75). In addition, one patient had a novel deletion of CFTR (GCTTCCTA from c.500 to c.508 leading to the shortened polypeptide molecule via a stop codon). Another patient had a novel missense in CLDN2 exon 2 (c.592A>C mutation). Clinically, patients with SPINK1 mutations had a higher rate of pancreatic duct stones, pancreatic pseudocyst and pancreatic calcification than those without SPINK1 mutations (p<0.05). CONCLUSIONS SPINK1 mutations were more commonly associated with Chinese children with ICP. SPINK1 IVS3+2T>C mutation may play an important role in the pathogenesis of Chinese paediatric ICP. However, further study is needed to confirm and to investigate the role of these genes in the development of Chinese ICP.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Xiao-Tian Sun
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Xiao-Ling Weng
- Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Dai-Zhan Zhou
- Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Tian Xia
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Liang-Hao Hu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Xiao-Wei Lai
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Bo Ye
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Mu-Yun Liu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Fei Jiang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Jun Gao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Lu-Min Bo
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Yun Liu
- Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medicine University, Shanghai, China
| |
Collapse
|
32
|
Masson E, Chen JM, Audrézet MP, Cooper DN, Férec C. A conservative assessment of the major genetic causes of idiopathic chronic pancreatitis: data from a comprehensive analysis of PRSS1, SPINK1, CTRC and CFTR genes in 253 young French patients. PLoS One 2013; 8:e73522. [PMID: 23951356 PMCID: PMC3738529 DOI: 10.1371/journal.pone.0073522] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/30/2013] [Indexed: 12/21/2022] Open
Abstract
Idiopathic chronic pancreatitis (ICP) has traditionally been defined as chronic pancreatitis in the absence of any obvious precipitating factors (e.g. alcohol abuse) and family history of the disease. Studies over the past 15 years have revealed that ICP has a highly complex genetic architecture involving multiple gene loci. Here, we have attempted to provide a conservative assessment of the major genetic causes of ICP in a sample of 253 young French ICP patients. For the first time, conventional types of mutation (comprising coding sequence variants and variants at intron/exon boundaries) and gross genomic rearrangements were screened for in all four major pancreatitis genes, PRSS1, SPINK1, CTRC and CFTR. For the purposes of the study, synonymous, intronic and 5'- or 3'-untranslated region variants were excluded from the analysis except where there was persuasive evidence of functional consequences. The remaining sequence variants/genotypes were classified into causative, contributory or neutral categories by consideration of (i) their allele frequencies in patient and normal control populations, (ii) their presumed or experimentally confirmed functional effects, (iii) the relative importance of their associated genes in the pathogenesis of chronic pancreatitis and (iv) gene-gene interactions wherever applicable. Adoption of this strategy allowed us to assess the pathogenic relevance of specific variants/genotypes to their respective carriers to an unprecedented degree. The genetic cause of ICP could be assigned in 23.7% of individuals in the study group. A strong genetic susceptibility factor was also present in an additional 24.5% of cases. Taken together, up to 48.2% of the studied ICP patients were found to display evidence of a genetic basis for their pancreatitis. Whereas these particular proportions may not be extrapolable to all ICP patients, the approach employed should serve as a useful framework for acquiring a better understanding of the role of genetic factors in causing this oligogenic disease.
Collapse
Affiliation(s)
- Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale, U1078, Brest, France
- Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire Brest, Hôpital Morvan, Brest, France
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale, U1078, Brest, France
- Etablissement Français du sang – Bretagne, Brest, France
- * E-mail: (J-MC); (CF)
| | - Marie-Pierre Audrézet
- Institut National de la Santé et de la Recherche Médicale, U1078, Brest, France
- Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire Brest, Hôpital Morvan, Brest, France
| | - David N. Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale, U1078, Brest, France
- Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire Brest, Hôpital Morvan, Brest, France
- Etablissement Français du sang – Bretagne, Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France
- * E-mail: (J-MC); (CF)
| |
Collapse
|
33
|
Geisz A, Hegyi P, Sahin-Tóth M. Robust autoactivation, chymotrypsin C independence and diminished secretion define a subset of hereditary pancreatitis-associated cationic trypsinogen mutants. FEBS J 2013; 280:2888-2899. [PMID: 23601753 PMCID: PMC3676443 DOI: 10.1111/febs.12292] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/30/2013] [Accepted: 04/14/2013] [Indexed: 12/14/2022]
Abstract
Mutations in human cationic trypsinogen cause hereditary pancreatitis by altering its proteolytic regulation of activation and degradation by chymotrypsin C (CTRC). CTRC stimulates trypsinogen autoactivation by processing the activation peptide to a shorter form, but also promotes degradation by cleaving the calcium-binding loop in trypsinogen. Mutations render trypsinogen resistant to CTRC-mediated degradation and/or increase processing of the activation peptide by CTRC. Here we demonstrate that the activation peptide mutations D19A, D22G, K23R and K23_I24insIDK robustly increased the rate of trypsinogen autoactivation, both in the presence and absence of CTRC. Degradation of the mutants by CTRC was unchanged, and processing of the activation peptide was increased fourfold in the D19A mutant only. Surprisingly, however, this increased processing had only a minimal effect on autoactivation. The tetra-aspartate motif in the trypsinogen activation peptide binds calcium (KD of ~ 1.6 mM), which stimulates autoactivation. Unexpectedly, calcium binding was not compromised by any of the activation peptide mutations. Despite normal binding, autoactivation of mutants D22G and K23_I24insIDK was not stimulated by calcium. Finally, the activation peptide mutants exhibited reduced secretion from transfected cells, and secreted trypsinogen levels were inversely proportional with autoactivation rates. We conclude that D19A, D22G, K23R and K23_I24insIDK form a mechanistically distinct subset of hereditary pancreatitis-associated mutations that exert their effect primarily through direct stimulation of autoactivation, independently of CTRC. The potentially severe clinical impact of the markedly increased autoactivation is offset by diminished secretion, resulting in a clinical phenotype that is indistinguishable from typical hereditary pancreatitis.
Collapse
Affiliation(s)
- Andrea Geisz
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118
| |
Collapse
|
34
|
Batra J, Szabó A, Caulfield TR, Soares AS, Sahin-Tóth M, Radisky ES. Long-range electrostatic complementarity governs substrate recognition by human chymotrypsin C, a key regulator of digestive enzyme activation. J Biol Chem 2013; 288:9848-9859. [PMID: 23430245 DOI: 10.1074/jbc.m113.457382] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Human chymotrypsin C (CTRC) is a pancreatic serine protease that regulates activation and degradation of trypsinogens and procarboxypeptidases by targeting specific cleavage sites within their zymogen precursors. In cleaving these regulatory sites, which are characterized by multiple flanking acidic residues, CTRC shows substrate specificity that is distinct from that of other isoforms of chymotrypsin and elastase. Here, we report the first crystal structure of active CTRC, determined at 1.9-Å resolution, revealing the structural basis for binding specificity. The structure shows human CTRC bound to the small protein protease inhibitor eglin c, which binds in a substrate-like manner filling the S6-S5' subsites of the substrate binding cleft. Significant binding affinity derives from burial of preferred hydrophobic residues at the P1, P4, and P2' positions of CTRC, although acidic P2' residues can also be accommodated by formation of an interfacial salt bridge. Acidic residues may also be specifically accommodated in the P6 position. The most unique structural feature of CTRC is a ring of intense positive electrostatic surface potential surrounding the primarily hydrophobic substrate binding site. Our results indicate that long-range electrostatic attraction toward substrates of concentrated negative charge governs substrate discrimination, which explains CTRC selectivity in regulating active digestive enzyme levels.
Collapse
Affiliation(s)
- Jyotica Batra
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, Florida 32224
| | - András Szabó
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts 02118
| | - Thomas R Caulfield
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, Florida 32224; Department of Neuroscience, Mayo Clinic Cancer Center, Jacksonville, Florida 32224
| | - Alexei S Soares
- Biology Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts 02118.
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, Florida 32224.
| |
Collapse
|
35
|
Graziani R, Frulloni L, Cicero C, Manfredi R, Ambrosetti MC, Mautone S, Pozzi Mucelli R. Bull's-eye pattern of pancreatic-duct stones on multidetector computed tomography and gene-mutation-associated pancreatitis (GMAP). Radiol Med 2012; 117:1275-86. [PMID: 23090249 DOI: 10.1007/s11547-012-0888-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/13/2012] [Indexed: 01/28/2023]
Abstract
PURPOSE This study prospectively assessed whether the presence of a bull's-eye pattern of pancreatic-duct stones on multidetector computed tomography (MDCT) correlated with gene-mutation-associated pancreatitis (GMAP) and whether other signs suggestive of GMAP can be detected with MDCT. MATERIALS AND METHODS Forty-seven patients with chronic calcific pancreatitis underwent genetic testing for CFTR, SPINK1 and PRSS1 mutations and an MDCT scan of the abdomen. Qualitative analysis assessed the presence or absence of pancreatic-duct stones with bull's-eye appearance. Quantitative analysis included the number and maximum diameter of stones and the diameter of the main pancreatic duct. RESULTS Fifteen of 47 patients (32%) were positive for gene mutations (GMAP patients). The bull's-eye pattern was found in 10/15 patients (67%) with GMAP and in 4/32 (12%) patients with chronic pancreatitis not associated with GMAP (NGMAP; p<0.0001). The mean diameter of duct stones was 15 mm in patients with GMAP and 10 mm in patients with NGMAP (p<0.04). CONCLUSIONS The presence of duct stones with a bull's-eye pattern correlates with GMAP. Duct stones with diameter ≥15 mm are another sign suggestive of GMAP.
Collapse
Affiliation(s)
- R Graziani
- Istituto di Radiologia, Università di Verona, P.le L.A. Scuro 10, 37134, Verona, Italy.
| | | | | | | | | | | | | |
Collapse
|
36
|
Genetics and pathogenesis of chronic pancreatitis: the 2012 update. Clin Res Hepatol Gastroenterol 2012; 36:334-40. [PMID: 22749696 DOI: 10.1016/j.clinre.2012.05.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/15/2012] [Indexed: 02/04/2023]
Abstract
Chronic pancreatitis is a persistent inflammation of the pancreas that results in irreversible morphological changes and impairment of both exocrine and endocrine functions. Genetic studies of the disease over the past 15 years have led to the identification of four firmly established susceptibility genes namely PRSS1, SPNIK1, CTRC and CFTR. Our previously published review (Chen and Férec. Annu Rev Genomics Hum Genet 2009) has comprehensively summarized the advances made in terms of genetics and pathogenesis of chronic pancreatitis, covering the data available up to early 2009. This review summarizes the important and representative findings published thereafter, focusing on (i) newly found disease-causing mutations, (ii) functional characterization of known variations and (iii) genotype and phenotype relationship.
Collapse
|
37
|
Strong purifying selection against gene conversions in the trypsin genes of primates. Hum Genet 2012; 131:1739-49. [PMID: 22752798 DOI: 10.1007/s00439-012-1196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/20/2012] [Indexed: 01/27/2023]
Abstract
The trypsin gene families of primate species are composed of members who share a remarkable level of sequence similarity. Here, we investigated the gene conversions occurring within the trypsin gene family in five primate species. A total of 36 conversion events, with an average length (±standard deviation) of 1,526 ± 1,124 nucleotides, were detected using two methods. Such extensive gene conversions are likely both the cause and the consequence of the high sequence similarity between primate trypsin genes. In the trypsins encoded by these genes, both the overall amino acid sequences and critical amino acid residues are conserved. Therefore, the numerous long gene conversions we detected between trypsin genes did not alter any of their functionally important amino acid sites. This suggest that, in the trypsin genes of the five primate species studied here, strong purifying selection against gene conversions is occurring in regions containing functionally important residues.
Collapse
|
38
|
Szabó A, Sahin-Tóth M. Increased activation of hereditary pancreatitis-associated human cationic trypsinogen mutants in presence of chymotrypsin C. J Biol Chem 2012; 287:20701-10. [PMID: 22539344 DOI: 10.1074/jbc.m112.360065] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Mutations in human cationic trypsinogen (PRSS1) cause autosomal dominant hereditary pancreatitis. Increased intrapancreatic autoactivation of trypsinogen mutants has been hypothesized to initiate the disease. Autoactivation of cationic trypsinogen is proteolytically regulated by chymotrypsin C (CTRC), which mitigates the development of trypsin activity by promoting degradation of both trypsinogen and trypsin. Paradoxically, CTRC also increases the rate of autoactivation by processing the trypsinogen activation peptide to a shorter form. The aim of this study was to investigate the effect of CTRC on the autoactivation of clinically relevant trypsinogen mutants. We found that in the presence of CTRC, trypsinogen mutants associated with classic hereditary pancreatitis (N29I, N29T, V39A, R122C, and R122H) autoactivated at increased rates and reached markedly higher active trypsin levels compared with wild-type cationic trypsinogen. The A16V mutant, known for its variable disease penetrance, exhibited a smaller increase in autoactivation. The mechanistic basis of increased activation was mutation-specific and involved resistance to degradation (N29I, N29T, V39A, R122C, and R122H) and/or increased N-terminal processing by CTRC (A16V and N29I). These observations indicate that hereditary pancreatitis is caused by CTRC-dependent dysregulation of cationic trypsinogen autoactivation, which results in elevated trypsin levels in the pancreas.
Collapse
Affiliation(s)
- András Szabó
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118, USA
| | | |
Collapse
|
39
|
Rebours V, Lévy P, Ruszniewski P. An overview of hereditary pancreatitis. Dig Liver Dis 2012; 44:8-15. [PMID: 21907651 DOI: 10.1016/j.dld.2011.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/05/2011] [Indexed: 12/11/2022]
Abstract
Hereditary pancreatitis is a rare cause of chronic pancreatitis. The prevalence was evaluated to 0.3/100000 in Western Countries. Genetic disorders are due to mutations of the PRSS1 gene on the long arm of the chromosome 7, encoding for the cationic trypsinogen. The inheritance pattern is autosomal dominant with an incomplete penetrance (80%). Since 1996, more than 30 mutations were found. The three more common mutations are R122H, N29I and A16V. First symptoms begin since childhood, mainly before 10 years old. Main symptoms are pancreatic pain and acute pancreatitis (>70%). CP morphological changes as pancreatic calcifications are diagnosed at a median age of 22-25 years. Exocrine and endocrine pancreatic insufficiency occurred in 34% and 26% at a median age of 29 and 38 years. No clinical differences exist according to the mutation type. No excess of mortality in hereditary pancreatitis population compared to general population was found, despite a real risk of cancer. The cumulative risks of pancreatic cancer at 50, 60 and, 75 years are 10%, 18.7% and, 53.5%, respectively. The relative risk of cancer increases in smokers and is evaluated to 8.55. Hereditary pancreatitis diagnosis permits to propose an adapted management in expert centres.
Collapse
Affiliation(s)
- Vinciane Rebours
- Pôle des Maladies de l'Appareil Digestif, Service de Gastroentérologie - Pancréatologie, Hôpital Beaujon, AP-HP, Université Denis Diderot-Paris VII, Clichy, France.
| | | | | |
Collapse
|
40
|
Abstract
Chronic pancreatitis is a persistent inflammatory disorder characterized by destruction of the pancreatic parenchyma, maldigestion, and chronic pain. Mutations in the chymotrypsin C (CTRC) gene encoding the digestive enzyme CTRC have been shown to increase the risk of chronic pancreatitis in European and Asian populations. Here, we review the biochemical properties and physiological functions of human CTRC, summarize the functional defects associated with CTRC mutations, and discuss mechanistic models that might explain the increased disease risk in carriers.
Collapse
Affiliation(s)
- Jiayi Zhou
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA 02118, USA
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA 02118, USA
| |
Collapse
|
41
|
da Costa MZG, Guarita DR, Ono-Nita SK, Paranaguá-Vezozzo DC, Felga GEG, Pedroso MRA, de Souza MMT, Nasser PD, Ferreira CDS, Carrilho FJ. Genetic risk for alcoholic chronic pancreatitis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2011; 8:2747-57. [PMID: 21845156 PMCID: PMC3155327 DOI: 10.3390/ijerph8072747] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/20/2011] [Indexed: 12/14/2022]
Abstract
In recent years many studies have examined the genetic predisposition to pancreatic diseases. Pancreatic disease of an alcoholic etiology was determined to be a multi-factorial disease, where environmental factors interact with the genetic profile of the individual. In this review we discuss the main results from studies examining the frequency of genetic mutations in alcoholic chronic pancreatitis.
Collapse
|
42
|
Sandhu B, Vitazka P, Ferreira-Gonzalez A, Pandya A, Vachhani R, Bouhaidar D, Zfass A, Sanyal A. Presence of SPINK-1 variant alters the course of chronic pancreatitis. J Gastroenterol Hepatol 2011; 26:965-9. [PMID: 21375584 DOI: 10.1111/j.1440-1746.2011.06713.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS There is growing evidence that genetic mutations/variants increase susceptibility to the development and progression of chronic pancreatitis (CP). Several mutations have been identified that have a direct and indirect role in events leading to CP. Mutations in the serine protease inhibitor, Kazal type-1 (SPINK-1) gene have been reported to lower the threshold for pancreatitis in the presence of other genetic or environmental factors. The prevalence and impact of SPINK-1 mutations on the clinical course and outcomes of CP remains unclear. This study was conducted to assess the prevalence of the SPINK-1/N34S variant in patients with CP, and to understand the impact of the SPINK-1 mutation on the natural history of CP. METHODS A retrospective-prospective analysis of 239 patients with CP was performed. A detailed history, including duration of symptoms, type of pain (intermittent flares or chronic continuous pain), number of flares requiring hospital admission, alcohol and smoking history, and family history was obtained. The baseline morphological stage of CP was categorized by Cambridge classification. Clinical outcome variables included frequency and severity of pain episodes, presence of exocrine failure (defined by presence of steatorrhea and/or fecal elastase < 200 ug/g), and diabetes. The genetic tests included the cationic trypsinogen gene-1 mutation, cystic fibrosis gene mutations (Genzyme assay), and the SPINK-1/N34S mutation. RESULTS Of the 239 patients with CP, 13 (5.4%) were positive for the SPINK-1/N34S mutation. There were 35 (14.6%) patients with idiopathic pancreatitis (IP) in this cohort. Most of the patients who were positive for the SPINK-1/N34S mutation had IP and were Caucasian (69.2%). The patients with the SPINK-1/N34S mutation had a younger age of onset (32.9 ± 10.2 vs 40.1 ± 13.6 years; P = 0.108) than those with IP and no mutation. Over a median follow up of 9.6 years, the patients with the SPINK-1/N34S mutation had a significantly greater number of acute flares each year, as compared to those without the mutation (11.8 ± 1.5 vs 4 ± 0.98; P = 0.0001). CONCLUSIONS The prevalence of the SPINK-1/N34S mutation in patients with CP is 5.4%, and is approximately 37.1% in patients with IP. These mutations are more prevalent in Caucasian patients with CP. The SPINK-1/N34S mutation predisposes to early onset IP and more frequent acute flares of pancreatitis that might ultimately lead to pancreatic insufficiency. The patients with IP and borderline alcohol history should be considered for testing for genetic analysis, including SPINK-1 mutations, initially restricted to clinical trials.
Collapse
Affiliation(s)
- Bimaljit Sandhu
- Department of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23233, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
It is now generally believed that pancreatitis results from pancreatic autodigestion. An inappropriate conversion of pancreatic zymogens to active enzymes within the pancreatic parenchyma is thought to initiate the inflammatory process. A key role has been attributed to the activation of trypsinogen to trypsin, converting all proteolytic proenzymes to their active form. Several gain-of-function mutations in the cationic trypsinogen gene (PRSS1) have been identified in patients with chronic pancreatitis (CP). These mutations lead to enhanced intrapancreatic trypsinogen activation. In contrast, a variant in the anionic trypsinogen (PRSS2) gene, p.G191R, has been described that mitigates intrapancreatic trypsin activity and thereby plays a protective role. Beside trypsinogen mutations, loss-of-function variants in SPINK1, encoding a pancreatic trypsin inhibitor, are strongly associated with idiopathic CP. Approximately 15-40% of patients with so-called idiopathic CP carry p.N34S on one allele or on both alleles. Chymotrypsin C (CTRC) degrades all human trypsin isoforms with high specificity. Two CTRC alterations, p.R254W and p.K247_R254del, are significantly associated with idiopathic as well as alcohol-related CP. Functional analysis of the variants revealed impaired activity and/or reduced secretion. Thus, loss-of-function mutations in CTRC predispose to pancreatitis by diminishing its protective trypsin-degrading activity. Albeit the association between CFTR, the gene mutated in cystic fibrosis, and idiopathic CP is now well established, the pathogenic mechanisms are poorly understood. Nearly 25-30% of patients carry at least one CFTR mutation, but few patients only were compound-heterozygous. Several patients, however, are trans-heterozygous for a CFTR alteration and a PRSS1, SPINK1, or CTRC variant, respectively.
Collapse
Affiliation(s)
- Heiko Witt
- Department of Pediatrics, Technische Universität München (TUM), Munich, Germany.
| |
Collapse
|
44
|
Szabó A, Héja D, Szakács D, Zboray K, Kékesi KA, Radisky ES, Sahin-Tóth M, Pál G. High affinity small protein inhibitors of human chymotrypsin C (CTRC) selected by phage display reveal unusual preference for P4' acidic residues. J Biol Chem 2011; 286:22535-45. [PMID: 21515688 DOI: 10.1074/jbc.m111.235754] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human chymotrypsin C (CTRC) is a pancreatic protease that participates in the regulation of intestinal digestive enzyme activity. Other chymotrypsins and elastases are inactive on the regulatory sites cleaved by CTRC, suggesting that CTRC recognizes unique sequence patterns. To characterize the molecular determinants underlying CTRC specificity, we selected high affinity substrate-like small protein inhibitors against CTRC from a phage library displaying variants of SGPI-2, a natural chymotrypsin inhibitor from Schistocerca gregaria. On the basis of the sequence pattern selected, we designed eight inhibitor variants in which amino acid residues in the reactive loop at P1 (Met or Leu), P2' (Leu or Asp), and P4' (Glu, Asp, or Ala) were varied. Binding experiments with CTRC revealed that (i) inhibitors with Leu at P1 bind 10-fold stronger than those with P1 Met; (ii) Asp at P2' (versus Leu) decreases affinity but increases selectivity, and (iii) Glu or Asp at P4' (versus Ala) increase affinity 10-fold. The highest affinity SGPI-2 variant (K(D) 20 pm) bound to CTRC 575-fold tighter than the parent molecule. The most selective inhibitor variant exhibited a K(D) of 110 pm and a selectivity ranging from 225- to 112,664-fold against other human chymotrypsins and elastases. Homology modeling and mutagenesis identified a cluster of basic amino acid residues (Lys(51), Arg(56), and Arg(80)) on the surface of human CTRC that interact with the P4' acidic residue of the inhibitor. The acidic preference of CTRC at P4' is unique among pancreatic proteases and might contribute to the high specificity of CTRC-mediated digestive enzyme regulation.
Collapse
Affiliation(s)
- András Szabó
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Heterologously expressed proteins in Escherichia coli may undergo unwanted N-terminal processing by methionine and proline aminopeptidases. To overcome this problem, we present a system where the gene of interest is cloned as a fusion to a self-splicing mini-intein. This fusion construct is expressed in an engineered E. coli strain from which the pepP gene coding for aminopeptidase P has been deleted. We describe a protocol using human cationic trypsinogen as an example to demonstrate that recombinant proteins produced in this expression system contain homogeneous, unprocessed N-termini.
Collapse
Affiliation(s)
- Orsolya Király
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA.
| | | | | |
Collapse
|
46
|
OHMURAYA M, YAMAMURA KI. The Roles of Serine Protease Inhibitor Kazal Type 1 (SPINK1) in Pancreatic Diseases. Exp Anim 2011; 60:433-44. [DOI: 10.1538/expanim.60.433] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Masaki OHMURAYA
- Priority Organization for Innovation and Excellence
- Institute of Resource Development and Analysis, Kumamoto University
| | | |
Collapse
|
47
|
Gasiorowska A, Talar-Wojnarowska R, Czupryniak L, Smolarz B, Romanowicz-Makowska H, Kulig A, Malecka-Panas E. The prevalence of cationic trypsinogen (PRSS1) and serine protease inhibitor, Kazal type 1 (SPINK1) gene mutations in Polish patients with alcoholic and idiopathic chronic pancreatitis. Dig Dis Sci 2011; 56:894-901. [PMID: 20676769 PMCID: PMC3041903 DOI: 10.1007/s10620-010-1349-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 07/12/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND The main cause of chronic pancreatitis (CP) is excessive alcohol consumption. On the other hand, only 5-10% of heavy drinkers develop chronic pancreatitis. We have only limited information regarding the pathogenic mechanism by which alcohol leads to the disease. Mutations of the PRSS1 and SPINK 1 have been mostly implicated in hereditary and idiopathic CP, but their presence in other types of this disease have also been reported. AIMS The aim of the study was to determine the frequency of PRSS1 and SPINK1 mutations in patients with chronic alcoholic (ACP) and idiopathic pancreatitis (ICP) as well as to investigate their relation to the clinical course of the disease. METHODS The study included 33 ACP and 14 ICP patients as well 46 healthy subjects. The diagnosis of CP was based on clinical data, ultrasound, and computed tomography. After isolation of DNA from peripheral blood two trypsinogen mutations were detected N29I and R122H by allelo-specific amplification polymerase chain reaction (ASA-PCR) and by the PCR-restriction fragment length polymorphism (RFLP). Beside this N34S mutation of SPINK1 was analyzed by PCR restriction fragment length polymorphism (PCR-RFLP). RESULTS PRSS1 mutations have been detected in 11 (33%) patients with ACP. The frequency of the PRSS1 mutations was higher in patients with ACP than in controls (4.3%) (p < 0.001). The frequency of PRSS1 mutation was present in 21.4% of ICP patients, which was significantly higher (p < 0.05) than in controls. Overall, six (18%) SPINK1 mutations in ACP group have been detected. Among 14 patients with ICP, in four (28.6%) of them SPINK1 has been detected. The same mutations have also been found in three (6.5%) control subjects. The frequency of the N34S mutation was higher in patients with ICP than in the controls (p < 0.05), but the frequency of N34S mutation did not differ between ACP and the control group. No relations have been detected between PRSS1 and SPINK1 mutations presence and clinical course and complications of CP. CONCLUSIONS Those preliminary data suggest the high prevalence of SPINK1 and PRSS1 mutations in the Polish population, generally, as well as in CP patients. It may be speculated that those mutations contribute to the development of chronic pancreatitis, especially in patients with alcohol overindulgence.
Collapse
Affiliation(s)
- Anita Gasiorowska
- Department of Digestive Tract Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| | - Renata Talar-Wojnarowska
- Department of Digestive Tract Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| | - Leszek Czupryniak
- Diabetology and Metabolic Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| | - Beata Smolarz
- Laboratory of Molecular Genetics, Institute of Polish Mother’s Memorial Hospital, Rzgowska 281/289, 93-338 Lodz, Poland
| | - Hanna Romanowicz-Makowska
- Laboratory of Molecular Genetics, Institute of Polish Mother’s Memorial Hospital, Rzgowska 281/289, 93-338 Lodz, Poland
| | - Andrzej Kulig
- Laboratory of Molecular Genetics, Institute of Polish Mother’s Memorial Hospital, Rzgowska 281/289, 93-338 Lodz, Poland
| | - Ewa Malecka-Panas
- Department of Digestive Tract Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| |
Collapse
|
48
|
Abstract
Hereditary chronic pancreatitis (HCP) is a rare disease in which chronic pancreatitis develops in childhood. HCP has autosomal dominant inheritance with approximately 80% penetrance. Diagnostic criteria are not universally agreed upon but the EUROPAC trial defined it as two first-degree relatives or at least 3 second-degree relatives in two or more generations, with chronic pancreatitis for which there is no other etiology. The gene for HCP was originally identified on chromosome 7 and subsequently many other genes have been reported to be associated with HCP. To date, no single genetic alteration has been found that is necessary for the development of HCP. In a recent study, 81% of patients with HCP were found to have a mutation of the PRSS1 gene. Patients with HCP are at risk for developing exocrine and endocrine insufficiency and there is a 50-fold increased risk of pancreatic cancer in HCP patients as compared with the general population.
Collapse
|
49
|
Genetic, epidemiological, and clinical aspects of hereditary pancreatitis: a population-based cohort study in Denmark. Am J Gastroenterol 2010; 105:1876-83. [PMID: 20502448 DOI: 10.1038/ajg.2010.193] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES In a population-based, well-defined group of patients first regarded as having pancreatitis of unknown origin (PUO), we identified, described, and compared the clinical and genetic aspects of patients with hereditary pancreatitis (HP) and with cystic fibrosis transmembrane conductance regulator gene (CFTR) and serine protease inhibitor Kazal type 1 gene (SPINK1) mutations with patients who retained the diagnosis of true idiopathic pancreatitis (tIP) after genetic testing for HP, SPINK1, and CFTR mutations. METHODS Patients with PUO were identified in the Danish National Registry of Patients or were referred by clinicians. DNA from blood was analyzed for cationic trypsinogen (PRSS1), SPINK1, and CFTR mutations. Considering the diagnosis of HP, a pedigree was drawn for each patient. RESULTS A genetic mutation was found in 40% of 122 patients with PUO. After testing first-degree relatives of the 18 initially identified HP patients, 38 HP patients in total were identified, and 28 patients had SPINK1-CFTR mutations. Among HP patients, no p.N29I mutations were found and the p.A16V mutation was more frequent than previously reported, 45 and 32% had exocrine and endocrine insufficiency, respectively, and among tIP patients 9 and 12%, respectively. Pancreatic cancer was diagnosed in 5% of the HP families. CONCLUSIONS The genotype of the Danish population with HP differs from that of previously described cohorts. The occurrence of exocrine and endocrine insufficiency is higher among patients with HP than in patients with SPINK1-CFTR mutations and tIP, and more HP families develop pancreatic cancer. Genetic testing thus helps to predict the prognosis of the pancreatitis.
Collapse
|
50
|
Rosendahl J, Teich N, Kovacs P, Szmola R, Blüher M, Gress TM, Hoffmeister A, Keim V, Löhr M, Mössner J, Nickel R, Ockenga J, Pfützer R, Schulz HU, Stumvoll M, Wittenburg H, Sahin-Tóth M, Witt H. Complete analysis of the human mesotrypsinogen gene (PRSS3) in patients with chronic pancreatitis. Pancreatology 2010; 10:243-9. [PMID: 20484962 PMCID: PMC2899151 DOI: 10.1159/000243769] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 09/21/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS A sustained imbalance of pancreatic proteases and their inhibitors seems to be important for the development of chronic pancreatitis (CP). Mesotrypsin (PRSS3) can degrade intrapancreatic trypsin inhibitors that protect against CP. Genetic variants that cause higher mesotrypsin activity might increase the risk for CP. METHODS We analyzed all 5 exons and the adjacent non-coding sequences of PRSS3 by direct sequencing of 313 CP patients and 327 controls. Additionally, exon 4 was investigated in 855 patients and 1,294 controls and a c.454+191G>A variant in 855 patients and 1,467 controls. The c.499A>G (p.T167A) variant was analyzed functionally using transiently transfected HEK 293T cells. RESULTS In the exonic regions, the previously described common c.94_96delGAG (p.E32del) variant and a novel p.T167A non-synonymous alteration were identified. Extended analysis of the p.T167A variant revealed no association to CP and in functional assays p.T167A showed normal secretion and activity. Variants of the intronic regions, including the extensively analyzed c.454+191G>A alteration, were not associated with the disease. Haplotype reconstruction using variants with a minor allele frequency of >1% revealed no CP-associated haplotype. CONCLUSIONS Although the trypsin inhibitor-degrading activity qualified PRSS3 as a candidate for a novel CP susceptibility gene, we found no association between a specific variant or haplotype and CP in our cohort with a high suspicion of genetically determined disease.
Collapse
Affiliation(s)
- Jonas Rosendahl
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany,*Jonas Rosendahl, MD, Medizinische Klinik und Poliklinik II, Universitätsklinikum Leipzig, Liebigstrasse 20, DE–04103 Leipzig (Germany), Tel. +49 341 971 3223, Fax +49 341 971 2209, E-Mail
| | - Niels Teich
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany,Internistische Gemeinschaftspraxis für Verdauungs- und Stoffwechselerkrankungen, Leipzig, Germany
| | - Peter Kovacs
- Interdisciplinary Center for Clinical Research Leipzig, University of Leipzig, Leipzig, Germany
| | - Richard Szmola
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, Mass., USA
| | - Matthias Blüher
- Department of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Thomas M. Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Albrecht Hoffmeister
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany
| | - Volker Keim
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Joachim Mössner
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany
| | | | - Johann Ockenga
- Department of Medicine II (Gastroenterology/Hepatology), Klinikum Bremen-Mitte, Bremen, Germany
| | - Roland Pfützer
- Department of Medicine II (Gastroenterology/Hepatology/Infectious Diseases), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Michael Stumvoll
- Department of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Henning Wittenburg
- Department of Gastroenterology and Hepatology, University of Leipzig, Leipzig, Germany
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, Mass., USA
| | - Heiko Witt
- Department of Hepatology and Gastroenterology, Charité, Berlin, Germany
| |
Collapse
|