1
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
2
|
Singh VP, Gorelick FS. Has a Hundred Years of Pursuing Proteases Helped to Palliate Pain in Chronic Pancreatitis More Than Placebo? Gastroenterology 2024; 166:559-561. [PMID: 38311123 DOI: 10.1053/j.gastro.2024.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Affiliation(s)
- Vijay P Singh
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona.
| | - Fred S Gorelick
- Department of Medicine, Section of Digestive Diseases, and Cell Biology, Yale University School of Medicine, New Haven, Connecticut; VA Connecticut, West Haven, Connecticut
| |
Collapse
|
3
|
Girodon E, Rebours V, Chen JM, Pagin A, Levy P, Ferec C, Bienvenu T. WITHDRAWN: Clinical interpretation of PRSS1 gene variants in patients with pancreatitis. Clin Res Hepatol Gastroenterol 2022; 46:101531. [PMID: 36057185 DOI: 10.1016/j.clinre.2020.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/27/2020] [Indexed: 02/04/2023]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published in Clinics and Research in Hepatology and Gastroenterology, Volume 45, Issue 1, 2021, 101497. https://doi.org/10.1016/j.clinre.2020.07.004. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal
Collapse
Affiliation(s)
- Emmanuelle Girodon
- Laboratoire de G..n..tique et Biologie Mol..culaires, H..pital Cochin, APHP.Centre-Universit.. de Paris, France
| | - Vinciane Rebours
- Service de Pancr..atologie-Gastroent..rologie, P..le des Maladies de l'Appareil Digestif, Universit.. Denis Diderot, H..pital Beaujon, APHP, DHU UNITY, Clichy, and Centre de R..f..rence des Maladies Rares du Pancr..as...PAncreaticRaresDISeases (PaRaDis), France
| | - Jian Min Chen
- INSERM UMR1078 "G..n..tique, G..nomique Fonctionnelle et Biotechnologies", EFS - Bretagne, Universit.. de Brest, CHRU Brest, Brest, France
| | - Adrien Pagin
- CHU Lille, Service de Toxicologie et G..nopathies, Lille, France
| | - Philippe Levy
- Service de Pancr..atologie-Gastroent..rologie, P..le des Maladies de l'Appareil Digestif, Universit.. Denis Diderot, H..pital Beaujon, APHP, DHU UNITY, Clichy, and Centre de R..f..rence des Maladies Rares du Pancr..as...PAncreaticRaresDISeases (PaRaDis), France
| | - Claude Ferec
- INSERM UMR1078 "G..n..tique, G..nomique Fonctionnelle et Biotechnologies", EFS - Bretagne, Universit.. de Brest, CHRU Brest, Brest, France
| | - Thierry Bienvenu
- Laboratoire de G..n..tique et Biologie Mol..culaires, H..pital Cochin, APHP.Centre-Universit.. de Paris, France.
| |
Collapse
|
4
|
Bennett C, Suguitan M, Abad J, Chawla A. Identification of high-risk germline variants for the development of pancreatic cancer: Common characteristics and potential guidance to screening guidelines. Pancreatology 2022; 22:719-729. [PMID: 35798629 DOI: 10.1016/j.pan.2022.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer (PC) is a product of a variety of environmental and genetic factors. Recent work has highlighted the influence of hereditary syndromes on pancreatic cancer incidence. The purpose of this review is to identify the high-risk syndromes, common variants, and risks associated with PC. The study also elucidates common characteristics of patients with these mutations, which is used to recommend potential changes to current screening protocols for greater screening efficacy. We analyzed 8 syndromes and their respective variants: Hereditary Breast and Ovarian Cancer (BRCA1/2), Familial Atypical Multiple Mole Melanoma Syndrome (CDKN2A), Peutz-Jeghers Syndrome (STK11), Lynch Syndrome (PMS2, MLH1, MSH2, MSH6, EPCAM), Ataxia Telangiectasia (ATM), Li-Fraumeni Syndrome (TP53), Fanconi Anemia (PALB2), and Hereditary Pancreatitis (PRSS1, SPINK1, CFTR). Of 587 studies evaluated, 79 studies fit into our inclusion criteria. Information from each study was analyzed to draw conclusions on these variants as well as their association with pancreatic cancer. Information from this review is intended to improve precision medicine and improve criteria for screening.
Collapse
Affiliation(s)
- Cade Bennett
- Division of Surgical Oncology, Department of Surgery, Northwestern Medicine Regional Medical Group, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mike Suguitan
- Division of Surgical Oncology, Department of Surgery, Northwestern Medicine Regional Medical Group, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John Abad
- Division of Surgical Oncology, Department of Surgery, Northwestern Medicine Regional Medical Group, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern Medicine Regional Medical Group, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
5
|
Wood LD, Canto MI, Jaffee EM, Simeone DM. Pancreatic Cancer: Pathogenesis, Screening, Diagnosis, and Treatment. Gastroenterology 2022; 163:386-402.e1. [PMID: 35398344 PMCID: PMC9516440 DOI: 10.1053/j.gastro.2022.03.056] [Citation(s) in RCA: 296] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/13/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging cancer, due to both its late stage at diagnosis and its resistance to chemotherapy. However, recent advances in our understanding of the biology of PDAC have revealed new opportunities for early detection and targeted therapy of PDAC. In this review, we discuss the pathogenesis of PDAC, including molecular alterations in tumor cells, cellular alterations in the tumor microenvironment, and population-level risk factors. We review the current status of surveillance and early detection of PDAC, including populations at high risk and screening approaches. We outline the diagnostic approach to PDAC and highlight key treatment considerations, including how therapeutic approaches change with disease stage and targetable subtypes of PDAC. Recent years have seen significant improvements in our approaches to detect and treat PDAC, but large-scale, coordinated efforts will be needed to maximize the clinical impact for patients and improve overall survival.
Collapse
Affiliation(s)
- Laura D Wood
- Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Sidney Kimmel Cancer Center, Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Diane M Simeone
- Departments of Surgery and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
6
|
Liu K, Liu J, Zou B, Li C, Zeh HJ, Kang R, Kroemer G, Huang J, Tang D. Trypsin-Mediated Sensitization to Ferroptosis Increases the Severity of Pancreatitis in Mice. Cell Mol Gastroenterol Hepatol 2021; 13:483-500. [PMID: 34562639 PMCID: PMC8688567 DOI: 10.1016/j.jcmgh.2021.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Pancreatitis is characterized by acinar cell death and persistent inflammation. Ferroptosis is a type of lipid peroxidation-dependent necrosis, which is negatively regulated by glutathione peroxidase 4. We studied how trypsin, a serine protease secreted by pancreatic acinar cells, affects the contribution of ferroptosis to triggering pancreatitis. METHODS In vitro, the mouse pancreatic acinar cell line 266-6 and mouse primary pancreatic acinar cells were used to investigate the effect of exogenous trypsin on ferroptosis sensitivity. Short hairpin RNAs were designed to silence gene expression, whereas a library of 1080 approved drugs was used to identify new ferroptosis inhibitors in 266-6 cells. In vivo, a Cre/LoxP system was used to generate mice with a pancreas-specific knockout of Gpx4 (Pdx1-Cre;Gpx4flox/flox mice). Acute or chronic pancreatitis was induced in these mice (Gpx4flox/flox mice served as controls) by cerulein injections or a Lieber-DeCarli alcoholic liquid diet. Pancreatic tissues, acinar cells, and serum were collected and analyzed by histology, immunoblot, quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, or immunohistochemical analyses. RESULTS Supraphysiological doses of trypsin (500 or 1000 ng/mL) alone did not trigger significant cell death in 266-6 cells and mouse primary pancreatic acinar cells, but did increase the sensitivity of these cells to ferroptosis upon treatment with cerulein, L-arginine, alcohol, erastin, or RSL3. Proteasome 26S subunit, non-adenosine triphosphatase 4-dependent lipid peroxidation caused ferroptosis in pancreatic acinar cells by promoting the proteasomal degradation of glutathione peroxidase 4. The drug screening campaign identified the antipsychotic drug olanzapine as an antioxidant inhibiting ferroptosis in pancreatic acinar cells. Mice lacking pancreatic Gpx4 developed more severe pancreatitis after cerulein infection or ethanol feeding than control mice. Conversely, olanzapine administration protected against pancreatic ferroptotic damage and experimental pancreatitis in Gpx4-deficient mice. CONCLUSIONS Trypsin-mediated sensitization to ferroptotic damage increases the severity of pancreatitis in mice, and this process can be reversed by olanzapine.
Collapse
Affiliation(s)
- Ke Liu
- Department of Ophthalmology, The 2nd Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Liu
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Borong Zou
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Changfeng Li
- Department of Endoscopy Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Herbert J. Zeh
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Jun Huang
- Department of Orthopaedics, The 2nd Xiangya Hospital, Central South University, Changsha, China,Jun Huang, MD, Department of Orthopaedics, The 2nd Xiangya Hospital, Central South University, Changsha 410011, China. fax: (86) 731-85295999
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas,Correspondence Address correspondence to: Daolin Tang, MD, Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390
| |
Collapse
|
7
|
Tomomura A, Bandow K, Tomomura M. Purification and Biological Function of Caldecrin. MEDICINES (BASEL, SWITZERLAND) 2021; 8:41. [PMID: 34436220 PMCID: PMC8398347 DOI: 10.3390/medicines8080041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Blood calcium homeostasis is critical for biological function. Caldecrin, or chymotrypsin-like elastase, was originally identified in the pancreas as a serum calcium-decreasing factor. The serum calcium-decreasing activity of caldecrin requires the trypsin-mediated activation of the protein. Protease activity-deficient mature caldecrin can also reduce serum calcium concentration, indicating that structural processing is necessary for serum calcium-decreasing activity. Caldecrin suppresses the differentiation of bone-resorbing osteoclasts from bone marrow macrophages (BMMs) by inhibiting receptor activator of NF-κB ligand (RANKL)-induced nuclear factor of activated T-cell cytoplasmic 1 expression via the Syk-PLCγ-Ca2+ oscillation-calcineurin signaling pathway. It also suppresses mature osteoclastic bone resorption by RANKL-stimulated TRAF6-c-Src-Syk-calcium entry and actin ring formation. Caldecrin inhibits lipopolysaccharide (LPS)-induced osteoclast formation in RANKL-primed BMMs by inducing the NF-κB negative regulator A20. In addition, caldecrin suppresses LPS-mediated M1 macrophage polarization through the immunoreceptor triggering receptor expressed on myeloid cells (TREM) 2, suggesting that caldecrin may function as an anti-osteoclastogenic and anti-inflammatory factor via TREM2. The ectopic intramuscular expression of caldecrin cDNA prevents bone resorption in ovariectomized mice, and the administration of caldecrin protein also prevents skeletal muscle destruction in dystrophic mice. In vivo and in vitro studies have indicated that caldecrin is a unique multifunctional protease and a possible therapeutic target for skeletal and inflammatory diseases.
Collapse
Affiliation(s)
- Akito Tomomura
- Division of Biochemistry, Department of Oral Biology & Tissue Engineering, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama 350-0283, Japan;
| | - Kenjiro Bandow
- Division of Biochemistry, Department of Oral Biology & Tissue Engineering, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama 350-0283, Japan;
| | - Mineko Tomomura
- Department of Oral Health Sciences, Meikai University School of Health Sciences, 1-1 Akemi, Urayasu, Chiba 279-8550, Japan;
| |
Collapse
|
8
|
Kulke M, Nagel F, Schulig L, Geist N, Gabor M, Mayerle J, M Lerch M, Link A, Delcea M. A Hypothesized Mechanism for Chronic Pancreatitis Caused by the N34S Mutation of Serine Protease Inhibitor Kazal-Type 1 Based on Conformational Studies. J Inflamm Res 2021; 14:2111-2119. [PMID: 34054303 PMCID: PMC8157096 DOI: 10.2147/jir.s304787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Although strongly related, the pathophysiological effect of the N34S mutation in the serine protease inhibitor Kazal type 1 (SPINK1) in chronic pancreatitis is still unknown. In this study, we investigate the conformational space of the human cationic trypsin-serine protease inhibitor complex. Methods Simulations with molecular dynamics, replica exchange, and transition pathway methods are used. Results Two main binding states of the inhibitor to the complex were found, which explicitly relate the influence of the mutation site to conformational changes in the active site of trypsin. Conclusion Based on our result, a hypothesis is formulated that explains the development of chronic pancreatitis through accelerated digestion of the mutant by trypsin.
Collapse
Affiliation(s)
- Martin Kulke
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Felix Nagel
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Lukas Schulig
- Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Norman Geist
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Marcel Gabor
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine II, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Markus M Lerch
- Department of Medicine a, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Link
- Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Mihaela Delcea
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| |
Collapse
|
9
|
Link JM, Liudahl SM, Betts CB, Sivagnanam S, Leis KR, McDonnell M, Pelz CR, Johnson B, Hamman KJ, Keith D, Sampson JE, Morgan TK, Lopez CD, Coussens LM, Sears RC. Tumor-Infiltrating Leukocyte Phenotypes Distinguish Outcomes in Related Patients With Pancreatic Adenocarcinoma. JCO Precis Oncol 2021; 5:PO.20.00287. [PMID: 34036232 PMCID: PMC8140804 DOI: 10.1200/po.20.00287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/23/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Jason M. Link
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
| | - Shannon M. Liudahl
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| | - Courtney B. Betts
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| | | | - Kenna R. Leis
- Computational Biology, Oregon Health and Science University, Portland, OR
| | - Mary McDonnell
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR
| | - Carl R. Pelz
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
- Computational Biology, Oregon Health and Science University, Portland, OR
| | - Brett Johnson
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR
| | - Kelly J. Hamman
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
| | - Jone E. Sampson
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
| | - Terry K. Morgan
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
- Department of Pathology, Oregon Health and Science University, Portland, OR
- Knight Cancer Institute, Portland, OR
| | - Charles D. Lopez
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
- Department of Hematology and Oncology, Portland, OR
- Knight Cancer Institute, Portland, OR
| | - Lisa M. Coussens
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Portland, OR
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR
- Knight Cancer Institute, Portland, OR
| |
Collapse
|
10
|
Clinical interpretation of PRSS1 variants in patients with pancreatitis. Clin Res Hepatol Gastroenterol 2021; 45:101497. [PMID: 33257277 DOI: 10.1016/j.clinre.2020.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/01/2020] [Indexed: 02/04/2023]
Abstract
Since the description of the PRSS1 gene encoding the cationic trypsinogen as being involved in dominant hereditary pancreatitis, more than 50 PRSS1 variants have been reported. Among the PRSS1 variants that have been classified as pathogenic, some have a high penetrance and others have a low penetrance. Assessing the clinical relevance of PRSS1 variants is often complicated in the absence of functional evidence and interpretation of rare variants is not very easy in clinical practice. The aim of this study was to review the different variants identified in the PRSS1 gene and to classify them according to their degree of deleterious effect. This classification was based on the results of several in vitro experiments and on population data, in comparing the allelic frequency of each variant in patients with pancreatitis and in unaffected individuals. This review should help geneticists and clinicians in charge of patient's care and genetic counseling to interpret molecular results.
Collapse
|
11
|
Huang H, Swidnicka-Siergiejko AK, Daniluk J, Gaiser S, Yao Y, Peng L, Zhang Y, Liu Y, Dong M, Zhan X, Wang H, Bi Y, Li Z, Ji B, Logsdon CD. Transgenic Expression of PRSS1 R122H Sensitizes Mice to Pancreatitis. Gastroenterology 2020; 158:1072-1082.e7. [PMID: 31419436 PMCID: PMC7580257 DOI: 10.1053/j.gastro.2019.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 07/19/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Mutations in the trypsinogen gene (PRSS1) cause human hereditary pancreatitis. However, it is not clear how mutant forms of PRSS1 contribute to disease development. We studied the effects of expressing mutant forms of human PRSS1 in mice. METHODS We expressed forms of PRSS1 with and without the mutation encoding R122H (PRSS1R122H) specifically in pancreatic acinar cells under control of a full-length pancreatic elastase gene promoter. Mice that did not express these transgenes were used as controls. Mice were given injections of caerulein to induce acute pancreatitis or injections of lipopolysaccharide to induce chronic pancreatitis. Other groups of mice were fed ethanol or placed on a high-fat diet to induce pancreatitis. Pancreata were collected and analyzed by histology, immunoblots, real-time polymerase chain reaction, and immunohistochemistry. Trypsin enzymatic activity and chymotrypsin enzymatic activity were measured in pancreatic homogenates. Blood was collected and serum amylase activity was measured. RESULTS Pancreata from mice expressing transgenes encoding PRSS1 or PRSS1R122H had focal areas of inflammation; these lesions were more prominent in mice that express PRSS1R122H. Pancreata from mice that express PRSS1 or PRSS1R122H had increased levels of heat shock protein 70 and nuclear factor (erythroid-derived 2)-like 2, and reduced levels of chymotrypsin C compared with control mice. Increased expression of PRSS1 or PRSS1R122H increased focal damage in pancreatic tissues and increased the severity of acute pancreatitis after caerulein injection. Administration of lipopolysaccharide exacerbated inflammation in mice that express PRSS1R122H compared to mice that express PRSS1 or control mice. Mice that express PRSS1R122H developed more severe pancreatitis after ethanol feeding or a high-fat diet than mice that express PRSS1 or control mice. Pancreata from mice that express PRSS1R122H had more DNA damage, apoptosis, and collagen deposition and increased trypsin activity and infiltration by inflammatory cells than mice that express PRSS1 or control mice. CONCLUSIONS Expression of a transgene encoding PRSS1R122H in mice promoted inflammation and increased the severity of pancreatitis compared with mice that express PRSS1 or control mice. These mice might be used as a model for human hereditary pancreatitis and can be studied to determine mechanisms of induction of pancreatitis by lipopolysaccharide, ethanol, or a high-fat diet.
Collapse
Affiliation(s)
- Haojie Huang
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX,Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Agnieszka Katarzyna Swidnicka-Siergiejko
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX,Department of Gastroenterology, Medical University of Bialystok, Bialystok, Poland
| | - Jaroslaw Daniluk
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX,Department of Gastroenterology, Medical University of Bialystok, Bialystok, Poland
| | - Sebastian Gaiser
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Yao Yao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Lisi Peng
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Yang Zhang
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX,Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Yan Liu
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Minyu Dong
- Department of Gastroenterology, Guangzhou Medical University, Guangzhou, China,Department of Cancer Biology, Mayo Clinic, Jacksonville, FL
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China,Department of Cancer Biology, Mayo Clinic, Jacksonville, FL
| | - Huamin Wang
- Department of Pathology, the University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Yan Bi
- Department of Gastroenterology, Mayo Clinic, Jacksonville, FL
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.
| | - Craig D. Logsdon
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
12
|
ATF6 regulates the development of chronic pancreatitis by inducing p53-mediated apoptosis. Cell Death Dis 2019; 10:662. [PMID: 31506423 PMCID: PMC6737032 DOI: 10.1038/s41419-019-1919-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/31/2019] [Accepted: 08/26/2019] [Indexed: 01/20/2023]
Abstract
Chronic pancreatitis (CP) is a progressive, recurrent inflammatory disorder of the pancreas. Initiation and progression of CP can result from serine protease 1 (PRSS1) overaccumulation and the ensuing endoplasmic reticulum (ER) stress. However, how ER stress pathways regulate the development and progression of CP remains poorly understood. In the present study we aimed to elucidate the ER stress pathway involved in CP. We found high expression of the ER stress marker genes ATF6, XBP1, and CHOP in human clinical specimens. A humanized PRSS1 transgenic mouse was established and treated with caerulein to mimic the development of CP, as evidenced by pathogenic alterations, collagen deposition, and increased expression of the inflammatory factors IL-6, IL-1β, and TNF-α. ATF6, XBP1, and CHOP expression levels were also increased during CP development in this model. Acinar cell apoptosis was also significantly increased, accompanied by upregulated p53 expression. Inhibition of ATF6 or p53 suppressed the expression of inflammatory factors and progression of CP in the mouse model. Finally, we showed that p53 expression could be regulated by the ATF6/XBP1/CHOP axis to promote the development of CP. We therefore conclude that ATF6 signalling regulates CP progression by modulating pancreatic acinar cell apoptosis, which provides a target for ER stress-based diagnosis and treatment of CP.
Collapse
|
13
|
Guerrero-Olazarán M, Castillo-Galván M, Gallegos-López JA, Fuentes-Garibay JA, Viader-Salvadó JM. Biochemical characterization of recombinant Penaeus vannamei trypsinogen. Comp Biochem Physiol B Biochem Mol Biol 2019; 238:110337. [PMID: 31476362 DOI: 10.1016/j.cbpb.2019.110337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/09/2019] [Accepted: 08/28/2019] [Indexed: 11/26/2022]
Abstract
Trypsinogens are the inactive precursors of trypsins (EC 3.4.21.4), which are digestive serine proteases. Despite knowing the properties of trypsins from Pacific white shrimp, Penaeus vannamei, the biochemical properties of shrimp trypsinogens including activation mechanisms and kinetics are unknown, due to difficulties isolating them from natural sources. In the present work, we describe the purification and biochemical characterization of four trypsinogen-like isoforms from recombinant P. vannamei trypsinogen, with a special emphasis on understanding its activation kinetics. The major trypsinogen-like isoform had an apparent molecular mass of 29 kDa. The other three forms of recombinant trypsinogen were: an N-glycosylated form of 32 kDa, a possibly O-glycosylated form of 41 kDa, and a likely double-chain form with a subunit of 23 kDa. The autoactivation profile of three-recombinant trypsinogen-like isoforms showed increased trypsin activity at a rate that was higher than that of bovine trypsinogen. This confirms the hypothesis proposed in the literature of a rapid trypsinogen autoactivation in the absence of aspartates in the activation peptide as it is for P. vannamei trypsinogen.
Collapse
Affiliation(s)
- Martha Guerrero-Olazarán
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Instituto de Biotecnología, 66455 San Nicolás de los Garza, NL, Mexico
| | - Mauricio Castillo-Galván
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Instituto de Biotecnología, 66455 San Nicolás de los Garza, NL, Mexico
| | - Juan Antonio Gallegos-López
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Instituto de Biotecnología, 66455 San Nicolás de los Garza, NL, Mexico
| | - José Antonio Fuentes-Garibay
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Instituto de Biotecnología, 66455 San Nicolás de los Garza, NL, Mexico
| | - José María Viader-Salvadó
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Instituto de Biotecnología, 66455 San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
14
|
Zhan X, Wan J, Zhang G, Song L, Gui F, Zhang Y, Li Y, Guo J, Dawra RK, Saluja AK, Haddock AN, Zhang L, Bi Y, Ji B. Elevated intracellular trypsin exacerbates acute pancreatitis and chronic pancreatitis in mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G816-G825. [PMID: 30943050 PMCID: PMC6620583 DOI: 10.1152/ajpgi.00004.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 01/31/2023]
Abstract
Intra-acinar trypsinogen activation occurs in the earliest stages of pancreatitis and is believed to play important roles in pancreatitis pathogenesis. However, the exact role of intra-acinar trypsin activity in pancreatitis remains elusive. Here, we aimed to examine the specific effects of intra-acinar trypsin activity on the development of pancreatitis using a transgenic mouse model. This transgenic mouse model allowed for the conditional expression of a mutant trypsinogen that can be activated specifically inside pancreatic acinar cells. We found that expression of this active mutated trypsin had no significant effect on triggering spontaneous pancreatitis. Instead, several protective compensatory mechanisms, including SPINK1 and heat shock proteins, were upregulated. Notably, these transgenic mice developed much more severe acute pancreatitis, compared with control mice, when challenged with caerulein. Elevated tissue edema, serum amylase, inflammatory cell infiltration and acinar cell apoptosis were dramatically associated with increased trypsin activity. Furthermore, chronic pathological changes were observed in the pancreas of all transgenic mice, including inflammatory cell infiltration, parenchymal atrophy and cell loss, fibrosis, and fatty replacement. These changes were not observed in control mice treated with caerulein. The alterations in pancreata from transgenic mice mimicked the histological changes common to human chronic pancreatitis. Taken together, we provided in vivo evidence that increased intra-acinar activation of trypsinogen plays an important role in the initiation and progression of both acute and chronic pancreatitis. NEW & NOTEWORTHY Trypsinogen is activated early in pancreatitis. However, the roles of trypsin in the development of pancreatitis have not been fully addressed. Using a genetic approach, we showed trypsin activity is critical for the severity of both acute and chronic pancreatitis.
Collapse
Affiliation(s)
- Xianbao Zhan
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
- Department of Oncology, Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Jianhua Wan
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Guowei Zhang
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University , Guangzhou , China
| | - Lele Song
- Department of Oncology, Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Fu Gui
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Yuebo Zhang
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Yinghua Li
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Jia Guo
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Rajinder K Dawra
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami , Miami, Florida
| | - Ashok K Saluja
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami , Miami, Florida
| | - Ashley N Haddock
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Lizhi Zhang
- Department of Pathology, Mayo Clinic , Rochester, Minnesota
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic , Jacksonville, Florida
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| |
Collapse
|
15
|
Saluja A, Dudeja V, Dawra R, Sah RP. Early Intra-Acinar Events in Pathogenesis of Pancreatitis. Gastroenterology 2019; 156:1979-1993. [PMID: 30776339 DOI: 10.1053/j.gastro.2019.01.268] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/09/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Abstract
Premature activation of digestive enzymes in the pancreas has been linked to development of pancreatitis for more than a century. Recent development of novel models to study the role of pathologic enzyme activation has led to advances in our understanding of the mechanisms of pancreatic injury. Colocalization of zymogen and lysosomal fraction occurs early after pancreatitis-causing stimulus. Cathepsin B activates trypsinogen in these colocalized organelles. Active trypsin increases permeability of these organelles resulting in leakage of cathepsin B into the cytosol leading to acinar cell death. Although trypsin-mediated cell death leads to pancreatic injury in early stages of pancreatitis, multiple parallel mechanisms, including activation of inflammatory cascades, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction in the acinar cells are now recognized to be important in driving the profound systemic inflammatory response and extensive pancreatic injury seen in acute pancreatitis. Chymotrypsin, another acinar protease, has recently been shown be play critical role in clearance of pathologically activated trypsin protecting against pancreatic injury. Mutations in trypsin and other genes thought to be associated with pathologic enzyme activation (such as serine protease inhibitor 1) have been found in familial forms of pancreatitis. Sustained intra-acinar activation of nuclear factor κB pathway seems to be key pathogenic mechanism in chronic pancreatitis. Better understanding of these mechanisms will hopefully allow us to improve treatment strategies in acute and chronic pancreatitis.
Collapse
|
16
|
Abu-El-Haija M, Lowe ME. Pediatric Pancreatitis-Molecular Mechanisms and Management. Gastroenterol Clin North Am 2018; 47:741-753. [PMID: 30337030 DOI: 10.1016/j.gtc.2018.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pediatric pancreatitis is an emerging field with an increasing incidence of disease. Management of pediatric pancreatitis is understudied and, therefore, extrapolated from adult studies (although the etiologies are different). There is evidence that feeding is safe in mild acute pancreatitis in children without increased pain or length of stay. Studies are needed to predict course of the disease, disease severity, and risk of chronic pancreatitis in children.
Collapse
Affiliation(s)
- Maisam Abu-El-Haija
- Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave MLC 2010, Cincinnati, Ohio 45229, USA
| | - Mark E Lowe
- Pediatric Gastroenterology, Hepatology and Nutrition, Washington University School of Medicine, 660 South Euclid Avenue, MPRB 4th Floor, Campus Box 8208, St Louis, MO 63110, USA.
| |
Collapse
|
17
|
Abstract
Individuals with acute recurrent and chronic pancreatitis may have an inherited predisposition to the development of the disease. Pancreatitis in the setting of a significant family history of the disease can be classified as hereditary or familial pancreatitis. In this article, the authors closely examine the specific genes implicated in pancreatitis, investigate the role of genetic testing for diagnosis, and describe the impact of genetic testing results on clinical management.
Collapse
Affiliation(s)
- Aws Hasan
- Department of Internal Medicine, Columbia University Medical Center, 630 West 168 Street, New York, NY, 10032, USA
| | - Dagmara I Moscoso
- Division of Digestive and Liver Diseases, Columbia University Medical Center, 630 West 168 Street, New York, NY, 10032, USA
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, 161 Fort Washington Avenue, Suite 862, New York, NY 10032, USA.
| |
Collapse
|
18
|
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: 4.3] [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
|
19
|
Cho SM, Shin S, Lee KA. PRSS1, SPINK1, CFTR, and CTRC Pathogenic Variants in Korean Patients With Idiopathic Pancreatitis. Ann Lab Med 2017; 36:555-60. [PMID: 27578509 PMCID: PMC5011109 DOI: 10.3343/alm.2016.36.6.555] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/15/2016] [Accepted: 07/19/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND This study aimed to identify pathogenic variants of PRSS1, SPINK1, CFTR, and CTRC genes in Korean patients with idiopathic pancreatitis. METHODS The study population consisted of 116 Korean subjects (65 males, 51 females; mean age, 30.4 yr, range, 1-88 yr) diagnosed with idiopathic chronic pancreatitis (ICP), idiopathic recurrent acute pancreatitis (IRAP), or idiopathic acute pancreatitis (IAP). We analyzed sequences of targeted regions in the PRSS1, SPINK1, CFTR, and CTRC genes, copy numbers of PRSS1 and SPINK1, and clinical data from medical records. RESULTS We identified three types of pathogenic PRSS1 variants in 11 patients, including p.N29I (n=1), p.R122H (n=1), and p.G208A (n=9). Sixteen patients exhibited heterozygous pathogenic variants of SPINK1, including c.194+2T>C (n=12), p.N34S (n=3), and a novel pathogenic splicing variation c.194+1G>A. A heterozygous CFTR p.Q1352H pathogenic variant was detected in eight patients. One patient carried a heterozygous CTRC p.P249L pathogenic variant, which is a known high-risk variant for pancreatitis. All patients had normal PRSS1 and SPINK1 gene copy numbers. Weight loss occurred more frequently in patients carrying the p.G208A pathogenic variant, while pancreatic duct stones occurred more frequently in patients with the c.194+2T>C pathogenic variant. CONCLUSIONS Pathogenic variants of PRSS1, SPINK1, and CFTR were associated with idiopathic pancreatitis, while pathogenic variants of CTRC were not. Copy number variations of PRSS1 and SPINK1 were not detected.
Collapse
Affiliation(s)
- Sun Mi Cho
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Kyung A Lee
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Chronic Pancreatitis in the 21st Century - Research Challenges and Opportunities: Summary of a National Institute of Diabetes and Digestive and Kidney Diseases Workshop. Pancreas 2016; 45:1365-1375. [PMID: 27748719 PMCID: PMC5117429 DOI: 10.1097/mpa.0000000000000713] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A workshop was sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases to focus on research gaps and opportunities in chronic pancreatitis (CP) and its sequelae. This conference marked the 20th year anniversary of the discovery of the cationic trypsinogen (PRSS1) gene mutation for hereditary pancreatitis. The event was held on July 27, 2016, and structured into 4 sessions: (1) pathophysiology, (2) exocrine complications, (3) endocrine complications, and (4) pain. The current state of knowledge was reviewed; many knowledge gaps and research needs were identified that require further investigation. Common themes included the need to design better tools to diagnose CP and its sequelae early and reliably, identify predisposing risk factors for disease progression, develop standardized protocols to distinguish type 3c diabetes mellitus from other types of diabetes, and design effective therapeutic strategies through novel cell culture technologies, animal models mimicking human disease, and pain management tools. Gene therapy and cystic fibrosis conductance regulator potentiators as possible treatments of CP were discussed. Importantly, the need for CP end points and intermediate targets for future drug trials was emphasized.
Collapse
|
21
|
Brandl T, Simic O, Skaanderup PR, Namoto K, Berst F, Ehrhardt C, Schiering N, Mueller I, Woelcke J. Trypsin inhibitors for the treatment of pancreatitis. Bioorg Med Chem Lett 2016; 26:4340-4. [DOI: 10.1016/j.bmcl.2016.07.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 02/08/2023]
|
22
|
Oracz G, Kolodziejczyk E, Sobczynska-Tomaszewska A, Wejnarska K, Dadalski M, Grabarczyk AM, Kierkus J, Woynarowski M, Wertheim-Tysarowska K, Ryzko J, Bal J, Rygiel AM. The clinical course of hereditary pancreatitis in children - A comprehensive analysis of 41 cases. Pancreatology 2016; 16:535-41. [PMID: 27179762 DOI: 10.1016/j.pan.2016.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 03/17/2016] [Accepted: 04/16/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Available data from adult patients do not reflect natural course of hereditary pancreatitis (HP) in children. To date, no study has assessed the clinical course of HP in children. OBJECTIVE To investigate the clinical course of HP in children and compare it to non-HP group with chronic pancreatitis (CP). METHODS A group of 265 children with CP, hospitalized from 1988 to 2014, were enrolled in the study. Medical records of those patients were reviewed for data on presentation, diagnostic findings and treatment. All children were screened for mutations in major pancreatitis-associated genes, i.e. PRSS1, SPINK1, and CFTR. RESULTS HP was diagnosed in 41 children (15.5%). Family history was positive in 88% of children with HP. Mutations of PRSS1 gene were found in 80% (33/41) of HP patients. We detected p.R122H, p.R122C, p.N29I, and p.E79K mutation in 34% (14/41), 27% (11/41), 12% (5/41), and 7% (3/41) of HP patients, respectively. Patients with paternal inheritance had first symptoms earlier than those with maternal inheritance (5.9 vs. 9.1 years; P < 0.05). Children with HP showed more severe changes in ERCP then those from non-HP group (2.05 Cambridge grade, vs. 1.6°; P < 0.05). ESWL was performed more frequently in HP group (12.2% vs. 3.1%; P < 0.05). There was no difference in age of disease onset (7.98 vs. 8.9 years; NS), pancreatic duct stenting (46.3% vs. 33%; NS), or number of surgical interventions (12.2% vs. 14.3%; NS) between both groups. CONCLUSIONS Children with HP reveal significantly more severe clinical presentation of the disease than non-HP patients, despite the same age of onset.
Collapse
Affiliation(s)
- Grzegorz Oracz
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland.
| | - Elwira Kolodziejczyk
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Agnieszka Sobczynska-Tomaszewska
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17A, 01-211 Warsaw, Poland; Medgen, ul. Orzycka 27, 02-695 Warsaw, Poland
| | - Karolina Wejnarska
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Maciej Dadalski
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Alicja Monika Grabarczyk
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17A, 01-211 Warsaw, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Marek Woynarowski
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | | | - Jozef Ryzko
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Jerzy Bal
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17A, 01-211 Warsaw, Poland
| | | |
Collapse
|
23
|
Abstract
Pancreatic adenocarcinoma is a leading cause of cancer death. Few patients are candidates for curative resection due to the late stage at diagnosis. While most pancreatic adenocarcinomas are sporadic, approximately 10% have an underlying hereditary basis. Known genetic syndromes account for only 20% of the familial clustering of pancreatic cancer cases. The majority are due to non-syndromic aggregation of pancreatic cancer cases or familial pancreatic cancer. Screening aims to identify high-risk lesions amenable to surgical resection. However, the optimal interval for screening and the management of pancreatic cancer precursor lesions detected on imaging are controversial.
Collapse
Affiliation(s)
- Shilpa Grover
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Kunal Jajoo
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
24
|
The genetic predisposition and its impact on the diabetes mellitus development in patients with alcoholic chronic pancreatitis. Gastroenterol Res Pract 2015; 2015:309156. [PMID: 25838820 PMCID: PMC4369946 DOI: 10.1155/2015/309156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/04/2015] [Indexed: 12/24/2022] Open
Abstract
The most common cause of chronic pancreatitis (CP) is alcohol abuse. The aim of the present study was to identify patients with genetic predisposition to CP abusing alcohol. The question posed was whether CP manifests at a younger age and diabetes mellitus develops earlier in individuals with genetic predisposition. The study encompassed 79 patients with alcoholic chronic pancreatitis (ACP) and control group (100 persons). The following mutations were determined: R122H and N29I of PRSS1 and N34S of SPINK1 as well as E366K and E288V of SERPINA 1. No R122H and N291 mutations were observed in the group of ACP patients and in controls. Moreover, there was no E288V mutation. In 79 ACP patients, six SPINK 1 (N34S/wt) mutations were observed. In the control group, one heterozygous SPINK 1N34S gene mutation was found (P = 0.0238). Two PiZ mutations were identified in patients with ACP and one analogical mutation in controls. Amongst patients with ACP as well as SPINK1 and PiZ mutations, the onset of disease was observed earlier and developed earlier. The prevalence of SPINK1 mutation is higher in patients with ACP than in healthy populations. This mutation together with the effects of alcohol accelerates the development of ACP and of diabetes mellitus.
Collapse
|
25
|
Protein surface charge of trypsinogen changes its activation pattern. BMC Biotechnol 2014; 14:109. [PMID: 25543846 PMCID: PMC4299543 DOI: 10.1186/s12896-014-0109-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/11/2014] [Indexed: 01/29/2023] Open
Abstract
Background Trypsinogen is the inactive precursor of trypsin, a serine protease that cleaves proteins and peptides after arginine and lysine residues. In this study, human trypsinogen was used as a model protein to study the influence of electrostatic forces on protein–protein interactions. Trypsinogen is active only after its eight-amino-acid-long activation peptide has been cleaved off by another protease, enteropeptidase. Trypsinogen can also be autoactivated without the involvement of enteropeptidase. This autoactivation process can occur if a trypsinogen molecule is activated by another trypsin molecule and therefore is based on a protein–protein interaction. Results Based on a rational protein design based on autoactivation-defective guinea pig trypsinogen, several amino acid residues, all located far away from the active site, were changed to modify the surface charge of human trypsinogen. The influence of the surface charge on the activation pattern of trypsinogen was investigated. The autoactivation properties of mutant trypsinogen were characterized in comparison to the recombinant wild-type enzyme. Surface-charged trypsinogen showed practically no autoactivation compared to the wild-type but could still be activated by enteropeptidase to the fully active trypsin. The kinetic parameters of surface-charged trypsinogen were comparable to the recombinant wild-type enzyme. Conclusion The variant with a modified surface charge compared to the wild-type enzyme showed a complete different activation pattern. Our study provides an example how directed modification of the protein surface charge can be utilized for the regulation of functional protein–protein interactions, as shown here for human trypsinogen. Electronic supplementary material The online version of this article (doi:10.1186/s12896-014-0109-5) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Sah RP, Garg SK, Dixit AK, Dudeja V, Dawra RK, Saluja AK. Endoplasmic reticulum stress is chronically activated in chronic pancreatitis. J Biol Chem 2014; 289:27551-61. [PMID: 25077966 DOI: 10.1074/jbc.m113.528174] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenesis of chronic pancreatitis (CP) is poorly understood. Endoplasmic reticulum (ER) stress has now been recognized as a pathogenic event in many chronic diseases. However, ER stress has not been studied in CP, although pancreatic acinar cells seem to be especially vulnerable to ER dysfunction because of their dependence on high ER volume and functionality. Here, we aim to investigate ER stress in CP, study its pathogenesis in relation to trypsinogen activation (widely regarded as the key event of pancreatitis), and explore its mechanism, time course, and downstream consequences during pancreatic injury. CP was induced in mice by repeated episodes of acute pancreatitis (AP) based on caerulein hyperstimulation. ER stress leads to activation of unfolded protein response components that were measured in CP and AP. We show sustained up-regulation of unfolded protein response components ATF4, CHOP, GRP78, and XBP1 in CP. Overexpression of GRP78 and ATF4 in human CP confirmed the experimental findings. We used novel trypsinogen-7 knock-out mice (T(-/-)), which lack intra-acinar trypsinogen activation, to clarify the relationship of ER stress to intra-acinar trypsinogen activation in pancreatic injury. Comparable activation of ER stress was seen in wild type and T(-/-) mice. Induction of ER stress occurred through pathologic calcium signaling very early in the course of pancreatic injury. Our results establish that ER stress is chronically activated in CP and is induced early in pancreatic injury through pathologic calcium signaling independent of trypsinogen activation. ER stress may be an important pathogenic mechanism in pancreatitis that needs to be explored in future studies.
Collapse
Affiliation(s)
- Raghuwansh P Sah
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Sushil K Garg
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Ajay K Dixit
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Vikas Dudeja
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Rajinder K Dawra
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Ashok K Saluja
- From the Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| |
Collapse
|
27
|
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: 5.5] [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
|
28
|
Chang MC, Wong JM, Chang YT. Screening and early detection of pancreatic cancer in high risk population. World J Gastroenterol 2014; 20:2358-2364. [PMID: 24605033 PMCID: PMC3942839 DOI: 10.3748/wjg.v20.i9.2358] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 01/05/2014] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a serious growing health issue in developed countries. For patients diagnosed with pancreatic cancer, the five year survival rate is below 5%. One major important reason leads to the poor survival rate is lack of early detection of pancreatic cancer. Over 80% of the patients are diagnosed in advanced disease stages. Screening for pancreatic cancer is a desirable option for high risk individuals to allow early detection and treatment of curable pancreatic neoplasms at a pre-invasive stage. This article highlights the need, endpoint, population, method, diagnostic yield, and the problems of current screening programs.
Collapse
|
29
|
Abstract
OBJECTIVE Hereditary pancreatitis is caused by mutations in human cationic trypsinogen (PRSS1) which lead to increased autoactivation by altering chymotrypsin C (CTRC)-dependent trypsinogen activation and degradation. Exceptions are some cysteine mutations which cause misfolding, intracellular retention and endoplasmic reticulum stress. Clinical relevance of many PRSS1 variants found in patients with sporadic chronic pancreatitis is unknown but often assumed by analogy with known disease-causing mutations. Functional comparison of PRSS1 variants found in sporadic and hereditary cases is needed to resolve this dilemma. DESIGN Here, we investigated the functional phenotype of 13 published PRSS1 variants with respect to autoactivation in the presence of CTRC and cellular secretion. RESULTS Only mutation p.D100H increased trypsinogen autoactivation, but this gain in function was offset by a marked reduction in secretion. Five mutants (p.P36R, p.G83E, p.I88N, p.V123M, p.S124F) showed decreased autoactivation due to increased degradation by CTRC. Five mutants exhibited strongly (p.D100H, p.C139F) or moderately (p.K92N, p.S124F, p.G208A) reduced secretion, whereas mutant p.K170E showed slightly increased secretion. Mutant p.I88N was also secreted to higher levels but was rapidly degraded by CTRC. Finally, three mutants (p.Q98K, p.T137M, p.S181G) had no phenotypic alterations relative to wild-type trypsinogen. CONCLUSIONS Rare PRSS1 variants found in sporadic chronic pancreatitis do not stimulate autoactivation but may cause increased degradation, impaired secretion or no functional change. Variants with reduced secretion are likely pathogenic due to mutation-induced misfolding and consequent endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Andrea Schnúr
- 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
| | - Sebastian Beer
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118
| | - Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ) & Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), 85350 Freising, Germany
- Department of Pediatrics, Klinikum rechts der Isar (MRI), Technische Universität München (TUM), 80804 Munich, Germany
| | - 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
- Correspondence to Miklós Sahin-Tóth, 72 East Concord Street, Evans-433; Boston, MA 02118; Tel: (617) 414-1070; Fax: (617) 414-1041;
| |
Collapse
|
30
|
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: 3.5] [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
|
31
|
Abstract
PURPOSE OF REVIEW In this article, we review important advances in our understanding of the mechanisms of pancreatitis. RECENT FINDINGS The relative contributions of intrapancreatic trypsinogen activation and nuclear factor kappa B (NFκB) activation, the two major early independent cellular events in pancreatitis, have been investigated using novel genetic models. Trypsinogen activation has traditionally held the spotlight for many decades as the central pathogenic event of pancreatitis. However, recent experimental evidence points to the role of trypsin activation in early acinar cell damage but not in the inflammatory response of acute pancreatitis, which was shown to be induced by NFκB activation. Further, chronic pancreatitis developed independently of trypsinogen activation in the caerulein model. Sustained NFκB activation, but not persistent intra-acinar expression of active trypsin, was shown to result in chronic pancreatitis. Calcineurin-NFAT (nuclear factor of activated T-cells) signaling was shown to mediate downstream effects of pathologic rise in intracellular calcium. Interleukin-6 was identified as a key cytokine mediating pancreatitis-associated lung injury. SUMMARY Recent advances challenge the long-believed trypsin-centered understanding of pancreatitis. It is becoming increasingly clear that activation of intense inflammatory signaling mechanisms in acinar cells is crucial to the pathogenesis of pancreatitis, which may explain the strong systemic inflammatory response in pancreatitis.
Collapse
|
32
|
Németh BC, Wartmann T, Halangk W, Sahin-Tóth M. Autoactivation of mouse trypsinogens is regulated by chymotrypsin C via cleavage of the autolysis loop. J Biol Chem 2013; 288:24049-62. [PMID: 23814066 DOI: 10.1074/jbc.m113.478800] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chymotrypsin C (CTRC) is a proteolytic regulator of trypsinogen autoactivation in humans. CTRC cleavage of the trypsinogen activation peptide stimulates autoactivation, whereas cleavage of the calcium binding loop promotes trypsinogen degradation. Trypsinogen mutations that alter these regulatory cleavages lead to increased intrapancreatic trypsinogen activation and cause hereditary pancreatitis. The aim of this study was to characterize the regulation of autoactivation of mouse trypsinogens by mouse Ctrc. We found that the mouse pancreas expresses four trypsinogen isoforms to high levels, T7, T8, T9, and T20. Only the T7 activation peptide was cleaved by mouse Ctrc, causing negligible stimulation of autoactivation. Surprisingly, mouse Ctrc poorly cleaved the calcium binding loop in all mouse trypsinogens. In contrast, mouse Ctrc readily cleaved the Phe-150-Gly-151 peptide bond in the autolysis loop of T8 and T9 and inhibited autoactivation. Mouse chymotrypsin B also cleaved the same peptide bond but was 7-fold slower. T7 was less sensitive to chymotryptic regulation, which involved slow cleavage of the Leu-149-Ser-150 peptide bond in the autolysis loop. Modeling indicated steric proximity of the autolysis loop and the activation peptide in trypsinogen, suggesting the cleaved autolysis loop may directly interfere with activation. We conclude that autoactivation of mouse trypsinogens is under the control of mouse Ctrc with some notable differences from the human situation. Thus, cleavage of the trypsinogen activation peptide or the calcium binding loop by Ctrc is unimportant. Instead, inhibition of autoactivation via cleavage of the autolysis loop is the dominant mechanism that can mitigate intrapancreatic trypsinogen activation.
Collapse
Affiliation(s)
- Balázs Csaba Németh
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
33
|
Sah RP, Dudeja V, Dawra RK, Saluja AK. Cerulein-induced chronic pancreatitis does not require intra-acinar activation of trypsinogen in mice. Gastroenterology 2013; 144:1076-1085.e2. [PMID: 23354015 PMCID: PMC3928043 DOI: 10.1053/j.gastro.2013.01.041] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/02/2013] [Accepted: 01/07/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Premature activation of trypsinogen activation can cause pancreatic injury and has been associated with chronic pancreatitis (CP). Mice that lack intra-acinar activation of trypsinogen, such as trypsinogen-7-null (T(-/-)) and cathepsin B-null (CB(-/-)) mice, have been used to study trypsin-independent processes of CP development. We compared histologic features and inflammatory responses of pancreatic tissues from these mice with those from wild-type mice after the development of CP. METHODS CP was induced in wild-type, T(-/-), and CB(-/-) mice by twice-weekly induction of acute pancreatitis for 10 weeks; acute pancreatitis was induced by hourly intraperitoneal injections of cerulein (50 μg/kg × 6). Pancreatic samples were collected and evaluated by histologic and immunohistochemical analyses. Normal human pancreas samples, obtained from the islet transplant program at the University of Minnesota, were used as controls and CP samples were obtained from surgical resections. RESULTS Compared with pancreatic tissues from wild-type mice, those from T(-/-) and CB(-/-) mice had similar levels of atrophy, histomorphologic features of CP, and chronic inflammation. All samples had comparable intra-acinar activation of nuclear factor (NF)-κB, a transcription factor that regulates the inflammatory response, immediately after injection of cerulein. Pancreatic tissue samples from patients with CP had increased activation of NF-κB (based on nuclear translocation of p65 in acinar cells) compared with controls. CONCLUSIONS Induction of CP in mice by cerulein injection does not require intra-acinar activation of trypsinogen. Pancreatic acinar cells of patients with CP have increased levels of NF-κB activation compared with controls; regulation of the inflammatory response by this transcription factor might be involved in the pathogenesis of CP.
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: 2.5] [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
|
Agarwal A, Boettcher A, Kneuer R, Sari-Sarraf F, Donovan A, Woelcke J, Simic O, Brandl T, Krucker T. In vivo imaging with fluorescent smart probes to assess treatment strategies for acute pancreatitis. PLoS One 2013; 8:e55959. [PMID: 23409095 PMCID: PMC3569412 DOI: 10.1371/journal.pone.0055959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 01/07/2013] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND AIMS Endoprotease activation is a key step in acute pancreatitis and early inhibition of these enzymes may protect from organ damage. In vivo models commonly used to evaluate protease inhibitors require animal sacrifice and therefore limit the assessment of dynamic processes. Here, we established a non-invasive fluorescence imaging-based biomarker assay to assess real-time protease inhibition and disease progression in a preclinical model of experimental pancreatitis. METHODS Edema development and trypsin activation were imaged in a rat caerulein-injection pancreatitis model. A fluorescent "smart" probe, selectively activated by trypsin, was synthesized by labeling with Cy5.5 of a pegylated poly-L-lysine copolymer. Following injection of the probe, trypsin activation was monitored in the presence or absence of inhibitors by in vivo and ex vivo imaging. RESULTS We established the trypsin-selectivity of the fluorescent probe in vitro using a panel of endopeptidases and specific inhibitor. In vivo, the probe accumulated in the liver and a region attributed to the pancreas by necropsy. A dose dependent decrease of total pancreatic fluorescence signal occurred upon administration of known trypsin inhibitors. The fluorescence-based method was a better predictor of trypsin inhibition than pancreatic to body weight ratio. CONCLUSIONS We established a fluorescence imaging assay to access trypsin inhibition in real-time in vivo. This method is more sensitive and dynamic than classic tissue sample readouts and could be applied to preclinically optimize trypsin inhibitors towards intrapancreatic target inhibition.
Collapse
Affiliation(s)
- Abhiruchi Agarwal
- Novartis Institute of BioMedical Research, Cambridge, Massachusetts, United States of America
| | | | - Rainer Kneuer
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Farid Sari-Sarraf
- Novartis Institute of BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Adriana Donovan
- Novartis Institute of BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Julian Woelcke
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Oliver Simic
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Trixi Brandl
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Krucker
- Novartis Institute of BioMedical Research, Cambridge, Massachusetts, United States of America
- Novartis Institutes of BioMedical Research, Emeryville, California, United States of America
| |
Collapse
|
36
|
Liu J, Zhang HX. A comprehensive study indicates PRSS1 gene is significantly associated with pancreatitis. Int J Med Sci 2013; 10:981-7. [PMID: 23801884 PMCID: PMC3691796 DOI: 10.7150/ijms.6164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/14/2013] [Indexed: 12/14/2022] Open
Abstract
This comprehensive meta-analysis was applied to case-control studies of the association between pancreatitis and PRSS1 gene to assess the joint evidence for the association, the influence of individual studies, and evidence for publication bias. PubMed, EMBASE, and Cochrane Library were searched in order to identify longitudinal studies evaluating pancreatitis disease and PRSS1 gene. Odds ratios (ORs) were pooled using a random-effects model. For the case-control studies, the authors found 1) support for the association between total pancreatitis and PRSS1 gene, both totally analyzed and subdivided analyzed {total: [OR:10.799, 95%CI:(5.489-21.242), p<0.000]; Europe: [OR:9.795, 95%CI:(2.923-32.819), p<0.000]; Asia: [OR:11.994, 95%CI:(5.156-27.898), p<0.000]}. 2) no evidence showed that this association was accounted for by any one study, and 3) no evidence showed any publication bias exist. In conclusion, PRSS1 gene was significantly associated with total pancreatitis disease, both totally and separately.
Collapse
Affiliation(s)
- Jie Liu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Orthopaedics and Traumatology, Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | | |
Collapse
|
37
|
Loos M, Michalski CW, Kleeff J. Asymptomatic pancreatic lesions: New insights and clinical implications. World J Gastroenterol 2012; 18:4474-7. [PMID: 22969218 PMCID: PMC3435770 DOI: 10.3748/wjg.v18.i33.4474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/03/2012] [Accepted: 08/15/2012] [Indexed: 02/06/2023] Open
Abstract
Despite great efforts in experimental and clinical research, the prognosis of pancreatic cancer (PC) has not changed significantly for decades. Detection of pre-invasive lesions or early-stage PC with small resectable cancers in asymptomatic individuals remains one of the most promising approaches to substantially improve the overall outcome of PC. Therefore, screening programs have been proposed to identify curable lesions especially in individuals with a familial or genetic predisposition for PC. In this regard, Canto et al recently contributed an important article comparing computed tomography, magnetic resonance imaging, and endoscopic ultrasound for the screening of 216 asymptomatic high-risk individuals (HRI). Pancreatic lesions were detected in 92 of 216 asymptomatic HRI (42.6%). The high diagnostic yield in this study raises several questions that need to be answered of which two will be discussed in detail in this commentary: First: which imaging test should be performed? Second and most importantly: what are we doing with incidentally detected pancreatic lesions? Which ones can be observed and which ones need to be resected?
Collapse
|
38
|
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
|
39
|
Ohmuraya M, Sugano A, Hirota M, Takaoka Y, Yamamura KI. Role of Intrapancreatic SPINK1/Spink3 Expression in the Development of Pancreatitis. Front Physiol 2012; 3:126. [PMID: 22586407 PMCID: PMC3345944 DOI: 10.3389/fphys.2012.00126] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 04/16/2012] [Indexed: 12/17/2022] Open
Abstract
Studies on hereditary pancreatitis have provided evidence in favor of central role for trypsin activity in the disease. Identification of genetic variants of trypsinogen linked the protease to the onset of pancreatitis, and biochemical characterization proposed an enzymatic gain of function as the initiating mechanism. Mutations of serine protease inhibitor Kazal type 1 gene (SPINK1) are shown to be associated with hereditary pancreatitis. We previously reported that Spink3 (a mouse homolog gene of human SPINK1) deficient mice showed excessive autophagy, followed by inappropriate trypsinogen activation in the exocrine pancreas. These data indicate that the role of SPINK1/Spink3 is not only trypsin inhibitor, but also negative regulator of autophagy. On the other hand, recent studies showed that high levels of SPINK1 protein detected in a serum or urine were associated with adverse outcome in various cancer types. It has been suggested that expression of SPINK1 and trypsin is balanced in normal tissue, but this balance could be disrupted during tumor progression. Based on the structural similarity between SPINK1 and epidermal growth factor (EGF), we showed that SPINK1 protein binds and activates EGF receptor, thus acting as a growth factor on tumor cell lines. In this review, we summarize the old and new roles of SPINK1/Spink3 in trypsin inhibition, autophagy, and cancer cell growth. These new functions of SPINK1/Spink3 may be related to the development of chronic pancreatitis.
Collapse
Affiliation(s)
- Masaki Ohmuraya
- Institute of Resource Development and Analysis, Kumamoto University Kumamoto, Japan
| | | | | | | | | |
Collapse
|
40
|
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: 5.8] [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
|
41
|
Abstract
Accumulating data indicate that clinically available abdominal imaging tests such as EUS and MRI/MRCP can detect asymptomatic precursor benign (IPMN, PanIN) and invasive malignant pancreatic neoplasms, such as ductal adenocarcinoma, in individuals with an inherited predisposition. These asymptomatic FPCs detected have been more likely to be resectable, compared to symptomatic tumors. The most challenging part of screening high-risk individuals is the selection of individuals with high-grade precursor neoplasms for preventive treatment (ie, surgical resection before development of invasive cancer). Ongoing and future research should focus on formulating and validating a model for FPC risk and neoplastic progression using patient characteristics, imaging, and biomarkers. The comparative cost and effectiveness of various approaches for screening and surveillance of high-risk individuals also deserves study. For now, screening is best performed in high-risk individuals within the research protocols in academic centers with multidisciplinary teams with expertise in genetics, gastroenterology, radiology, surgery, and pathology.
Collapse
|
42
|
Dawra R, Sah RP, Dudeja V, Rishi L, Saluja AK, Garg P, Saluja AK. Intra-acinar trypsinogen activation mediates early stages of pancreatic injury but not inflammation in mice with acute pancreatitis. Gastroenterology 2011; 141:2210-2217.e2. [PMID: 21875495 PMCID: PMC3587766 DOI: 10.1053/j.gastro.2011.08.033] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/11/2011] [Accepted: 08/18/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The role of trypsinogen activation in the pathogenesis of acute pancreatitis (AP) has not been clearly established. METHODS We generated and characterized mice lacking trypsinogen isoform 7 (T7) gene (T(-/-)). The effects of pathologic activation of trypsinogen were studied in these mice during induction of AP with cerulein. Acinar cell death, tissue damage, early intra-acinar activation of the transcription factor nuclear factor κB (NF-κB), and local and systemic inflammation were compared between T(-/-) and wild-type mice with AP. RESULTS Deletion of T7 reduced the total trypsinogen content by 60% but did not affect physiologic function. T(-/-) mice lacked pathologic activation of trypsinogen, which occurs within acinar cells during early stages of AP progression. Absence of trypsinogen activation in T(-/-) mice led to near complete inhibition of acinar cell death in vitro and a 50% reduction in acinar necrosis during AP progression. However, T(-/-) mice had similar degrees of local and systemic inflammation during AP progression and comparable levels of intra-acinar NF-κB activation, which was previously shown to occur concurrently with trypsinogen activation during early stages of pancreatitis. CONCLUSIONS T7 is activated during pathogenesis of AP in mice. Intra-acinar trypsinogen activation leads to acinar death during early stages of pancreatitis, which accounts for 50% of the pancreatic damage in AP. However, progression of local and systemic inflammation in AP does not require trypsinogen activation. NF-κB is activated early in acinar cells, independently of trypsinogen activation, and might be responsible for progression of AP.
Collapse
|
43
|
Familial pancreatic cancer and hereditary syndromes: screening strategy for high-risk individuals. J Gastroenterol 2011; 46:1249-59. [PMID: 21847571 DOI: 10.1007/s00535-011-0457-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/25/2011] [Indexed: 02/04/2023]
Abstract
Globally, and almost evenly across nations, a familial disposition can be found in 4-10% of patients with pancreatic cancer (PC). A family history of PC is a risk for this disease and the risk level changes in correlation with the number of affected relatives. Several hereditary syndromes with potential germline mutation also have a high risk for PC; however, little is yet known regarding the genes responsible for familial pancreatic cancer (FPC). Characteristics of FPC cases are similar to those of other familial tumors, including younger onset than in sporadic cases and an ethnic difference (Ashkenazi Jewish > other Caucasian). Other risks resemble those of sporadic cases and include smoking and diabetes mellitus. People with several genetic syndromes, including Peutz-Jeghers syndrome, hereditary pancreatitis, breast-ovarian cancer syndrome, hereditary nonpolyposis colorectal cancer, and familial adenomatous polyposis also have an increased risk of PC. In many countries, but not yet in Japan, screening of these high-risk individuals is now ongoing for the detection of early PC under established familial pancreatic cancer registries. In addition to the ordinary risk factors, such as smoking, diabetes, pancreatitis, cysts, duct ectasia, and intraductal papillary mucinous neoplasm (IPMN), individuals with a family history of PC and hereditary syndromes are expected to be entered into the screening protocol.
Collapse
|
44
|
Gaiser S, Daniluk J, Liu Y, Tsou L, Chu J, Lee W, Longnecker DS, Logsdon CD, Ji B. Intracellular activation of trypsinogen in transgenic mice induces acute but not chronic pancreatitis. Gut 2011; 60:1379-88. [PMID: 21471572 PMCID: PMC4304390 DOI: 10.1136/gut.2010.226175] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Premature intra-acinar activation of trypsinogen is widely considered key for both the initiation of acute pancreatitis and the development of chronic pancreatitis. However, the biological consequences of intracellular trypsinogen activation have not been directly examined. To do so, a new mouse model was developed. METHODS Mice were engineered to conditionally express an endogenously activated trypsinogen within pancreatic acinar cells (PACE-tryp(on)). Hallmarks of pancreatitis were determined and findings were correlated to the level (zygosity) and extent (temporal and spatial) of conditional PACE-tryp(on) expression. Furthermore, the impact of acinar cell death in PACE-tryp(on) mice was assessed and compared with a model of selective diphtheria toxin (DT)-mediated induction of acinar apoptosis. RESULTS Initiation of acute pancreatitis was observed with high (homozygous), but not low (heterozygous) levels of PACE-tryp(on) expression. Subtotal (maximal-rapid induction) but not limited (gradual-repetitive induction) conditional PACE-tryp(on) expression was associated with systemic complications and mortality. Rapid caspase-3 activation and apoptosis with delayed necrosis was observed, and loss of acinar cells led to replacement with fatty tissue. Chronic inflammation or fibrosis did not develop. Selective depletion of pancreatic acinar cells by apoptosis using DT evoked similar consequences. CONCLUSIONS Intra-acinar activation of trypsinogen is sufficient to initiate acute pancreatitis. However, the primary response to intracellular trypsin activity is rapid induction of acinar cell death via apoptosis which facilitates resolution of the acute inflammation rather than causing chronic pancreatitis. This novel model provides a powerful tool to improve our understanding of basic mechanisms occurring during pancreatitis.
Collapse
Affiliation(s)
- Sebastian Gaiser
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Jaroslaw Daniluk
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Yan Liu
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Lilian Tsou
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Jun Chu
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Woojin Lee
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Daniel S Longnecker
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Craig D Logsdon
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
- Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Baoan Ji
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Abstract
Trypsinogen activation is sufficient to induce acute pancreatitis in an experimental model. However, whether it is a requirement for the pathogenesis of acute and chronic pancreatitis remains to be explored.
Collapse
Affiliation(s)
- Raghuwansh P Sah
- Department of Surgery, Division of Basic and Translational Research, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
46
|
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)
| | - Miklós Sahin-Tóth
- To whom correspondence should be addressed: 72 East Concord Street, Evans-433, Boston, MA 02118, USA.
| |
Collapse
|
47
|
Farris AB, Basturk O, Adsay NV. Pancreatitis, Other Inflammatory Lesions, and Pancreatic Pseudotumors. Surg Pathol Clin 2011; 4:625-650. [PMID: 26837491 DOI: 10.1016/j.path.2011.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The pancreas is versatile in the diversity of disorders that it can exhibit. In this article, characteristics of disorders such as chronic, autoimmune, eosinophilic, hereditary, and infectious pancreatitis are described. With regard to autoimmune pancreatitis, the role of clinical evaluation, histologic examination, and IgG4 immunohistochemistry is discussed. The role of pancreatitis in the pathogenesis of diabetes is also mentioned. Some implications of pancreatitis are highlighted, including the neoplastic predisposition caused by inflammatory lesions of the pancreas. The goal of this article is to convey an appreciation of these disorders because their recognition can benefit patients tremendously, as inflammatory lesions of the pancreas can be mass-forming, giving rise to pseudotumors, and leading to surgical resection that may otherwise be unnecessary.
Collapse
Affiliation(s)
- Alton B Farris
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Emory University, 1364 Clifton Road Northeast, Room H-188, Atlanta, GA 30322, USA.
| | - Olca Basturk
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - N Volkan Adsay
- Department of Pathology and Laboratory Medicine, Emory University, 1364 Clifton Road NE, Room H-180B, Atlanta, GA 30322, USA
| |
Collapse
|
48
|
Abstract
OBJECTIVES In a hereditary pancreatitis family from Denmark, we identified a novel intragenic duplication of 9 nucleotides in exon-2 of the human cationic trypsinogen (PRSS1) gene (c.63_71dup) which at the amino-acid level resulted in the insertion of 3 amino acids within the activation peptide of cationic trypsinogen (p.K23_I24insIDK). The aim of the present study was to characterize the effect of this unique genetic alteration on the function of human cationic trypsinogen. METHODS Wild-type and mutant cationic trypsinogens were produced recombinantly and purified to homogeneity. Trypsinogen activation was followed by enzymatic assays and sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Trypsinogen secretion was measured from transfected HEK 293T cells. RESULTS Recombinant cationic trypsinogen carrying the p.K23_I24insIDK mutation exhibited greater than 10-fold increased autoactivation. Activation by human cathepsin B also was accelerated by 10-fold. Secretion of the p.K23_I24insIDK mutant from transfected cells was diminished, consistent with intracellular autoactivation. CONCLUSIONS This is the first report of an intragenic duplication within the PRSS1 gene causing hereditary pancreatitis. The accelerated activation of p.K23_I24insIDK by cathepsin B is a unique biochemical property not found in any other pancreatitis-associated trypsinogen mutant. In contrast, the robust autoactivation of the novel mutant confirms the notion that increased autoactivation is a disease-relevant mechanism in hereditary pancreatitis.
Collapse
|
49
|
Abstract
There is an unacceptably high mortality in acute pancreatitis, which is due to the lack of specific treatments for the disease. A major reason stated to account for the inability to develop effective treatments is that there are multiple pathobiologic pathways activated in the acinar cell mediating pancreatitis making it difficult to choose molecular targets for therapeutic strategies. However, this reasoning limits opportunities for therapeutic development because it does include another important participant in pancreatitis - the pancreatic duct cells. The most recent advance in pancreatitis research is that depletion of both glycolytic and oxidative ATP synthesis is a common event in both acinar and ductal cells. Although ATP has a very short half-life in the blood and is hydrolysed to ADP, there is clear evidence that encapsulating ATP into liposomes can effectively drive ATP into the cells which can be effective in protecting them from necrosis. In this review, we will examine the effects of different insults associated with pancreatitis on both the acinar and ductal components of the exocrine pancreas pointing out the role of the ductal epithelial responses in both attenuating and increasing the severity of pancreatitis. In addition, we propose that exogenous ATP administration may restore ductal and acinar function providing therapeutic benefit.
Collapse
Affiliation(s)
- Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged, Hungary.
| | - Stephen Pandol
- Department of Medicine, Veterans Affairs and University of California, Los Angeles, California, USA
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Zoltán Rakonczay
- First Department of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
50
|
Abstract
Chronic pancreatitis (CP) is a disease characterized by irreversible destruction and fibrosis of the parenchyma, leading to pancreatic exocrine insufficiency. In developed countries, the etiology for 60% to 70% of CP amongst male patients is alcohol and 25% are classified as idiopathic chronic pancreatitis (ICP). The genetic predisposition to CP could be an inappropriate activation of trypsinogen in the pancreas. Two common haplotypes, c.101A>G (p.N34S) and c.-215G>A, and four intronic alterations of the serine protease inhibitor Kazal type 1 (SPINK1) gene have been found to increase the risk for CP in the Asia Pacific region. Hence, SPINK1 is thought to be a candidate gene for pancreatitis. A loss-of-function alteration in chymotrypsinogen C (CTRC) gene has been shown to be associated with tropical calcific pancreatitis (TCP). Cathepsin B (CTSB) is also found to be associated with TCP. However mutations in cationic and anionic trypsinogen gene do not play an important role in causing CP in Asia Pacific region.
Collapse
Affiliation(s)
- D Nageshwar Reddy
- Asian Healthcare Foundation, Asian Institute of Gastroenterology, Somajiguda, Hyderabad, Andhra Pradesh, India.
| | | |
Collapse
|