1
|
Du ZK, Wang YC, Wang YH, Li XY, Zheng YZ, Wu D, Qu WM, Liao Z, Zou WB. Associations between ABO, FUT2 and chronic pancreatitis: A comprehensive meta-analysis of multiple cohorts and public biobanks. Pancreatology 2025; 25:58-64. [PMID: 39732592 DOI: 10.1016/j.pan.2024.12.016] [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: 11/02/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
OBJECTIVES Associations of ABO blood group specifying transferases A/B (ABO) and fucosyltransferase 2 (FUT2) with CP remain inconclusive. We aimed to comprehensively investigate the associations by Chinese sequencing cohorts and external cohorts. METHODS First, we analyzed the distributions of ABO blood groups and FUT2 status, along with lead single nucleotide polymorphisms (SNPs) at ABO (rs8176693 C/T) and FUT2 (rs632111 A/G) gene loci in Chinese low-coverage whole-genome sequencing discovery cohort. Subsequently, we investigated the associations of CP with ABO and FUT2 SNPs in Chinese whole-exome sequencing validation cohort and three public biobanks (FinnGen, UK Biobank, and BioBank Japan). Finally, comprehensive meta-analysis was performed by integrating data from two Chinese cohorts, reported cohorts and public biobanks. RESULTS Firstly, in Chinese discovery cohort, the distribution of blood types in CP patients showed no significant difference compared to healthy controls, and similar results were observed in subgroup analyses and in meta-analysis with the reported cohorts. Secondly, results indicated no association between rs8176693 or rs632111 and CP in Chinese cohorts and meta-analysis of three biobanks, though ABO SNP was found to be significantly associated with CP in UK Biobank (odds ratio [OR] = 1.27, P = 0.04). Finally, no association was observed between rs8176693 (OR = 1.03, P = 0.29) or rs632111 (OR = 1.04, P = 0.10) and CP in comprehensive meta-analysis. CONCLUSIONS No association was found between lead SNPs of ABO or FUT2 and CP in meta-analysis, nor was there an association between ABO blood group or FUT2 secretor status and CP in Chinese cohort. ABO and FUT2 might play limited role in CP development.
Collapse
Affiliation(s)
- Zeng-Kan Du
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuan-Chen Wang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ya-Hui Wang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiao-Yu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yi-Zhou Zheng
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Di Wu
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wei-Ming Qu
- Gastroenterology Ward, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, China
| | - Zhuan Liao
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Wen-Bin Zou
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
2
|
Shieh C, Dean RJ, Silva SA, Rodriguez L, Perez JM, Mendoza Ladd A. Association between acute pancreatitis severity and ABO/Rh blood group. Medicine (Baltimore) 2024; 103:e40789. [PMID: 39612377 PMCID: PMC11608716 DOI: 10.1097/md.0000000000040789] [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: 07/08/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
Acute pancreatitis (AP) is an inflammatory disorder associated with a significant mortality rate in its severe form. This study aimed to evaluate the association between severity of AP and ABO/Rh blood type. Retrospective chart review was conducted on hospitalized patients who met the diagnostic criteria for AP. Data collected included patient demographics, ABO/Rh blood type, etiology of pancreatitis, severity based on the Atlanta classification, and hospital length of stay. The proportion of patients who experienced severe AP was compared amongst combinations of ABO/Rh blood group. Of the 358 patients included in the study, 20.9% had non-mild AP. The proportion of patients in each blood group with non-mild AP was as follows: A: 21.1%, B: 21.4%, AB: 5.9%, O: 30.9%, Rh+: 22.0%, Rh-: 14.8%. When comparing across A, B, AB, O and Rh groups separately and in combination, there was no statistically significant correlation found between AP severity and ABO/Rh blood type. In this retrospective cohort study, no significant association between ABO/Rh blood group and severity of AP was found, suggesting that the inflammatory cascade in AP is not directly influenced by blood groups.
Collapse
Affiliation(s)
- Christine Shieh
- Division of Gastroenterology, University of California, Davis, Sacramento, CA
| | - Richard J. Dean
- Division of Gastroenterology, University of California, Davis, Sacramento, CA
| | - Spring A. Silva
- Division of Gastroenterology, University of California, Davis, Sacramento, CA
| | - Lizette Rodriguez
- Division of Internal Medicine, University of California, Davis, Sacramento, CA
| | - Jose Martinez Perez
- Division of Internal Medicine, University of California, Davis, Sacramento, CA
| | | |
Collapse
|
3
|
Zhernakova DV, Wang D, Liu L, Andreu-Sánchez S, Zhang Y, Ruiz-Moreno AJ, Peng H, Plomp N, Del Castillo-Izquierdo Á, Gacesa R, Lopera-Maya EA, Temba GS, Kullaya VI, van Leeuwen SS, Xavier RJ, de Mast Q, Joosten LAB, Riksen NP, Rutten JHW, Netea MG, Sanna S, Wijmenga C, Weersma RK, Zhernakova A, Harmsen HJM, Fu J. Host genetic regulation of human gut microbial structural variation. Nature 2024; 625:813-821. [PMID: 38172637 PMCID: PMC10808065 DOI: 10.1038/s41586-023-06893-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 11/23/2023] [Indexed: 01/05/2024]
Abstract
Although the impact of host genetics on gut microbial diversity and the abundance of specific taxa is well established1-6, little is known about how host genetics regulates the genetic diversity of gut microorganisms. Here we conducted a meta-analysis of associations between human genetic variation and gut microbial structural variation in 9,015 individuals from four Dutch cohorts. Strikingly, the presence rate of a structural variation segment in Faecalibacterium prausnitzii that harbours an N-acetylgalactosamine (GalNAc) utilization gene cluster is higher in individuals who secrete the type A oligosaccharide antigen terminating in GalNAc, a feature that is jointly determined by human ABO and FUT2 genotypes, and we could replicate this association in a Tanzanian cohort. In vitro experiments demonstrated that GalNAc can be used as the sole carbohydrate source for F. prausnitzii strains that carry the GalNAc-metabolizing pathway. Further in silico and in vitro studies demonstrated that other ABO-associated species can also utilize GalNAc, particularly Collinsella aerofaciens. The GalNAc utilization genes are also associated with the host's cardiometabolic health, particularly in individuals with mucosal A-antigen. Together, the findings of our study demonstrate that genetic associations across the human genome and bacterial metagenome can provide functional insights into the reciprocal host-microbiome relationship.
Collapse
Affiliation(s)
- Daria V Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Daoming Wang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Lei Liu
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Yue Zhang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Angel J Ruiz-Moreno
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Haoran Peng
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Niels Plomp
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Ángela Del Castillo-Izquierdo
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Esteban A Lopera-Maya
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Godfrey S Temba
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vesla I Kullaya
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Sander S van Leeuwen
- University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen, The Netherlands
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Serena Sanna
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- Institute for Genetic and Biomedical Research, National Research Council, Cagliari, Italy
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands.
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands.
| |
Collapse
|
4
|
Spagnolo DM, Greer PJ, Ohlsen CS, Mance S, Ellison M, Breze C, Busby B, Whitcomb DC, Haupt M. Acute and Chronic Pancreatitis Disease Prevalence, Classification, and Comorbidities: A Cohort Study of the UK BioBank. Clin Transl Gastroenterol 2022; 13:e00455. [PMID: 35060944 PMCID: PMC8806365 DOI: 10.14309/ctg.0000000000000455] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Pancreatitis is a complex syndrome that results from many etiologies. Large well-characterized cohorts are needed to further understand disease risk and prognosis. METHODS A pancreatitis cohort of more than 4,200 patients and 24,000 controls were identified in the UK BioBank (UKBB) consortium. A descriptive analysis was completed, comparing patients with acute (AP) and chronic pancreatitis (CP). The Toxic-metabolic, Idiopathic, Genetic, Autoimmune, Recurrent, and severe pancreatitis and Obstructive checklist Version 2 classification was applied to patients with AP and CP and compared with the control population. RESULTS CP prevalence in the UKBB is 163 per 100,000. AP incidence increased from 21.4/100,000 per year from 2001 to 2005 to 48.2/100,000 per year between 2016 and 2020. Gallstones and smoking were confirmed as key risk factors for AP and CP, respectively. Both populations carry multiple risk factors and a high burden of comorbidities, including benign and malignant neoplastic disorders. DISCUSSION The UKBB serves as a rich cohort to evaluate pancreatitis. Disease burden of AP and CP was high in this population. The association of common risk factors identified in other cohort studies was confirmed in this study. Further analysis is needed to link genomic risks and biomarkers with disease features in this population.
Collapse
Affiliation(s)
- Daniel M. Spagnolo
- Ariel Precision Medicine, Inc., Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Phil J. Greer
- Ariel Precision Medicine, Inc., Pittsburgh, Pennsylvania, USA
| | | | | | | | - Cameron Breze
- Ariel Precision Medicine, Inc., Pittsburgh, Pennsylvania, USA
| | - Ben Busby
- DNAnexus, Mountain View, California, USA
| | - David C. Whitcomb
- Ariel Precision Medicine, Inc., Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh, Pennsylvania, USA
| | - Mark Haupt
- Ariel Precision Medicine, Inc., Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Ng JW, Chong ETJ, Lee PC. An Updated Review on the Role of Single Nucleotide Polymorphisms in COVID-19 Disease Severity: A Global Aspect. Curr Pharm Biotechnol 2022; 23:1596-1611. [PMID: 35034591 DOI: 10.2174/1389201023666220114162347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/09/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and recently has become a serious global pandemic. Age, gender, and comorbidities are known to be common risk factors for severe COVID-19 but are not enough to fully explain the magnitude of their effect on the risk of severity of the disease. Single nucleotide polymorphisms (SNPs) in several genes have been reported as a genetic factor contributing to COVID-19 severity. This comprehensive review focuses on the association between SNPs in four important genes and COVID-19 severity in a global aspect. We discuss a total of 39 SNPs in this review: five SNPs in the ABO gene, nine SNPs in the angiotensin-converting enzyme 2 (ACE2) gene, 19 SNPs in the transmembrane protease serine 2 (TMPRSS2) gene, and six SNPs in the toll-like receptor 7 (TLR7) gene. These SNPs data could assist in monitoring an individual's risk of severe COVID-19 disease, and therefore personalized management and pharmaceutical treatment could be planned in COVID-19 patients.
Collapse
Affiliation(s)
- Jun Wei Ng
- Biotechnology Programme, Faculty of Science and Natural Resources, Universiti Malaysia, Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia
| | - Eric Tzyy Jiann Chong
- Biotechnology Programme, Faculty of Science and Natural Resources, Universiti Malaysia, Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia
| | - Ping-Chin Lee
- Biotechnology Programme, Faculty of Science and Natural Resources, Universiti Malaysia, Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia;
- Biotechnology Research Institute, Universiti Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia
| |
Collapse
|
6
|
Abstract
The Toxic-metabolic, Idiopathic, Genetic, Autoimmune, Recurrent and severe acute pancreatitis and Obstructive (TIGAR-O) Pancreatitis Risk/Etiology Checklist (TIGAR-O_V1) is a broad classification system that lists the major risk factors and etiologies of recurrent acute pancreatitis, chronic pancreatitis, and overlapping pancreatic disorders with or without genetic, immunologic, metabolic, nutritional, neurologic, metaplastic, or other features. New discoveries and progressive concepts since the 2001 TIGAR-O list relevant to understanding and managing complex pancreatic disorders require an update to TIGAR-O_V2 with both a short (S) and long (L) form. The revised system is designed as a hierarchical checklist for health care workers to quickly document and track specific factors that, alone or in combinations, may contribute to progressive pancreatic disease in individual patients or groups of patients and to assist in treatment selection. The rationale and key clinical considerations are summarized for each updated classification item. Familiarity with the structured format speeds up the completion process and supports thoroughness and consideration of complex or alternative diagnoses during evaluation and serves as a framework for communication. The structured approach also facilitates the new health information technologies that required high-quality data for accurate precision medicine. A use primer accompanies the TIGAR-O_V2 checklist with rationale and comments for health care workers and industries caring for patients with pancreatic diseases.
Collapse
|
7
|
Peng Y, Goyal H, Lin H, Liu D, Li L. Serum amylase activity altered by the ABO blood group system in Chinese subjects. J Clin Lab Anal 2019; 33:e22883. [PMID: 30938472 PMCID: PMC6595472 DOI: 10.1002/jcla.22883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE The underlying interactions between ABO blood group antigens and pancreatic exocrine tissue have been demonstrated, and serum amylase was synthesized by pancreatic ductal cells. Thus, we investigated the link between ABO blood type and serum amylase activity in Chinese subjects. METHODS Our study included 343 relatively healthy Chinese individuals, and the data were retrieved from electronic medical record database. RESULTS A increased trend was observed for serum amylase activity in ABO blood type distribution, and we found that serum amylase activity was remarkable increased in subjects with O blood type compared to those with non-O blood type (P = 0.013). Logistic regression analysis indicated that serum amylase was independently associated with individuals with O blood group (adjusted odds ratio 1.574; 95% CI, 1.022-2.425, P = 0.039). CONCLUSIONS The present evidence suggests a significant link between serum amylase activity and ABO blood type in the study population, indicating ABO blood type may be associated with the susceptibility of pancreatic disease.
Collapse
Affiliation(s)
- You‐Fan Peng
- Department of Endocrinology, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingChina
- Pancreatic Research InstituteSoutheast UniversityNanjingChina
| | - Hemant Goyal
- Department of Internal Medicine, School of MedicineMercer UniversityMaconGeorgiaUSA
| | - Hao Lin
- Pancreatic Research InstituteSoutheast UniversityNanjingChina
- Department of Clinical Science and Research, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingChina
| | - De‐Chen Liu
- Pancreatic Research InstituteSoutheast UniversityNanjingChina
- Department of Clinical Science and Research, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingChina
| | - Ling Li
- Department of Endocrinology, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingChina
- Pancreatic Research InstituteSoutheast UniversityNanjingChina
| |
Collapse
|
8
|
Mayerle J, Sendler M, Hegyi E, Beyer G, Lerch MM, Sahin-Tóth M. Genetics, Cell Biology, and Pathophysiology of Pancreatitis. Gastroenterology 2019; 156:1951-1968.e1. [PMID: 30660731 PMCID: PMC6903413 DOI: 10.1053/j.gastro.2018.11.081] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023]
Abstract
Since the discovery of the first trypsinogen mutation in families with hereditary pancreatitis, pancreatic genetics has made rapid progress. The identification of mutations in genes involved in the digestive protease-antiprotease pathway has lent additional support to the notion that pancreatitis is a disease of autodigestion. Clinical and experimental observations have provided compelling evidence that premature intrapancreatic activation of digestive proteases is critical in pancreatitis onset. However, disease course and severity are mostly governed by inflammatory cells that drive local and systemic immune responses. In this article, we review the genetics, cell biology, and immunology of pancreatitis with a focus on protease activation pathways and other early events.
Collapse
Affiliation(s)
- Julia Mayerle
- Medical Department II, University Hospital, LMU, Munich, Germany,Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Eszter Hegyi
- Institute for Translational Medicine, University of Pécs, Hungary
| | - Georg Beyer
- Medical Department II, University Hospital, LMU, Munich, Germany
| | - Markus M. Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Miklós Sahin-Tóth
- Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA 02118
| |
Collapse
|
9
|
El Jellas K, Johansson BB, Fjeld K, Antonopoulos A, Immervoll H, Choi MH, Hoem D, Lowe ME, Lombardo D, Njølstad PR, Dell A, Mas E, Haslam SM, Molven A. The mucinous domain of pancreatic carboxyl-ester lipase (CEL) contains core 1/core 2 O-glycans that can be modified by ABO blood group determinants. J Biol Chem 2018; 293:19476-19491. [PMID: 30315106 DOI: 10.1074/jbc.ra118.001934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Carboxyl-ester lipase (CEL) is a pancreatic fat-digesting enzyme associated with human disease. Rare mutations in the CEL gene cause a syndrome of pancreatic exocrine and endocrine dysfunction denoted MODY8, whereas a recombined CEL allele increases the risk for chronic pancreatitis. Moreover, CEL has been linked to pancreatic ductal adenocarcinoma (PDAC) through a postulated oncofetal CEL variant termed feto-acinar pancreatic protein (FAPP). The monoclonal antibody mAb16D10 was previously reported to detect a glycotope in the highly O-glycosylated, mucin-like C terminus of CEL/FAPP. We here assessed the expression of human CEL in malignant pancreatic lesions and cell lines. CEL was not detectably expressed in neoplastic cells, implying that FAPP is unlikely to be a glycoisoform of CEL in pancreatic cancer. Testing of the mAb16D10 antibody in glycan microarrays then demonstrated that it recognized structures containing terminal GalNAc-α1,3(Fuc-α1,2)Gal (blood group A antigen) and also repeated protein sequences containing GalNAc residues linked to Ser/Thr (Tn antigen), findings that were supported by immunostainings of human pancreatic tissue. To examine whether the CEL glycoprotein might be modified by blood group antigens, we used high-sensitivity MALDI-TOF MS to characterize the released O-glycan pool of CEL immunoprecipitated from human pancreatic juice. We found that the O-glycome of CEL consisted mainly of core 1/core 2 structures with a composition depending on the subject's FUT2 and ABO gene polymorphisms. Thus, among digestive enzymes secreted by the pancreas, CEL is a glycoprotein with some unique characteristics, supporting the view that it could serve additional biological functions to its cholesteryl esterase activity in the duodenum.
Collapse
Affiliation(s)
- Khadija El Jellas
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| | - Bente B Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Karianne Fjeld
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Center for Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Aristotelis Antonopoulos
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Heike Immervoll
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Ålesund Hospital, N-6017 Ålesund, Norway
| | - Man H Choi
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Mark E Lowe
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Dominique Lombardo
- INSERM, CRO2, Center for Research in Biological Oncology and Oncopharmacology, Aix-Marseille University, 13284 Marseille Cedex 07, France
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Anne Dell
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Eric Mas
- INSERM, CRO2, Center for Research in Biological Oncology and Oncopharmacology, Aix-Marseille University, 13284 Marseille Cedex 07, France
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Anders Molven
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway, .,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Genetic mutations in genes within and outside of the trypsin-dependent pathologic pathway have been found to be associated with chronic pancreatitis. This review highlights recent developments. RECENT FINDINGS CTRB1-CTRB2 has been identified as a new risk locus for chronic pancreatitis and the disease mechanism may involve trypsin degradation. Misfolding mutations in PRSS1, CPA1, and CEL, as well as environmental stress factors like tobacco and alcohol can trigger endoplasmic reticulum stress (ER-Stress). SUMMARY Protein misfolding as well as enzyme activity changes due to altered autoactivation, intracellular degradation, or enzyme inhibition represent the most important pathological mechanisms of chronic pancreatitis to date. Analysis of composite risk patterns by next-generation sequencing will help elucidate complex gene interactions and identify new potential therapeutic targets.
Collapse
|
11
|
Streicher SA, Klein AP, Olson SH, Amundadottir LT, DeWan AT, Zhao H, Risch HA. Impact of Sixteen Established Pancreatic Cancer Susceptibility Loci in American Jews. Cancer Epidemiol Biomarkers Prev 2017; 26:1540-1548. [PMID: 28754795 PMCID: PMC5626623 DOI: 10.1158/1055-9965.epi-17-0262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/19/2017] [Accepted: 07/20/2017] [Indexed: 01/04/2023] Open
Abstract
Background: The higher risk of pancreatic cancer in Ashkenazi Jews compared with non-Jews is only partially explained by the increased frequency of BRCA1 and BRCA2 mutations in Ashkenazi Jews.Methods: We evaluated the impact of 16 established pancreatic cancer susceptibility loci in a case-control sample of American Jews, largely Ashkenazi, including 406 full-Jewish pancreatic cancer patients and 2,332 full-Jewish controls, genotyped as part of the Pancreatic Cancer Cohort and Case-Control Consortium I/II (PanScan I/II), Pancreatic Cancer Case-Control Consortium (PanC4), and Resource for Genetic Epidemiology Research on Adult Health and Aging (GERA) datasets. We compared risk in full-Jewish subjects with risk in part-Jewish; non-Jewish Southern European; and in the combined non-Jewish Eastern, Northern, Southern, and Western European (non-Jewish white European) subjects from the same datasets. Jewish ancestries were genetically identified using seeded Fast principal component analysis. Data were analyzed by unconditional logistic regression, and adjusted for age, sex, and principal components.Results: One SNP on chromosome 13q22.1 (rs9543325; OR, 1.36; 95% confidence interval, 1.16-1.58; P = 10-4.1) was significant in full-Jews. Individual ORs and minor allele frequencies were similar between Jewish and non-Jewish white European subjects. The average ORs across the 16 pancreatic cancer susceptibility loci for full-Jewish, full- plus part-Jewish, non-Jewish Southern European, and non-Jewish white European subjects were 1.25, 1.30, 1.31, and 1.26, respectively.Conclusions: The 16 pancreatic cancer susceptibility loci similarly impact Jewish and non-Jewish white European subjects, both individually and as summary odds.Impact: These 16 pancreatic cancer susceptibility loci likely do not explain the higher risk seen in Ashkenazi Jews. Cancer Epidemiol Biomarkers Prev; 26(10); 1540-8. ©2017 AACR.
Collapse
Affiliation(s)
- Samantha A Streicher
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut.
| |
Collapse
|
12
|
Abstract
Chronic pancreatitis is defined as a pathological fibro-inflammatory syndrome of the pancreas in individuals with genetic, environmental and/or other risk factors who develop persistent pathological responses to parenchymal injury or stress. Potential causes can include toxic factors (such as alcohol or smoking), metabolic abnormalities, idiopathic mechanisms, genetics, autoimmune responses and obstructive mechanisms. The pathophysiology of chronic pancreatitis is fairly complex and includes acinar cell injury, acinar stress responses, duct dysfunction, persistent or altered inflammation, and/or neuro-immune crosstalk, but these mechanisms are not completely understood. Chronic pancreatitis is characterized by ongoing inflammation of the pancreas that results in progressive loss of the endocrine and exocrine compartment owing to atrophy and/or replacement with fibrotic tissue. Functional consequences include recurrent or constant abdominal pain, diabetes mellitus (endocrine insufficiency) and maldigestion (exocrine insufficiency). Diagnosing early-stage chronic pancreatitis is challenging as changes are subtle, ill-defined and overlap those of other disorders. Later stages are characterized by variable fibrosis and calcification of the pancreatic parenchyma; dilatation, distortion and stricturing of the pancreatic ducts; pseudocysts; intrapancreatic bile duct stricturing; narrowing of the duodenum; and superior mesenteric, portal and/or splenic vein thrombosis. Treatment options comprise medical, radiological, endoscopic and surgical interventions, but evidence-based approaches are limited. This Primer highlights the major progress that has been made in understanding the pathophysiology, presentation, prevalence and management of chronic pancreatitis and its complications.
Collapse
|
13
|
Conwell DL, Banks PA, Sandhu BS, Sherman S, Al-Kaade S, Gardner TB, Anderson MA, Wilcox CM, Lewis MD, Muniraj T, Forsmark CE, Cote GA, Guda NM, Tian Y, Romagnuolo J, Wisniewski SR, Brand R, Gelrud A, Slivka A, Whitcomb DC, Yadav D. Validation of Demographics, Etiology, and Risk Factors for Chronic Pancreatitis in the USA: A Report of the North American Pancreas Study (NAPS) Group. Dig Dis Sci 2017; 62:2133-2140. [PMID: 28600657 PMCID: PMC6040886 DOI: 10.1007/s10620-017-4621-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/16/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND/OBJECTIVES Our aim was to validate recent epidemiologic trends and describe the distribution of TIGAR-O risk factors in chronic pancreatitis (CP) patients. METHODS The NAPS-2 Continuation and Validation (NAPS2-CV) study prospectively enrolled 521 CP patients from 13 US centers from 2008 to 2012. CP was defined by definitive changes in imaging, endoscopy, or histology. Data were analyzed after stratification by demographic factors, physician-defined etiology, participating center, and TIGAR-O risk factors. RESULTS Demographics and physician-defined etiology in the NAPS2-CV study were similar to the original NAPS2 study. Mean age was 53 years (IQR 43, 62) with 55% males and 87% white. Overall, alcohol was the single most common etiology (46%) followed by idiopathic etiology (24%). Alcohol etiology was significantly more common in males, middle-aged (35-65 years), and non-whites. Females and elderly (≥65 years) were more likely to have idiopathic etiology, while younger patients (<35 years) to have genetic etiology. Variability in etiology was noted by participating centers (e.g., alcohol etiology ranged from 27 to 67% among centers enrolling ≥25 patients). Smoking was the most commonly identified (59%) risk factor followed by alcohol (53%), idiopathic (30%), obstructive (19%), and hyperlipidemia (13%). The presence of multiple TIGAR-O risk factors was common, with 1, 2, ≥3 risk factors observed in 27.6, 47.6, and 23.6% of the cohort, respectively. CONCLUSION Our data validate the current epidemiologic trends in CP. Alcohol remains the most common physician-defined etiology, while smoking was the most commonly identified TIGAR-O risk factor. Identification of multiple risk factors suggests CP to be a complex disease.
Collapse
Affiliation(s)
- Darwin L Conwell
- Brigham and Women's Hospital, Boston, MA, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | | | - Bimaljit S Sandhu
- Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Stuart Sherman
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Samer Al-Kaade
- Saint Louis University School of Medicine, St. Louis, MO, USA
| | | | | | - C Mel Wilcox
- University of Alabama at Birmingham Hospital, Birmingham, AL, USA
| | | | | | | | - Gregory A Cote
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nalini M Guda
- University of Wisconsin School of Medicine, Milwaukee, WI, USA
| | - Ye Tian
- Department of Epidemiology, University of Pittsburg Graduate School of Public Health, Pittsburgh, PA, USA
| | | | - Stephen R Wisniewski
- Department of Epidemiology, University of Pittsburg Graduate School of Public Health, Pittsburgh, PA, USA
| | - Randall Brand
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Andres Gelrud
- University of Chicago School of Medicine, Chicago, IL, USA
| | - Adam Slivka
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Dhiraj Yadav
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Pădureanu V, Enescu AŞ, Siloşi I, Forţofoiu M, Enescu A, Bogdan M, Forţofoiu MC, Dumitrescu AG, Tudoraşcu DR, Mita A, Streata I, Ioana M, Petrescu F, Săftoiu A. The association between chronic pancreatitis and the iNOS-2087A>G polymorphism. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2017; 55:89-95. [PMID: 28125406 DOI: 10.1515/rjim-2017-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Chronic pancreatitis is morphologically characterized by ductal dysplasia, breeding grounds for the proliferation of the ductal cells, the degenerative changes in pancreatic acinar cells and fibrosis, and it is defined on the basis of the clinical, morphological and functional criteria. AIM The aim of our study is to examine the existence of a possible correlation between the iNOS-2087A>G polymorphism and chronic pancreatitis by means of the genetic analysis. MATERIAL AND METHOD We have conducted the study at the Gastroenterology Clinic and the Research Center of Gastroenterology and Hepatology of the University of Medicine and Pharmacy, Craiova, between March 2015 - September 2016. The study had a prospective character. Both for the 58 patients diagnosed with chronic pancreatitis and for the 132 patients in the witness group, the biological material was represented by blood, (around 2.5 - 5 milliliters of venous blood) let on EDTA and kept at 4°C up to the separation of the DNA molecule. All the patients were genotyped for the iNOS - 2087A>G polymorphism, by means of the Real Time PCR technique with TaqMan probes. RESULTS Analysing the prevalence of the iNOS genotypes within the study group and witness group, we have noticed that, statistically speaking, there are no significant differences between the two groups. CONCLUSION As a conclusion, in the study lot we can sustain that the risk of developing chronic pancreatitis is not increased by the presence of the iNOS-2087A>G polymorphism.
Collapse
Affiliation(s)
- Vlad Pădureanu
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | | | - Isabela Siloşi
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Maria Forţofoiu
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Aurelia Enescu
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Maria Bogdan
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | | | | | | | - Adrian Mita
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ioana Streata
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Mihai Ioana
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Florin Petrescu
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Adrian Săftoiu
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| |
Collapse
|
15
|
Abstract
Despite decades of scientific and clinical research, pancreatic ductal adenocarcinoma (PDAC) remains a lethal malignancy. The clinical and pathologic features of PDAC, specifically the known environmental and genetic risk factors, are reviewed here with special emphasis on the hereditary pancreatic cancer (HPC) syndromes. For these latter conditions, strategies are described for their identification, for primary and secondary prevention in unaffected carriers, and for disease management in affected carriers. Nascent steps have been made toward personalized medicine based on the rational use of screening, tumor subtyping, and targeted therapies; these have been guided by growing knowledge of HPC syndromes in PDAC.
Collapse
Affiliation(s)
- Ashton A Connor
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Gallinger
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
16
|
Weiss FU, Schurmann C, Guenther A, Ernst F, Teumer A, Mayerle J, Simon P, Völzke H, Radke D, Greinacher A, Kuehn JP, Zenker M, Völker U, Homuth G, Lerch MM. Fucosyltransferase 2 (FUT2) non-secretor status and blood group B are associated with elevated serum lipase activity in asymptomatic subjects, and an increased risk for chronic pancreatitis: a genetic association study. Gut 2015; 64:646-56. [PMID: 25028398 DOI: 10.1136/gutjnl-2014-306930] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Serum lipase activities above the threefold upper reference limit indicate acute pancreatitis. We investigated whether high lipase activity-within the reference range and in the absence of pancreatitis-are associated with genetic single nucleotide polymorphisms (SNP), and whether these identified SNPs are also associated with clinical pancreatitis. METHODS Genome-wide association studies (GWAS) on phenotypes 'serum lipase activity' and 'high serum lipase activity' were conducted including 3966 German volunteers from the population-based Study-of-Health-in-Pomerania (SHIP). Lead SNPs associated on a genome-wide significance level were replicated in two cohorts, 1444 blood donors and 1042 pancreatitis patients. RESULTS Initial discovery GWAS detected SNPs within or near genes encoding the ABO blood group specifying transferases A/B (ABO), Fucosyltransferase-2 (FUT2), and Chymotrypsinogen-B2 (CTRB2), to be significantly associated with lipase activity levels in asymptomatic subjects. Replication analyses in blood donors confirmed the association of FUT-2 non-secretor status (OR=1.49; p=0.012) and ABO blood-type-B (OR=2.48; p=7.29×10(-8)) with high lipase activity levels. In pancreatitis patients, significant associations were found for FUT-2 non-secretor status (OR=1.53; p=8.56×10(-4)) and ABO-B (OR=1.69, p=1.0×10(-4)) with chronic pancreatitis, but not with acute pancreatitis. Conversely, carriers of blood group O were less frequently affected by chronic pancreatitis (OR=0.62; p=1.22×10(-05)) and less likely to have high lipase activity levels (OR=0.59; p=8.14×10(-05)). CONCLUSIONS These are the first results indicating that ABO blood type-B as well as FUT2 non-secretor status are common population-wide risk factors for developing chronic pancreatitis. They also imply that, even within the reference range, elevated lipase activities may indicate subclinical pancreatic injury in asymptomatic subjects.
Collapse
Affiliation(s)
- Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Claudia Schurmann
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany The Charles Bronfman Institute for Personalized Medicine, Genetics of Obesity & Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Annett Guenther
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Florian Ernst
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Teumer
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Peter Simon
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Dörte Radke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Department of Transfusion Medicine, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Jens-Peter Kuehn
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Martin Zenker
- Institute of Human Genetics, Otto-von-Guericke-Universität Magdeburg, University Hospital Magdeburg, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
17
|
Muniraj T, Aslanian HR, Farrell J, Jamidar PA. Chronic pancreatitis, a comprehensive review and update. Part I: epidemiology, etiology, risk factors, genetics, pathophysiology, and clinical features. Dis Mon 2015; 60:530-50. [PMID: 25510320 DOI: 10.1016/j.disamonth.2014.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
Whitcomb DC. Innovation and hard work: The 2015 George E. Palade Medal Award Lecture. Pancreatology 2015; 15:611-5. [PMID: 26481054 PMCID: PMC4688455 DOI: 10.1016/j.pan.2015.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/04/2015] [Indexed: 12/11/2022]
Abstract
The George E Palade Medal is the highest honor awarded by the International Association of Pancreatology and is given to an individual who has made outstanding contributions to the understanding of the pancreas and pancreatic diseases. Professor David C Whitcomb, University of Pittsburgh, is the 2015 recipient. The Palade Lecture, presented in Shanghai, China on August 28, 2015, included 5 personal stories that provide perspective from a life-time of achievement: "My life in a nutshell"; "Two old ladies"; "7777"; "I helped put a man on the moon"; and "Rugby". Together, the stories provide encouragement to a younger generation, struggling to find their way as physicians and scientists who are working on pancreatic or other diseases in a challenging world.
Collapse
Affiliation(s)
- David C. Whitcomb
- University of Pittsburgh, Gastroenterology, Room 401.4, 3708 Fifth Ave, Pittsburgh, PA 15213, USA. Tel.: +1 412 578 9515; fax: +1 412 578 9537.
| |
Collapse
|
19
|
Becker AE, Hernandez YG, Frucht H, Lucas AL. Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection. World J Gastroenterol 2014; 20:11182-11198. [PMID: 25170203 PMCID: PMC4145757 DOI: 10.3748/wjg.v20.i32.11182] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/15/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer-related deaths in the United States, with over 38000 deaths in 2013. The opportunity to detect pancreatic cancer while it is still curable is dependent on our ability to identify and screen high-risk populations before their symptoms arise. Risk factors for developing pancreatic cancer include multiple genetic syndromes as well as modifiable risk factors. Genetic conditions include hereditary breast and ovarian cancer syndrome, Lynch Syndrome, familial adenomatous polyposis, Peutz-Jeghers Syndrome, familial atypical multiple mole melanoma syndrome, hereditary pancreatitis, cystic fibrosis, and ataxia-telangiectasia; having a genetic predisposition can raise the risk of developing pancreatic cancer up to 132-fold over the general population. Modifiable risk factors, which include tobacco exposure, alcohol use, chronic pancreatitis, diet, obesity, diabetes mellitus, as well as certain abdominal surgeries and infections, have also been shown to increase the risk of pancreatic cancer development. Several large-volume centers have initiated such screening protocols, and consensus-based guidelines for screening high-risk groups have recently been published. The focus of this review will be both the genetic and modifiable risk factors implicated in pancreatic cancer, as well as a review of screening strategies and their diagnostic yields.
Collapse
|
20
|
Abstract
BACKGROUND There are few data on blood group (BG) types and types of pancreatic cancers. The aims of this study were to study BG types and BG-antigens in pancreatic intraductal papillary mucinous neoplasms (IPMNs). METHODS BG type and tumor BG-antigen (glycoprotein) expression (studied by immunohistochemistry on tissue microarrays) were analyzed with regard to characteristics of 101 surgically resected pancreatic IPMNs. RESULTS Non-O BG type predicted invasive carcinoma independently from high serum CA19-9 and male gender. BG type A was observed more frequently in women than in men. Chronic pancreatitis was more frequently seen in patients with BG type B or AB. Aberrant tumor expression (with regard to BG type) of loss of A antigen expression type occurred in 15.0% of IPMNs and of loss of B antigen expression type in 62.5% of IPMNs. Intraneoplasm BG-antigen expression was not related to dysplasia grade or invasion. CONCLUSION The results of the study suggest that in pancreatic IPMN, non-O BG type predicted invasive carcinoma, whereas for intratumor BG-antigen expression no specific patterns were detected with regard to the progression of glandular epithelial dysplasia or invasion.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Acute pancreatitis, recurrent acute pancreatitis (RAP) and chronic pancreatitis are interrelated and progressive inflammatory disorders of the pancreas with highly variable and complex susceptibility, severity and outcomes. The role of genetics in acute pancreatitis, RAP and progression to chronic pancreatitis within a new framework is needed. RECENT FINDINGS The first genome-wide association study in the pancreas has been published with genome-wide significance linked with noncoding variants at the PRSS1-PRSS2 locus on chromosome seven and the CLDN2 locus on the X chromosome. A new personalized medicine paradigm is being considered to facilitate organization of genetic and other susceptibility risk compared with the risk of disease progression or resolution and risk of complications. SUMMARY A new framework for organizing multiple, complex data sets is emerging. The role of genetics in the context of other variables is important in understanding susceptibility to RAP and in the modification of disease severity and progression to chronic pancreatitis. Questions of when to order testing, what to order and how to use the data in real time remains an area for future research and development.
Collapse
|
22
|
Abstract
OBJECTIVE Individuals with chronic pancreatitis are at increased risk for pancreatic cancer. We hypothesized that genetic variation in the γ-glutamyltransferase 1 (GGT1) gene, which was recently reported associated with pancreatic cancer risk in a genome-wide association study, is also associated with risk of chronic pancreatitis. METHODS Associations between common polymorphisms in GGT1 and chronic pancreatitis were evaluated using data and samples from the North American Pancreatitis Study 2. Patients (n = 496) and control subjects (n = 465) were genotyped for 4 single-nucleotide polymorphisms: rs4820599, rs2017869, rs8135987, and rs5751901. Odds ratios (ORs) and corresponding 95% confidence intervals (95% CI) for chronic pancreatitis risk were calculated using multiple logistic regression models. Interactions with cigarette smoking and alcohol use were explored. RESULTS Single-nucleotide polymorphisms rs8135987 and rs4820599 were both statistically significantly associated with risk of chronic pancreatitis; compared with common allele homozygotes, individuals with at least 1 minor allele were at increased risk (rs8135987: OR, 1.36; 95% CI, 1.03-1.80 [P(trend) = 0.01]; rs4820599: OR, 1.39; 95% CI, 1.04-1.84 [P(trend) = 0.0]; adjusted for age, sex, race, smoking status, and alcohol use). No significant interactions with cigarette smoking and alcohol use were observed. CONCLUSION Our results suggest that common variation in the GGT1 gene may also affect risk of chronic pancreatitis.
Collapse
|
23
|
Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 2013; 144:1252-61. [PMID: 23622135 PMCID: PMC3662544 DOI: 10.1053/j.gastro.2013.01.068] [Citation(s) in RCA: 1331] [Impact Index Per Article: 110.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/25/2013] [Accepted: 01/30/2013] [Indexed: 02/07/2023]
Abstract
Acute pancreatitis is one of the most frequent gastrointestinal causes of hospital admission in the United States. Chronic pancreatitis, although lower in incidence, significantly reduces patients' quality of life. Pancreatic cancer is associated with a high mortality rate and is one of the top 5 causes of death from cancer. The burden of pancreatic disorders is expected to increase over time. The risk and etiology of pancreatitis differ with age and sex, and all pancreatic disorders affect the black population more than any other race. Gallstones are the most common cause of acute pancreatitis, and early cholecystectomy eliminates the risk of future attacks. Alcohol continues to be the single most important risk factor for chronic pancreatitis. Smoking is an independent risk factor for acute and chronic pancreatitis, and its effects could synergize with those of alcohol. Significant risk factors for pancreatic cancer include smoking and non-O blood groups. Alcohol abstinence and smoking cessation can alter the progression of pancreatitis and reduce recurrence; smoking cessation is the most effective strategy to reduce the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Dhiraj Yadav
- Division of Gastroenterology & Hepatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, USA.
| | | |
Collapse
|
24
|
Pelzer U, Klein F, Bahra M, Sinn M, Dörken B, Neuhaus P, Meyer O, Riess H. Blood group determinates incidence for pancreatic cancer in Germany. Front Physiol 2013; 4:118. [PMID: 23745115 PMCID: PMC3662880 DOI: 10.3389/fphys.2013.00118] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 05/07/2013] [Indexed: 12/14/2022] Open
Abstract
Background: Genetic risk factors for sporadic pancreatic cancer are largely unknown but actually under high exposure. Findings of correlations between the AB0 blood group system (Chromosome 9q34,1—q34,2) and the risk of pancreatic cancer (PC) in patients from Asia, America and south Europe have already been published. So far it is unclear, whether this correlation between blood group an PC incidence can be found in German patients as well. Methods: One hundred and sixty-six patients who underwent a resection of PC were evaluated in a period between 2000 and 2010. Blood group reference distribution for the German population is given as: 0: 41%; A: 43%; B: 11%; AB: 5%; Rhesus positive: 85%; Rhesus negative: 15%. Analyses were done using the non-parametric Chi2-test (p-value two sided; SPSS 19.0). Results: Median age was 62 (34–82) years. Gender: female 73/44%; male: 93/56%. Observed blood group proportions: 0: 43 (25.9%)/A: 94 (56.6%)/B: 16 (9.6%)/AB: 13 (7.8%)/Rhesus positive: 131 (78.9%)/negative: 35 (21.1%). We detected a significant difference to the German reference distribution of the AB0 system (Chi2 19.34, df 3, p < 0.001). Rhesus factor has no impact on AB0-distribution (Chi2 4.13, df 3, p = 0.25), but differs significantly from reference distribution—probably due to initial AB0-variation (Chi2 4.82, df 1, p = 0.028). The odds ratio for blood group A is 2.01 and for blood group 0 is 0.5. Conclusions: The incidence of PC in the German cohort is highly associated with the AB0-system as well. More patients with blood group A suffer from PC (p < 0.001) whereas blood group 0 was less frequent in patients with PC (p < 0.001). Thus, our findings support the results from other non-German surveys. The causal trigger points of this carcinogenesis correlation are still not known.
Collapse
Affiliation(s)
- U Pelzer
- Department of Hematology/Oncology, Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Handra-Luca A. A and B blood group antigens in human pancreatic ductal adenocarcinomas. Oncology 2012; 84:141-9. [PMID: 23257765 DOI: 10.1159/000345028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/28/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The aim of this study was to determine the clinicopathological significance of blood group expression patterns in patients with surgically resected pancreatic ductal adenocarcinomas. METHODS Pancreatic ductal adenocarcinomas from 459 patients were analyzed for immunohistochemical expression of A and B blood group antigens on tissue microarrays. Correlations were established by Fisher's test, χ(2) test, and logistic regression models, and relationships to postsurgical overall survival were analyzed by Kaplan-Meier method, log-rank test, and multivariate Cox models. RESULTS Tumors expressing blood group A antigen predicted fewer lymph node metastases (<3) and had longer postoperative survival by univariate and multivariate analysis. Loss of A antigen tumor expression in A blood group type patients correlated to vascular invasion. CONCLUSION In patients with resectable pancreatic ductal adenocarcinomas, blood group A antigen expression predicted fewer lymph node metastases and was associated with improved outcome after pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Adriana Handra-Luca
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Md. 21231, USA
| |
Collapse
|