51
|
Andersson R, Löhr JM. Swedish national guidelines for chronic pancreatitis. Scand J Gastroenterol 2021; 56:469-483. [PMID: 33617407 DOI: 10.1080/00365521.2021.1881815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 02/04/2023]
Abstract
Chronic pancreatitis (CP) should be suspected in the case of recurrent upper abdominal pain of unknown origin and/or clinical signs of exocrine pancreatic insufficiency (EPI). Alcohol is the most common etiological factor associated with CP, others being smoking, male gender, and hereditary forms. CP is often associated with recurrent episodes of acute exacerbations.As of today, there is no accepted clinical definition of CP. However, irreversible morphological changes within the pancreas often occur, including dilatation of the main and branch pancreatic ducts, calcifications in ducts and parenchyma, parenchymal atrophy, and development of pseudocysts, though less so in the early phase of CP.
Collapse
|
52
|
Yamamiya A, Irisawa A, Tominaga K, Tsuchida K, Sugaya T, Tsunemi M, Hoshi K, Jinnai H, Yamabe A, Izawa N, Iwasaki M, Takimoto Y, Kanamori A, Nagashima K, Minaguchi T, Kashima K, Kunogi Y, Sato A, Goda K, Iijima M, Haruyama Y. Interobserver Reliability of the Endoscopic Ultrasound Criteria for the Diagnosis of Early Chronic Pancreatitis: Comparison between the 2009 and 2019 Japanese Diagnostic Criteria. Diagnostics (Basel) 2021; 11:431. [PMID: 33802623 PMCID: PMC8000630 DOI: 10.3390/diagnostics11030431] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
In 2009, diagnostic criteria for early chronic pancreatitis (DCECP2009) were proposed by the Japan Pancreas Society. This study aimed to evaluate the interobserver reliability (IOR) of endoscopic ultrasound (EUS) criteria for diagnosis of early chronic pancreatitis (ECP) between DCECP2009 and 2019 diagnostic criteria for ECP (DCECP2019) to assess the validity of the revision from the perspective of EUS findings. Among patients who underwent a detailed observation of the pancreas by EUS at our institution between January 2018 and March 2019, EUS images of 97 patients were extracted. Images were reviewed by 12 gastrointestinal endoscopy experts (eight pancreatologists, group A and four nonpancreatologists, group B). The overall kappa (K)-values for the IOR of the DCECP2009 EUS criteria were 0.424 and 0.563:0.231 for groups A:B, whereas the overall K-values for the DCECP2019 criteria were 0.618, and 0.733:0.442 for groups A:B. Regarding changes in the final diagnosis of ECP based on clinical features and EUS findings, 20 cases were definite ECP, 53 were probable ECP, and 24 were normal according to DCECP2009. In contrast, seven were definite ECP, 19 were probable ECP, and 71 were normal according to DECEP2019. IOR of DCECP2019 was higher than that of DCECP2009, which indicates an improvement in precision.
Collapse
Affiliation(s)
- Akira Yamamiya
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Atsushi Irisawa
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Keiichi Tominaga
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Kohei Tsuchida
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Takeshi Sugaya
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Misako Tsunemi
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Koki Hoshi
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Hidehito Jinnai
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Akane Yamabe
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Naoya Izawa
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Mari Iwasaki
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Yoichi Takimoto
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Akira Kanamori
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Kazunori Nagashima
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Takahito Minaguchi
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Ken Kashima
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Yasuhito Kunogi
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Ai Sato
- Department of Gastroenterology, Dokkyo Medical University Nikko Medical Center, 632 Takatoku Nikko, Tochigi 321-2593, Japan;
| | - Kenichi Goda
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Makoto Iijima
- Department of Gastroenterology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan; (A.Y.); (K.T.); (K.T.); (T.S.); (M.T.); (K.H.); (H.J.); (A.Y.); (N.I.); (M.I.); (Y.T.); (A.K.); (K.N.); (T.M.); (K.K.); (Y.K.); (K.G.); (M.I.)
| | - Yasuo Haruyama
- Department of Public Health, Dokkyo Medical University School of Medicine, 880 Kitakobayashi Mibu, Tochigi 321-0293, Japan;
| |
Collapse
|
53
|
Haber PS, Kortt NC. Alcohol use disorder and the gut. Addiction 2021; 116:658-667. [PMID: 32511812 DOI: 10.1111/add.15147] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/14/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Acute and chronic gastrointestinal problems are common in the setting of excessive alcohol use, and excessive alcohol use is associated with injury to all parts of the gastrointestinal tract. There is mounting evidence of gastrointestinal injury and increased cancer risk even from moderate alcohol consumption. The major causes of alcohol-related morbidity and mortality within the gastrointestinal system are liver disease, pancreatitis and gastrointestinal cancer. Other alcohol-related intestinal dysfunction is common but not life-threatening, leading to diarrhoea, malabsorption and nutritional deficiencies. This review describes non-neoplastic and neoplastic alcohol-related disorders of the gastrointestinal tract, omitting the liver, which has been reviewed elsewhere.
Collapse
Affiliation(s)
- Paul S Haber
- Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.,University of Sydney, Sydney, NSW, 2050, Australia
| | | |
Collapse
|
54
|
A high prevalence of genetic polymorphisms in idiopathic and alcohol-associated chronic pancreatitis patients in Ireland. HPB (Oxford) 2021; 23:231-237. [PMID: 32669225 DOI: 10.1016/j.hpb.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Individual genetic architecture is considered central to susceptibility and progression of disease in chronic pancreatitis. The study aimed to evaluate the presence of common pancreatic gene mutations in a defined cohort of idiopathic and alcohol-induced chronic pancreatitis patients in Ireland. METHODS The study comprised patients with idiopathic and alcohol-induced chronic pancreatitis and historic controls. Variants in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, cationic trypsinogen (PRSS1) gene and serine protease inhibitor kazal type-1 (SPINK1) gene, were assessed by Taqman© genotyping assay. RESULTS Of n = 126 patients and n = 167 controls, mutations were detected in 23 (20%) and in 10 (6%) respectively (P < 0.001). The majority of mutations found were in the SPINK1 gene variant N34S (13%) which increased disease risk almost six-fold (OR 5.9). Neither CFTR severe mutation (F508del) (P = 0.649) nor mild variant (R117H) (P = 0.327) were over-represented amongst patients compared to control subjects. PRSS1 variants were not detected in either patient or control subjects. CONCLUSION There was a significant prevalence of chronic pancreatitis-associated gene mutations in this well-phenotyped cohort. In patients with alcohol-related or idiopathic chronic pancreatitis, the possibility of genetic mutations in the SPINK 1 gene should be considered as a contributing aetiology factor.
Collapse
|
55
|
Alkhayyat M, Abou Saleh M, Coronado W, Abureesh M, Al-Otoom O, Qapaja T, Mansoor E, Simons-Linares CR, Stevens T, Chahal P. Increasing Prevalence of Anxiety and Depression Disorders After Diagnosis of Chronic Pancreatitis: A 5-Year Population-Based Study. Pancreas 2021; 50:153-159. [PMID: 33565791 DOI: 10.1097/mpa.0000000000001746] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Chronic pancreatitis (CP) is often associated with poor quality of life. Only a few small associative studies have reported the prevalence of mood disorders in CP. Using a large database, we sought to describe the epidemiology and risk association of anxiety and depression in CP and evaluate their outcomes. METHODS A multicenter database (Explorys), an aggregate of electronic health record data from 26 US healthcare systems, was surveyed. A cohort of patients with a diagnosis of CP between 2014 and 2019 was identified. Within this cohort, rates of anxiety and depression were calculated. Demographics, comorbidities, and outcomes were described. RESULTS Of the 30,276,810 individuals in the database (2014-2019), 67,260 patients had a CP diagnosis (0.22%). When compared with patients with no history of CP, patients with CP were more likely to develop anxiety (odds ratio, 6.94; 95% confidence interval, 6.85-7.04) and depression (odds ratio, 5.09; 95% confidence interval, 5.01-5.17). Chronic pancreatitis patients with depression had an increased risk of suicidal ideation compared with controls. CONCLUSIONS Patients with CP are at a higher risk of developing anxiety and depression compared with those without CP, with overall worse outcomes. Clinicians should screen CP patients and make appropriate referral to psychiatry when indicated.
Collapse
Affiliation(s)
| | - Mohannad Abou Saleh
- Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH
| | | | - Mohammad Abureesh
- Department of Internal Medicine, Staten Island University Hospital, New York City, NY
| | | | | | - Emad Mansoor
- Department of Gastroenterology and Hepatology, University Hospitals Cleveland Medical Center, Cleveland, OH
| | | | - Tyler Stevens
- Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH
| | - Prabhleen Chahal
- Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH
| |
Collapse
|
56
|
Gopalakrishnan G, Kalayarasan R, Gnanasekaran S, Pottakkat B. Frey's plus versus Frey's procedure for chronic pancreatitis: Analysis of postoperative outcomes and quality of life. Ann Hepatobiliary Pancreat Surg 2020; 24:496-502. [PMID: 33234753 PMCID: PMC7691205 DOI: 10.14701/ahbps.2020.24.4.496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 02/06/2023] Open
Abstract
Backgrounds/Aims Additional surgical procedures are often required in patients with chronic pancreatitis (CP) related complications. The present study aims to analyze the type of additional procedures required in patients who underwent Frey’s procedure (Frey’s plus) and to compare the short-term outcomes and quality of life with patients who underwent only Frey’s procedure. Methods Retrospective analysis of a prospectively maintained database of patients who underwent surgery for CP between January 2012 and February 2018 and completed at least one year of follow-up. Patients who underwent non-Frey’s surgical procedures were excluded. Clinical parameters, postoperative pain relief (using Izbicki pain score) and functioning scale score (EORTC QLQ C30) of patients who underwent Frey’s plus procedure and only Frey’s procedure were compared. Results Of the 146 patients who underwent surgery for CP during the study period, 100 patients (Frey’s procedure–68, Frey’s plus procedure–32) were included in this study. Roux-en-Y hepaticojejunostomy was the commonly performed additional procedure (n=12). The demographic and clinical parameters were comparable, except for more patients with jaundice (28.1% vs. 2.9%, p=0.01) and prolonged operative time (374.6 mins vs. 326.3 mins, p=0.01) in Frey’s plus group. However, there was no significant difference in mean intraoperative blood loss, postoperative morbidity or duration of hospital stay. At median (range) follow up of 34 (12-86) months, there was no significant difference in the pain control and quality of life between two groups. Conclusions Frey’s plus procedure for chronic pancreatitis can be safely performed wherever indicated without adversely affecting the postoperative outcome or quality of life.
Collapse
Affiliation(s)
| | - Raja Kalayarasan
- Department of Surgical Gastroenterology, JIPMER, Puducherry, India
| | | | - Biju Pottakkat
- Department of Surgical Gastroenterology, JIPMER, Puducherry, India
| |
Collapse
|
57
|
Pancreatic Diseases and Microbiota: A Literature Review and Future Perspectives. J Clin Med 2020; 9:jcm9113535. [PMID: 33139601 PMCID: PMC7692447 DOI: 10.3390/jcm9113535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota represent an interesting worldwide research area. Several studies confirm that microbiota has a key role in human diseases, both intestinal (such as inflammatory bowel disease, celiac disease, intestinal infectious diseases, irritable bowel syndrome) and extra intestinal disorders (such as autism, multiple sclerosis, rheumatologic diseases). Nowadays, it is possible to manipulate microbiota by administering prebiotics, probiotics or synbiotics, through fecal microbiota transplantation in selected cases. In this scenario, pancreatic disorders might be influenced by gut microbiota and this relationship could be an innovative and inspiring field of research. However, data are still scarce and controversial. Microbiota manipulation could represent an important therapeutic strategy in the pancreatic diseases, in addition to standard therapies. In this review, we analyze current knowledge about correlation between gut microbiota and pancreatic diseases, by discussing on the one hand existing data and on the other hand future possible perspectives.
Collapse
|
58
|
Yuan F, Hung RJ, Walsh N, Zhang H, Platz EA, Wheeler W, Song L, Arslan AA, Beane Freeman LE, Bracci P, Canzian F, Du M, Gallinger S, Giles GG, Goodman PJ, Kooperberg C, Le Marchand L, Neale RE, Rosendahl J, Scelo G, Shu XO, Visvanathan K, White E, Zheng W, Albanes D, Amiano P, Andreotti G, Babic A, Bamlet WR, Berndt SI, Brennan P, Bueno-de-Mesquita B, Buring JE, Campbell PT, Chanock SJ, Fuchs CS, Gaziano JM, Goggins MG, Hackert T, Hartge P, Hassan MM, Holly EA, Hoover RN, Katzke V, Kirsten H, Kurtz RC, Lee IM, Malats N, Milne RL, Murphy N, Ng K, Oberg AL, Porta M, Rabe KG, Real FX, Rothman N, Sesso HD, Silverman DT, Thompson IM, Wactawski-Wende J, Wang X, Wentzensen N, Wilkens LR, Yu H, Zeleniuch-Jacquotte A, Shi J, Duell EJ, Amundadottir LT, Li D, Petersen GM, Wolpin BM, Risch HA, Yu K, Klein AP, Stolzenberg-Solomon R. Genome-Wide Association Study Data Reveal Genetic Susceptibility to Chronic Inflammatory Intestinal Diseases and Pancreatic Ductal Adenocarcinoma Risk. Cancer Res 2020; 80:4004-4013. [PMID: 32641412 PMCID: PMC7861352 DOI: 10.1158/0008-5472.can-20-0447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/27/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022]
Abstract
Registry-based epidemiologic studies suggest associations between chronic inflammatory intestinal diseases and pancreatic ductal adenocarcinoma (PDAC). As genetic susceptibility contributes to a large proportion of chronic inflammatory intestinal diseases, we hypothesize that the genomic regions surrounding established genome-wide associated variants for these chronic inflammatory diseases are associated with PDAC. We examined the association between PDAC and genomic regions (±500 kb) surrounding established common susceptibility variants for ulcerative colitis, Crohn's disease, inflammatory bowel disease, celiac disease, chronic pancreatitis, and primary sclerosing cholangitis. We analyzed summary statistics from genome-wide association studies data for 8,384 cases and 11,955 controls of European descent from two large consortium studies using the summary data-based adaptive rank truncated product method to examine the overall association of combined genomic regions for each inflammatory disease group. Combined genomic susceptibility regions for ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis were associated with PDAC at P values < 0.05 (0.0040, 0.0057, 0.011, and 3.4 × 10-6, respectively). After excluding the 20 PDAC susceptibility regions (±500 kb) previously identified by GWAS, the genomic regions for ulcerative colitis, Crohn disease, and inflammatory bowel disease remained associated with PDAC (P = 0.0029, 0.0057, and 0.0098, respectively). Genomic regions for celiac disease (P = 0.22) and primary sclerosing cholangitis (P = 0.078) were not associated with PDAC. Our results support the hypothesis that genomic regions surrounding variants associated with inflammatory intestinal diseases, particularly, ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis are associated with PDAC. SIGNIFICANCE: The joint effects of common variants in genomic regions containing susceptibility loci for inflammatory bowel disease and chronic pancreatitis are associated with PDAC and may provide insights to understanding pancreatic cancer etiology.
Collapse
Affiliation(s)
- Fangcheng Yuan
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Ontario, Canada
| | - Naomi Walsh
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - William Wheeler
- Information Management Services, Inc., Silver Spring, Maryland
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Alan A Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, New York, USA
- Department of Population Health, New York University School of Medicine, New York, New York
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | | | - Paige Bracci
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Ontario, Canada
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Phyllis J Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Loic Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Rachel E Neale
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | | | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Emily White
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Pilar Amiano
- Public Health Division of Gipuzkoa, Ministry of Health of the Basque Government, Donostia-San Sebastian, Spain
- Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Julie E Buring
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peter T Campbell
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Smilow Cancer Hospital, New Haven, Connecticut
| | - J Michael Gaziano
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Boston Veteran Affairs Healthcare System, Boston, Massachusetts
| | - Michael G Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Manal M Hassan
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Holly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Robert N Hoover
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE-Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Robert C Kurtz
- Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - I-Min Lee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nuria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Miquel Porta
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital del Mar Institute of Medical Research (IMIM), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kari G Rabe
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Francisco X Real
- CIBERONC, Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Ian M Thompson
- CHRISTUS Santa Rosa Hospital - Medical Center, San Antonio, Texas
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, New York
| | - Xiaoliang Wang
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University School of Medicine, New York, New York
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Eric J Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology (ICO), Barcelona, Spain
| | | | - Donghui Li
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Alison P Klein
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- 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
| | | |
Collapse
|
59
|
Zhang T, Lu Y, Yang B, Zhang C, Li J, Liu H, Wang H, Wang D. Diffusion Metrics for Staging Pancreatic Fibrosis and Correlating With Epithelial‐Mesenchymal Transition Markers in a Chronic Pancreatitis Rat Model at 11.7T MRI. J Magn Reson Imaging 2020; 52:197-206. [DOI: 10.1002/jmri.26995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022] Open
Abstract
BackgroundChronic pancreatitis (CP) is characterized by pancreatic fibrosis, in which a epithelial‐mesenchymal transition (EMT)‐like process is observed. However, few noninvasive approaches have been reported to evaluate pancreatic fibrosis and EMT in an animal model based on diffusion imaging.PurposeTo evaluate pancreatic fibrosis in CP by conventional diffusion‐weighted imaging (DWI), intravoxel incoherent motion (IVIM), and diffusion kurtosis imaging (DKI) and then explore the correlation between diffusion parameters and the EMT markers in an animal model.Study TypeProspective controlled imaging histological correlation.PopulationForty‐five rats with CP induced by injecting dibutyltin dichloride solution and 10 normal rats comprised the control group.Field Strength/Sequence11.7T MR, diffusion imaging with 10 b‐values.AssessmentApparent diffusion coefficient (ADC), IVIM‐associated perfusion fraction (f), pseudodiffusion coefficient (D*), diffusion coefficient (D), DKI‐associated mean kurtosis (MK), and mean corrected diffusion coefficient (MD) were quantitatively measured and correlated with pancreatic fibrosis stages as well as the EMT markers E‐cadherin and α‐smooth muscle actin (α‐SMA) expression. The discriminative performance of diffusion parameters for staging fibrosis was compared.Statistical TestsSpearman's correlation, Student's t‐test, and a receiver operating characteristic curve was conducted for statistical analysis.ResultsADC, D, and MD (r = –0.637, –0.688, and –0.535; P < 0.001) were negatively correlated with pancreatic fibrosis staging, but MK (r = 0.740, P < 0.001) had a positive correlation. ADC, D, MD, and MK were significantly correlated with α‐SMA (r = –0.684, –0.728, –0.627, and 0.721, all P < 0.001), while MK was significantly correlated with E‐cadherin (r = –0.606, P < 0.001). The area under the curve (AUC) was not significantly different (P > 0.05) among ADC (0.797, 0.816, 0.873), D (0.862, 0.810, 0.895), MD (0.767, 0.772, 0.801), and MK (0.836, 0.893, 0.951) for F1 or greater, F2 or greater, and F3 pancreatic fibrosis separately.Data ConclusionADC, D, MD, and MK were helpful for assessing pancreatic fibrosis staging, and these diffusion parameters were also significantly correlated with the expression of EMT markers in pancreatic fibrosis.Level of Evidence: 2Technical Efficacy Stage: 2J. Magn. Reson. Imaging 2020;52:197–206.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yimei Lu
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Baofeng Yang
- Institute of Science and Technology for Brain‐Inspired Intelligence Fudan University Shanghai China
| | - Caiyuan Zhang
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jinning Li
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - He Wang
- Institute of Science and Technology for Brain‐Inspired Intelligence Fudan University Shanghai China
- Human Phenome Institute Fudan University Shanghai China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
60
|
Zhou D, Bai R, Wang L. The Cystic Fibrosis Transmembrane Conductance Regulator 470 Met Allele Is Associated with an Increased Risk of Chronic Pancreatitis in Both Asian and Caucasian Populations: A Meta-Analysis. Genet Test Mol Biomarkers 2020; 24:24-32. [PMID: 31940241 DOI: 10.1089/gtmb.2019.0199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: The Met470Val polymorphism (1540A>G [rs213950]) within the cystic fibrosis transmembrane conductance regulator (CFTR) protein has been reported to be associated with chronic pancreatitis (CP). The results remain inconclusive, and therefore, we performed this meta-analysis to clarify the association between M470V and CP risk. Methodology/Results: We conducted a meta-analysis of 7 case-control studies, including a total of 1121 CP patients and 2209 controls from Asian and Caucasian populations. We calculated the odds ratio (OR) and 95% confidence intervals (95% CI). Met470Val was found to be significantly associated with an increased risk of CP under all the genetic models (M vs. V, OR = 1.260, 95% CI: 1.134-1.399; MV vs. VV, OR = 1.292, 95% CI: 1.091-1.530; MM vs. VV, OR = 1.579, 95% CI: 1.274-1.956; MV/MV vs. VV, OR = 1.366, 95% CI: 1.165-1.603; MM vs. MV/VV, OR = 1.346, 95% CI: 1.114-1.621). Met470Val was also found to be significantly associated with an increased risk of idiopathic CP (ICP) in allele contrast, codominant, and recessive models (M vs. V, OR = 1.298, 95% CI: 1.020-1.653; MV vs. VV, OR = 1.297, 95% CI: 1.074-1.566; MM vs. VV, OR = 1.473, 95% CI: 1.165-1.862; MM vs. MV/VV, OR = 1.254, 95% CI: 1.023-1.538). Conclusions: The CFTR 470 M allele is significantly associated with an increased risk of CP in both Asian and Caucasian populations. The CFTR 470 M allele is also significantly associated with risk of ICP.
Collapse
Affiliation(s)
- Donger Zhou
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Rui Bai
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Liang Wang
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| |
Collapse
|
61
|
Magnetic resonance cholangiopancreatography findings in early chronic pancreatitis diagnosed according to the Japanese Diagnostic Criteria. Pancreatology 2020; 20:596-601. [PMID: 32371200 DOI: 10.1016/j.pan.2020.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/20/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES It is important for diagnosing early chronic pancreatitis (CP), which may be improved by therapeutic intervention. We aimed to examine the pancreatic ductal changes on magnetic resonance cholangiopancreatography (MRCP) in patients with early CP defined by the Japanese Diagnostic Criteria. METHODS This retrospective study included patients suspected early CP and performed both endoscopic ultrasonography (EUS) and MRCP from January 2010 to August 2018. We assessed the diameter of the main pancreatic duct (MPD) and the number of irregularly dilated duct branches using MRCP imaging in early CP. RESULTS We enrolled 165 patients and 25 patients (15%) fulfilled the diagnostic criteria for early CP. Irregular dilatation of ≥ 3 duct branches on MRCP was more often observed in early CP compared to non-early CP (P = 0.004), although MPD diameter was comparable (2.06 mm in early CP vs. 1.96 in non-early CP, P = 0.698). The sensitivity and specificity were 45% and 74%, respectively. The prevalence of positive MRCP findings in patients with ≥ 2 positive EUS findings was higher than that in patients with 1 positive EUS finding (P = 0.08) and in patients without an EUS finding (P < 0.001). There was no difference in the average diameter of MPD. CONCLUSION Patients with early CP often exhibit alteration in duct branches and not in MPD in addition to parenchymal alteration. Both pancreatic parenchyma and duct branches might need to be evaluated by EUS and MRCP.
Collapse
|
62
|
Jiang M, Li Z, Fu S, Xu Y, Tan Y, Jia W, Jiang Z, Mo N, Wei X, Zhang R, Zhang Z, Jiang G, Yang X. IVS8-5T Allele of CFTR is the Risk Factor in Chronic Pancreatitis, Especially in Idiopathic Chronic Pancreatitis. Am J Med Sci 2020; 360:55-63. [PMID: 32439152 DOI: 10.1016/j.amjms.2020.04.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/04/2020] [Accepted: 04/17/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Cystic fibrosis transmembrane conductance regulator IVS8-5T gene variation appears to be associated with a higher risk of chronic pancreatitis (CP); however, there is inconsistency between previous reported studies. Here, we performed a meta-analysis to investigate this relationship. MATERIALS AND METHODS PubMed and WANFANG databases were searched for the case-control studies that contained Patients with CP with IVS8-5T variation. Odd ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the relevance of IVS8-5T gene variation and CP. RESULTS Analysis showed that the frequency of the 5T allele was significantly higher in CP subjects than that in control subjects (OR = 1.43, 95% CI: 1.13-1.81, I2 = 1.2%). Based on the subgroup analysis stratified by etiology, the 5T allele was associated with a higher risk of idiopathic chronic pancreatitis (ICP) (OR = 1.80, 95% CI: 1.18-2.76, I2 = 0.0%) and not alcoholic CP (OR = 2.14, 95% CI: 0.98-4.66, I2 = 0.0%). Further study indicated that the 5T allele was related to higher ICP prevalence in the European population (OR = 1.79, 95% CI: 1.06-3.03, I2 = 0.0%). In contrast, there was no significant difference between ICP subjects and healthy controls within the Asian population (OR = 1.84, 95% CI: 0.91-3.72, I2 = 38.0%). CONCLUSIONS Cystic fibrosis transmembrane conductance regulator IVS8-5T is a risk factor in patients with CP. IVS8-5T variation may play a significant role in the occurrence of ICP, especially in the European population.
Collapse
Affiliation(s)
- Min Jiang
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhouquan Li
- Scientific Research Center, Guilin Medical University, Nanning, Guilin, China
| | - Shien Fu
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanzhen Xu
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanjun Tan
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Wenxian Jia
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhiwen Jiang
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Nanfang Mo
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Xinyan Wei
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Ruoheng Zhang
- Penn State College of Medicine, Hershey, Pennsylvania
| | - Zaiping Zhang
- Penn State College of Medicine, Hershey, Pennsylvania
| | - Guangjian Jiang
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, China..
| | - Xiaoli Yang
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China; Scientific Research Center, Guilin Medical University, Nanning, Guilin, China.
| |
Collapse
|
63
|
Ren YC, Zhao Q, He Y, Li B, Wu Z, Dai J, Wen L, Wang X, Hu G. Legumain promotes fibrogenesis in chronic pancreatitis via activation of transforming growth factor β1. J Mol Med (Berl) 2020; 98:863-874. [PMID: 32415356 DOI: 10.1007/s00109-020-01911-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 01/09/2023]
Abstract
Chronic pancreatitis (CP) is a major risk factor for pancreatic cancer; however, little is known about the pathogenic mechanisms underlying the development of CP. Legumain (Lgmn) has been linked to some chronic inflammatory diseases. The present study investigated the role of legumain in pancreatic fibrogenesis. We induced CP in wild type C57BL6 (WT), Lgmn-deficient (Lgmn-/-), Lgmnflox/flox and Lgmnflox/flox × LysMCre mice by intraperitoneal injection of caerulein for 4 weeks. Pancreata were collected and analyzed by quantitative reverse transcription polymerase chain reaction, Western blotting, and histology. Pancreatic stellate cells and macrophages were isolated and studied using immunofluorescence, gelatin zymography, and enzyme-linked immunosorbent assay. The effects of inhibition of legumain were investigated in vivo by administration of the specific legumain inhibitor, RR-11a. Legumain was found to be upregulated in the serum and pancreatic tissues of mice with caerulein-induced CP. Mice with global and macrophage-specific legumain deficiency exhibited significantly reduced development of pancreatic fibrosis compared with control mice, based on pancreas size, histology, and expression of fibrosis-associated genes. Our results indicate that legumain promotes activation of pancreatic stellate cells and increases synthesis of extracellular matrix proteins via activation of matrix metalloproteinase-2(MMP-2), which hydrolyzes the transforming growth factor-β1 (TGF-β1) precursor to form active TGF-β1. Administration of RR-11a markedly attenuated pancreatic fibrosis in mice with CP. Deficiency or inhibition of legumain significantly reduces the severity of pancreatic fibrosis by suppressing activation of the TGF-β1 precursor. Our results highlight the potential of legumain as a novel therapeutic target for CP. KEY MESSAGES: • Legumain expression was markedly upregulated in CP mice. • Deletion of legumain attenuated pancreatic fibrosis in CP mice. • Legumain promotes fibrosis via MMP-2 activation, which hydrolyzed the TGF-β1 precursor to the active form. • Legumain is a potential therapeutic target for the management of CP.
Collapse
Affiliation(s)
- Ying-Chun Ren
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qiuyan Zhao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan He
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Bin Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zengkai Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Juanjuan Dai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Li Wen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
64
|
Abstract
Multidisciplinary collaboration (MDC) has been widely adopted in healthcare to optimize patient care. MDC brings several specialized healthcare providers to the table using several methods, including multidisciplinary meetings (MDMs), multidisciplinary clinics, teleconferences, and online multidisciplinary expert panels, to reach the goal of achieving the best diagnosis and treatment plan for complex diseases. Diagnosis and management of acute/chronic pancreatitis is complex which necessitates the development and utilization of MDC. The key members of pancreatitis MDM include gastroenterologists, radiologists, pathologists, hepatobiliary surgeons, chairperson, and a coordinator. After selection of admitted or referred patients, the availability of required information is reviewed, and then each case is discussed. The final diagnosis and treatment plan is confirmed by consensus, especially for complex cases that require endoscopic intervention or pancreatectomy and patients with the possibility of pancreatic adenocarcinoma. It has been shown that MDMs have improved the clinical outcome of patients with acute/chronic pancreatitis. In addition to MDM, the feasibility of multidisciplinary clinics, teleconferences, and online multidisciplinary expert panels for the management of pancreatic disorders has been investigated. Understanding structure, potential advantages, and limitations of MDC will help clinicians and healthcare systems in developing an optimized MDC to improve the management of acute/chronic pancreatitis. This narrative review summarized prior recommendations and explored the impact of MDC on clinical outcomes of patients with pancreatitis. Our recommendations offer a generalizable method that can be utilized by healthcare systems.
Collapse
|
65
|
Dugic A, Nikolic S, Mühldorfer S, Bulajic M, Pozzi Mucelli R, Tsolakis AV, Löhr JM, Vujasinovic M. Clinical importance of main pancreatic duct variants and possible correlation with pancreatic diseases. Scand J Gastroenterol 2020; 55:517-527. [PMID: 32393143 DOI: 10.1080/00365521.2020.1760345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/04/2023]
Abstract
Background: Except for pancreas divisum (PD), the prevalence of anatomic variants of the main pancreatic duct (MPD) seems to be insufficiently investigated. To date, their role in the occurrence of pancreatic exocrine insufficiency (PEI) and morphological changes suggestive of chronic pancreatitis (CP) has remained unclear.Methods: A systematic review was performed, searching MEDLINE and Web of Science, limited to articles published between 1960 and 1 June 2019.Results: Our review included a total number of 3234 subjects. The most common variant of MPD was type 3, followed by type 1, indicating MPD drainage pattern into major papilla (MP) as the most frequent. A sub-variant of type 3, known as 'reverse pancreas divisum' had a prevalence of 2.2%. Type 4 variant- PD, was found in 6.4% of all cases. The most common sub-variant of PD was complete PD, followed by incomplete PD and variant with MPD as only pancreatic duct. Type 5 variant (including ansa pancreatica) was present in 2.9% of subjects. Apart from one study with a significantly higher frequency of morphological changes suggestive of CP in patients with ansa pancreatica, the studies stated no significant association between pancreatic disease and MPD variants. Furthermore, only one study examined the influence of MPD variants on exocrine pancreatic function. Although equivocal, this association is most likely found to be insignificant.Conclusion: To elucidate linkage between MPD variants and the occurrence of chronic pancreatitis and impairment of pancreatic exocrine function, further clinical investigations are warranted.
Collapse
Affiliation(s)
- Ana Dugic
- Department of Internal Medicine, Clinic for Gastroenterology, Endocrinology and Metabolic Disorders, Bayreuth, Germany
- Department of Medicine, Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Sara Nikolic
- Department of Medicine, Huddinge, Karolinska Institute, Stockholm, Sweden
- Department of Gastroenterology, Division of Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Steffen Mühldorfer
- Department of Internal Medicine, Clinic for Gastroenterology, Endocrinology and Metabolic Disorders, Bayreuth, Germany
| | - Milutin Bulajic
- Gastroenterology and Digestive Endoscopy Department, Mater Olbia Hospital, Olbia, Italy
| | - Raffaella Pozzi Mucelli
- Department of Abdominal Radiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Apostolos V Tsolakis
- Department for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna (MedS, K2), Division of Clinical Medicine, Karolinska Institute, Stockholm, Sweden
| | - J-Matthias Löhr
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
- Department for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Miroslav Vujasinovic
- Department of Medicine, Huddinge, Karolinska Institute, Stockholm, Sweden
- Department for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
66
|
Masamune A, Kotani H, Sörgel FL, Chen JM, Hamada S, Sakaguchi R, Masson E, Nakano E, Kakuta Y, Niihori T, Funayama R, Shirota M, Hirano T, Kawamoto T, Hosokoshi A, Kume K, Unger L, Ewers M, Laumen H, Bugert P, Mori MX, Tsvilovskyy V, Weißgerber P, Kriebs U, Fecher-Trost C, Freichel M, Diakopoulos KN, Berninger A, Lesina M, Ishii K, Itoi T, Ikeura T, Okazaki K, Kaune T, Rosendahl J, Nagasaki M, Uezono Y, Algül H, Nakayama K, Matsubara Y, Aoki Y, Férec C, Mori Y, Witt H, Shimosegawa T. Variants That Affect Function of Calcium Channel TRPV6 Are Associated With Early-Onset Chronic Pancreatitis. Gastroenterology 2020; 158:1626-1641.e8. [PMID: 31930989 DOI: 10.1053/j.gastro.2020.01.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/09/2019] [Accepted: 01/02/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Changes in pancreatic calcium levels affect secretion and might be involved in development of chronic pancreatitis (CP). We investigated the association of CP with the transient receptor potential cation channel subfamily V member 6 gene (TRPV6), which encodes a Ca2+-selective ion channel, in an international cohort of patients and in mice. METHODS We performed whole-exome DNA sequencing from a patient with idiopathic CP and from his parents, who did not have CP. We validated our findings by sequencing DNA from 300 patients with CP (not associated with alcohol consumption) and 1070 persons from the general population in Japan (control individuals). In replication studies, we sequenced DNA from patients with early-onset CP (20 years or younger) not associated with alcohol consumption from France (n = 470) and Germany (n = 410). We expressed TRPV6 variants in HEK293 cells and measured their activity using Ca2+ imaging assays. CP was induced by repeated injections of cerulein in TRPV6mut/mut mice. RESULTS We identified the variants c.629C>T (p.A210V) and c.970G>A (p.D324N) in TRPV6 in the index patient. Variants that affected function of the TRPV6 product were found in 13 of 300 patients (4.3%) and 1 of 1070 control individuals (0.1%) from Japan (odds ratio [OR], 48.4; 95% confidence interval [CI], 6.3-371.7; P = 2.4 × 10-8). Twelve of 124 patients (9.7%) with early-onset CP had such variants. In the replication set from Europe, 18 patients with CP (2.0%) carried variants that affected the function of the TRPV6 product compared with 0 control individuals (P = 6.2 × 10-8). Variants that did not affect the function of the TRPV6 product (p.I223T and p.D324N) were overrepresented in Japanese patients vs control individuals (OR, 10.9; 95% CI, 4.5-25.9; P = 7.4 × 10-9 for p.I223T and P = .01 for p.D324N), whereas the p.L299Q was overrepresented in European patients vs control individuals (OR, 3.0; 95% CI, 1.9-4.8; P = 1.2 × 10-5). TRPV6mut/mut mice given cerulein developed more severe pancreatitis than control mice, as shown by increased levels of pancreatic enzymes, histologic alterations, and pancreatic fibrosis. CONCLUSIONS We found that patients with early-onset CP not associated with alcohol consumption carry variants in TRPV6 that affect the function of its product, perhaps by altering Ca2+ balance in pancreatic cells. TRPV6 regulates Ca2+ homeostasis and pancreatic inflammation.
Collapse
Affiliation(s)
- Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Hiroshi Kotani
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Franziska Lena Sörgel
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Paediatric Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Jian-Min Chen
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France
| | - Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Emmanuelle Masson
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France; CHU Brest, Service de Génétique, Brest, France
| | - Eriko Nakano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoichi Kakuta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Funayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Matsuyuki Shirota
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuya Hirano
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Tetsuya Kawamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Atsuki Hosokoshi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kiyoshi Kume
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Lara Unger
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Paediatric Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Maren Ewers
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Paediatric Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Helmut Laumen
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Paediatric Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg-Hessen, Mannheim, Germany
| | - Masayuki X Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Volodymyr Tsvilovskyy
- Pharmakologisches Institut, Universität Heidelberg, Heidelberg, Germany; German Center for Cardiovascular Research, partner site Heidelberg, Germany
| | - Petra Weißgerber
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, Germany
| | - Ulrich Kriebs
- Pharmakologisches Institut, Universität Heidelberg, Heidelberg, Germany; German Center for Cardiovascular Research, partner site Heidelberg, Germany
| | - Claudia Fecher-Trost
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Universität Heidelberg, Heidelberg, Germany; German Center for Cardiovascular Research, partner site Heidelberg, Germany
| | - Kalliope N Diakopoulos
- Mildred Scheel Chair of Tumor Metabolism and Comprehensive Cancer Center Munich at the Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Alexandra Berninger
- Mildred Scheel Chair of Tumor Metabolism and Comprehensive Cancer Center Munich at the Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marina Lesina
- Mildred Scheel Chair of Tumor Metabolism and Comprehensive Cancer Center Munich at the Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kentaro Ishii
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Tsukasa Ikeura
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Kazuichi Okazaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Tom Kaune
- Department of Internal Medicine I, Martin Luther University, Halle (Saale), Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle (Saale), Germany
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yasuhito Uezono
- Cancer Pathophysiology Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Hana Algül
- Mildred Scheel Chair of Tumor Metabolism and Comprehensive Cancer Center Munich at the Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Keiko Nakayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Yoko Aoki
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Claude Férec
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France; CHU Brest, Service de Génétique, Brest, France
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Paediatric Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
67
|
Ruggeri JM, Franco-Barraza J, Sohail A, Zhang Y, Long D, Pasca di Magliano M, Cukierman E, Fridman R, Crawford HC. Discoidin Domain Receptor 1 (DDR1) Is Necessary for Tissue Homeostasis in Pancreatic Injury and Pathogenesis of Pancreatic Ductal Adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1735-1751. [PMID: 32339496 DOI: 10.1016/j.ajpath.2020.03.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 01/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) and chronic pancreatitis are characterized by a dense collagen-rich desmoplastic reaction. Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase activated by collagens that can regulate cell proliferation, migration, adhesion, and remodeling of the extracellular matrix. To address the role of DDR1 in PDA, Ddr1-null (Ddr-/-) mice were crossed with the KrasG12D/+; Trp53R172H/+; Ptf1aCre/+ (KPC) model of metastatic PDA. Ddr1-/-; KPC mice progress to differentiated PDA but resist progression to poorly differentiated cancer compared with KPC control mice. Strikingly, severe pancreatic atrophy accompanied tumor progression in Ddr1-/-; KPC mice. To further explore the effects of Ddr1 ablation, Ddr1-/- mice were crossed with the KrasG12D/+; Ptf1aCre/+ neoplasia model and subjected to cerulein-induced experimental pancreatitis. Similar to KPC mice, tissue atrophy was a hallmark of both neoplasia and pancreatitis models in the absence of Ddr1. Compared with controls, Ddr1-/- models had increased acinar cell dropout and reduced proliferation with no difference in apoptotic cell death between control and Ddr1-/- animals. In most models, organ atrophy was accompanied by increased fibrillar collagen deposition, suggesting a compensatory response in the absence of this collagen receptor. Overall, these data suggest that DDR1 regulates tissue homeostasis in the neoplastic and injured pancreas.
Collapse
Affiliation(s)
- Jeanine M Ruggeri
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Janusz Franco-Barraza
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Anjum Sohail
- Department of Pathology, Wayne State University, Detroit, Michigan; Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Daniel Long
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Edna Cukierman
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Rafael Fridman
- Department of Pathology, Wayne State University, Detroit, Michigan; Karmanos Cancer Institute, Wayne State University, Detroit, Michigan; Department of Oncology, Wayne State University, Detroit, Michigan.
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
68
|
Orekhova A, Geisz A, Sahin-Tóth M. Ethanol feeding accelerates pancreatitis progression in CPA1 N256K mutant mice. Am J Physiol Gastrointest Liver Physiol 2020; 318:G694-G704. [PMID: 32116022 PMCID: PMC7191466 DOI: 10.1152/ajpgi.00007.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alcoholic pancreatitis is a multifactorial, progressive, inflammatory disorder of the pancreas. Alcohol initiates pancreatitis and promotes its progression in the context of genetic susceptibility and/or other environmental risk factors such as smoking. Genetic mutations can cause digestive enzyme misfolding, which induces endoplasmic reticulum (ER) stress and elicits pancreatitis. Here, we tested the hypothesis that alcohol synergizes with misfolding in promoting ER stress and thereby accelerates chronic pancreatitis progression. To this end, we fed an ethanol-containing diet to CPA1 N256K mice, which carry the human p.N256K CPA1 mutation and develop spontaneous chronic pancreatitis. Inexplicably, CPA1 N256K mice suffered generalized seizures after 2-3 wk of ethanol feeding, which resulted in high mortality and the early termination of the study. Analysis of CPA1 N256K mice euthanized after 3-3.5 wk of ethanol feeding revealed more severe chronic pancreatitis associated with significantly increased Hspa5 [ER chaperone immunoglobulin heavy chain-binding protein (BiP)] mRNA levels when compared with CPA1 N256K mice on a control liquid diet. In contrast, ethanol feeding of C57BL/6N mice for 4 wk increased Hspa5 levels to a lesser degree and caused no pancreatitis. We conclude that ethanol feeding synergizes with the misfolding CPA1 mutant in promoting ER stress and thereby accelerates progression of chronic pancreatitis in CPA1 N256K mice.NEW & NOTEWORTHY Alcoholic pancreatitis is a multifactorial, progressive, inflammatory disorder of the pancreas. This study demonstrates that alcohol synergizes with digestive enzyme misfolding in promoting endoplasmic reticulum stress and thereby accelerates progression of chronic pancreatitis.
Collapse
Affiliation(s)
- Anna Orekhova
- 1Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University, Henry M. Goldman School of Dental Medicine, Boston, Massachusetts
| | - Andrea Geisz
- 1Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University, Henry M. Goldman School of Dental Medicine, Boston, Massachusetts
| | - Miklós Sahin-Tóth
- 1Center for Exocrine Disorders, Department of Molecular and Cell Biology, Boston University, Henry M. Goldman School of Dental Medicine, Boston, Massachusetts,2Department of Surgery, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
69
|
Lin CH, Yeh NC, Wang JJ, Ho CH, Her SH, Tsay WI, Chien CC. Effect of Chronic Pancreatitis on Complications and Mortality in DM Patients: A 10-year Nationwide Cohort Study. J Clin Endocrinol Metab 2020; 105:5715202. [PMID: 31974550 DOI: 10.1210/clinem/dgaa035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/23/2020] [Indexed: 02/08/2023]
Abstract
CONTEXT Chronic pancreatitis (CP), is a long-term inflammation of the pancreatic parenchyma, and might increase risk of a hyperglycemia crisis or hypoglycemia in patients with diabetes mellitus (DM); however, the relationship has not been previously investigated. OBJECTIVE To investigate the risk of diabetic ketoacidosis (DKA), hyperglycemic hyperosmolar state (HHS), hypoglycemia, and long-term outcomes in DM patients with CP. DESIGN A population-based cohort study. SETTING AND PARTICIPANTS Tapping Taiwan's National Health Insurance Research Database, we identified 506 DM patients with newly diagnosed CP from 1999 to 2010 and created a control cohort consisting of 5060 age- and sex-matched DM patients without CP from the same time period. We followed those 2 cohorts from the index date to occurrence of outcomes, the date of death or 31 December 2012. MAIN OUTCOME MEASURES DKA, HHS, hypoglycemia and mortality. RESULTS DM patients with CP, who were predominantly male (88%) and younger (60% < 45 years old), had a 9.5-, 5.0-, and 3.0-fold higher risk for DKA (95% confidence interval [CI]: 6.51-13.91), HHS (95% CI: 2.85-8.62), and hypoglycemia (95% CI: 2.23-4.08), respectively. They also had lower 1-, 5-, and 10-year cumulative survival rates (98.4% vs 99.0%, 87.7% vs 96.6%, and 78.7% vs 93.6%, respectively) (log-rank test: P < .001), and a 2.43-fold higher risk for death (HR: 2.43, 95% CI: 1.82-3.27). CONCLUSIONS In Taiwan, DM patients with CP have a higher incidence of DKA, HHS, hypoglycemia, and mortality. More attention is needed for preventing hyperglycemia crisis and hypoglycemia prevention in this population.
Collapse
Affiliation(s)
- Cheng-Heng Lin
- Department of Gastroenterology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Nai-Cheng Yeh
- Department of Endocrinology and Metabolism, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jhi-Joung Wang
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chung-Han Ho
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Shwu-Huey Her
- Division of Controlled Drugs, Food and Drug Administration, Ministry of Health and Welfare, Taiwan
| | - Wen-Ing Tsay
- Division of Controlled Drugs, Food and Drug Administration, Ministry of Health and Welfare, Taiwan
| | | |
Collapse
|
70
|
Xu X, Yu H, Sun L, Zheng C, Shan Y, Zhou Z, Wang C, Chen B. Adipose‑derived mesenchymal stem cells ameliorate dibutyltin dichloride‑induced chronic pancreatitis by inhibiting the PI3K/AKT/mTOR signaling pathway. Mol Med Rep 2020; 21:1833-1840. [PMID: 32319628 PMCID: PMC7057804 DOI: 10.3892/mmr.2020.10995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) play a positive role in tissue injury repair and regeneration. The aim of this study was to determine whether ASCs could ameliorate chronic pancreatitis (CP) induced by the injection of dibutyltin dichloride (DBTC) and to elucidate its potential mechanisms. Furthermore, this study also explored whether there was a significant difference if the ASCs were injected via the inferior vena cava or the left gastric artery. CP was induced in rats by a single intravenous administration of DBTC, and the accumulation of collagen and apoptotic rates of pancreatic acinar cells were analyzed. According to the results, ASCs markedly reduced DBTC-induced pancreatic damage and collagen deposition in the rat model of CP. Moreover, ASCs significantly decreased pancreatic cell apoptosis by regulating the expression levels of caspase-3, BAX and Bcl-2. These effects were observed regardless of whether the injection was in the inferior vena cava or the left gastric artery. It was also found that the expression levels of phosphorylated PI3K, AKT and mTOR in pancreatic tissues of the DBTC-induced CP model group were significantly increased, while the expression levels of phosphorylated PI3K, AKT and mTOR in the two treatment groups were markedly decreased. ASCs noticeably suppressed the PI3K/AKT/mTOR pathway in the pancreatic tissue of DBTC-induced CP. This study indicated that ASCs protect against pancreatic fibrosis by modulating the PI3K/AKT/mTOR pathway, and have the potential to be a new strategy for the treatment of CP in the future.
Collapse
Affiliation(s)
- Xiangxiang Xu
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Huajun Yu
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Linxiao Sun
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chenlei Zheng
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yunfeng Shan
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhenxu Zhou
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Cheng Wang
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
71
|
J. C. C, Parks RW. Chronic Pancreatitis—Update on Pathophysiology and Therapeutic Approaches. Indian J Surg 2020. [DOI: 10.1007/s12262-019-02059-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
AbstractChronic pancreatitis is an inflammatory condition characterized by structural change within the pancreas, that leads to progressive and irreversible loss of functioning pancreatic parenchyma, exocrine/endocrine dysfunction and an increased risk of pancreatic ductal adenocarcinoma. Whilst hallmarks of advanced disease are readily identifiable on routine clinical imaging, concordance between structural changes within the pancreas and symptoms is poor, such that early diagnosis can be challenging. Traditionally, chronic pancreatitis has been managed with a ‘step-up’ approach of measures including analgesia, therapeutic endoscopy and surgery (in a select minority of patients). Accumulating evidence is emerging to challenge this approach: early surgical intervention may offer the opportunity to interrupt the disease process before irreversible sequelae become established. This article provides an overview of the pathophysiology underlying chronic pancreatitis together with a review of the current evidence to support established and novel therapeutic approaches to the disease.
Collapse
|
72
|
Chen H, Tan P, Qian B, Du Y, Wang A, Shi H, Huang Z, Huang S, Liang T, Fu W. Hic-5 deficiency protects cerulein-induced chronic pancreatitis via down-regulation of the NF-κB (p65)/IL-6 signalling pathway. J Cell Mol Med 2019; 24:1488-1503. [PMID: 31797546 PMCID: PMC6991662 DOI: 10.1111/jcmm.14833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/16/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic pancreatitis (CP), characterized by pancreatic fibrosis, is a recurrent, progressive and irreversible disease. Activation of the pancreatic stellate cells (PSCs) is considered a core event in pancreatic fibrosis. In this study, we investigated the role of hydrogen peroxide-inducible clone-5 (Hic-5) in CP. Analysis of the human pancreatic tissue samples revealed that Hic-5 was overexpressed in patients with CP and was extremely low in healthy pancreas. Hic-5 was significant up-regulated in the activated primary PSCs independently from transforming growth factor beta stimulation. CP induced by cerulein injection was ameliorated in Hic-5 knockout (KO) mice, as shown by staining of tissue level. Simultaneously, the activation ability of the primary PSCs from Hic-5 KO mice was significantly attenuated. We also found that the Hic-5 up-regulation by cerulein activated the NF-κB (p65)/IL-6 signalling pathway and regulated the downstream extracellular matrix (ECM) genes such as α-SMA and Col1a1. Therefore, we determined whether suppressing NF-κB/p65 alleviated CP by treating mice with the NF-κB/p65 inhibitor triptolide in the cerulein-induced CP model and found that pancreatic fibrosis was alleviated by NF-κB/p65 inhibition. These findings provide evidence for Hic-5 as a therapeutic target that plays a crucial role in regulating PSCs activation and pancreatic fibrosis.
Collapse
Affiliation(s)
- Hao Chen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Peng Tan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Baolin Qian
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yichao Du
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ankang Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hao Shi
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhiwei Huang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shiyao Huang
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tiancheng Liang
- Luzhou Municipal Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
73
|
Vepakomma D. Pediatric Pancreatitis: Outcomes and Current Understanding. J Indian Assoc Pediatr Surg 2019; 25:22-27. [PMID: 31896895 PMCID: PMC6910056 DOI: 10.4103/jiaps.jiaps_223_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/12/2019] [Accepted: 02/02/2019] [Indexed: 12/13/2022] Open
Abstract
Aim: The aim of this study was to analyze the clinical spectrum of pediatric pancreatitis and review current literature with regard to concepts of disease and management approaches. Materials and Methods: This is a retrospective analysis of pancreatitis patients admitted to a tertiary referral pediatric surgical unit from March 2013 to September 2018. Results: There were 106 patients from 1 year to 18 years with median age group between 11 and 15 years and equal gender distribution. Sixty-two children had acute pancreatitis (AP), with 13 of them having recurrent episodes. Forty-four patients had chronic pancreatitis (CP). Definite etiology was found in 11 AP patients. All patients had radiological changes. All AP patients were treated symptomatically except those with correctable underlying etiology. Sixteen with CP required intervention in the form of endoscopic retrograde cholangiopancreatography sphincterotomy/stenting or surgery. There were two mortalities. One with acute necrotizing pancreatitis and one CP girl due to the complication of diabetes mellitus, unrelated to episode of pancreatitis. All others are well at the last follow-up. Conclusion: Pediatric pancreatitis is a disease with a wide spectrum, but management can be standardized. Newer studies reveal that certain genetic mutations make children more susceptible to pancreatitis. Increasing incidence must prompt us to evaluate further so as to better equip ourselves to managing this disease entity in all its forms and evolve preventive strategies.
Collapse
Affiliation(s)
- Deepti Vepakomma
- Department of Pediatric Surgery, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| |
Collapse
|
74
|
Li Z, Yu X, Werner J, Bazhin AV, D'Haese JG. The role of interleukin-18 in pancreatitis and pancreatic cancer. Cytokine Growth Factor Rev 2019; 50:1-12. [PMID: 31753718 DOI: 10.1016/j.cytogfr.2019.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023]
Abstract
Originally described as an interferon (IFN)-γ-inducing factor, interleukin (IL)-18 has been reported to be involved in Th1 and Th2 immune responses, as well as in activation of NK cells and macrophages. There is convincing evidence that IL-18 plays an important role in various pathologies (i.e. inflammatory diseases, cancer, chronic obstructive pulmonary disease, Crohn's disease and others). Recently, IL-18 has also been shown to execute specific effects in pancreatic diseases, including acute and chronic pancreatitis, as well as pancreatic cancer. The aim of this study was to give a profound review of recent data on the role of IL-18 and its potential as a therapeutic target in pancreatic diseases. The existing data on this topic are in part controversial and will be discussed in detail. Future studies should aim to confirm and clarify the role of IL-18 in pancreatic diseases and unravel their molecular mechanisms.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; Department of Hepatopancreatobiliary Surgery, The third Xiangya hospital, Central south university, Changsha 410013, Hunan, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, The third Xiangya hospital, Central south university, Changsha 410013, Hunan, China
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany.
| | - Jan G D'Haese
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| |
Collapse
|
75
|
Bai Y, Chen YB, Qiu XT, Chen YB, Ma LT, Li YQ, Sun HK, Zhang MM, Zhang T, Chen T, Fan BY, Li H, Li YQ. Nucleus tractus solitarius mediates hyperalgesia induced by chronic pancreatitis in rats. World J Gastroenterol 2019; 25:6077-6093. [PMID: 31686764 PMCID: PMC6824279 DOI: 10.3748/wjg.v25.i40.6077] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Central sensitization plays a pivotal role in the maintenance of chronic pain induced by chronic pancreatitis (CP). We hypothesized that the nucleus tractus solitarius (NTS), a primary central site that integrates pancreatic afferents apart from the thoracic spinal dorsal horn, plays a key role in the pathogenesis of visceral hypersensitivity in a rat model of CP.
AIM To investigate the role of the NTS in the visceral hypersensitivity induced by chronic pancreatitis.
METHODS CP was induced by the intraductal injection of trinitrobenzene sulfonic acid (TNBS) in rats. Pancreatic hyperalgesia was assessed by referred somatic pain via von Frey filament assay. Neural activation of the NTS was indicated by immunohistochemical staining for Fos. Basic synaptic transmission within the NTS was assessed by electrophysiological recordings. Expression of vesicular glutamate transporters (VGluTs), N-methyl-D-aspartate receptor subtype 2B (NR2B), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subtype 1 (GluR1) was analyzed by immunoblotting. Membrane insertion of NR2B and GluR1 was evaluated by electron microscopy. The regulatory role of the NTS in visceral hypersensitivity was detected via pharmacological approach and chemogenetics in CP rats.
RESULTS TNBS treatment significantly increased the number of Fos-expressing neurons within the caudal NTS. The excitatory synaptic transmission was substantially potentiated within the caudal NTS in CP rats (frequency: 5.87 ± 1.12 Hz in CP rats vs 2.55 ± 0.44 Hz in sham rats, P < 0.01; amplitude: 19.60 ± 1.39 pA in CP rats vs 14.71 ± 1.07 pA in sham rats; P < 0.01). CP rats showed upregulated expression of VGluT2, and increased phosphorylation and postsynaptic trafficking of NR2B and GluR1 within the caudal NTS. Blocking excitatory synaptic transmission via the AMPAR antagonist CNQX and the NMDAR antagonist AP-5 microinjection reversed visceral hypersensitivity in CP rats (abdominal withdraw threshold: 7.00 ± 1.02 g in CNQX group, 8.00 ± 0.81 g in AP-5 group and 1.10 ± 0.27 g in saline group, P < 0.001). Inhibiting the excitability of NTS neurons via chemogenetics also significantly attenuated pancreatic hyperalgesia (abdominal withdraw threshold: 13.67 ± 2.55 g in Gi group, 2.00 ± 1.37 g in Gq group, and 2.36 ± 0.67 g in mCherry group, P < 0.01).
CONCLUSION Our findings suggest that enhanced excitatory transmission within the caudal NTS contributes to pancreatic pain and emphasize the NTS as a pivotal hub for the processing of pancreatic afferents, which provide novel insights into the central sensitization of painful CP.
Collapse
Affiliation(s)
- Yang Bai
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ying-Biao Chen
- Department of Anatomy, Fujian Health College, Fuzhou 350101, Fujian Province, China
| | - Xin-Tong Qiu
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Yan-Bing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Li-Tian Ma
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ying-Qi Li
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hong-Ke Sun
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ming-Ming Zhang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Bo-Yuan Fan
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
- Joint Laboratory of Neuroscience at Hainan Medical University and Fourth Military Medical University, Hainan Medical University, Haikou 571199, Hainan Province, China
| |
Collapse
|
76
|
Wolske KM, Ponnatapura J, Kolokythas O, Burke LMB, Tappouni R, Lalwani N. Chronic Pancreatitis or Pancreatic Tumor? A Problem-solving Approach. Radiographics 2019; 39:1965-1982. [PMID: 31584860 DOI: 10.1148/rg.2019190011] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Certain inflammatory pancreatic abnormalities may mimic pancreatic ductal adenocarcinoma at imaging, which precludes accurate preoperative diagnosis and may lead to unnecessary surgery. Inflammatory conditions that may appear masslike include mass-forming chronic pancreatitis, focal autoimmune pancreatitis, and paraduodenal pancreatitis or "groove pancreatitis." In addition, obstructive chronic pancreatitis can mimic an obstructing ampullary mass or main duct intraductal papillary mucinous neoplasm. Secondary imaging features such as the duct-penetrating sign, biliary or main pancreatic duct skip strictures, a capsulelike rim, the pancreatic duct-to-parenchyma ratio, displaced calcifications in patients with chronic calcific pancreatitis, the "double duct" sign, and vessel encasement or displacement can help to suggest the possibility of an inflammatory mass or a neoplastic process. An awareness of the secondary signs that favor a diagnosis of malignant or inflammatory lesions in the pancreas can help the radiologist to perform the differential diagnosis and determine the degree of suspicion for malignancy. Repeat biopsy or surgical resection may be necessary to achieve an accurate diagnosis and prevent unnecessary surgery for inflammatory conditions. Online supplemental material and DICOM image stacks are available for this article. ©RSNA, 2019.
Collapse
Affiliation(s)
- Kristy Marie Wolske
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| | - Janardhana Ponnatapura
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| | - Orpheus Kolokythas
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| | - Lauren M B Burke
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| | - Rafel Tappouni
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| | - Neeraj Lalwani
- From the Departments of Radiology of Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27157 (K.M.W., J.P., R.T., N.L.); University of Washington, Seattle, Wash (O.K.); and University of North Carolina at Chapel Hill, Chapel Hill, NC (L.M.B.B.)
| |
Collapse
|
77
|
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: 27] [Impact Index Per Article: 4.5] [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
|
78
|
Abstract
OBJECTIVES Premature activation of the digestive protease trypsin within the pancreatic parenchyma is a critical factor in the pathogenesis of pancreatitis. Alterations in genes that affect intrapancreatic trypsin activity are associated with chronic pancreatitis (CP). Recently, carboxyl ester lipase emerged as a trypsin-independent risk gene. Here, we evaluated pancreatic lipase (PNLIP) as a potential novel susceptibility gene for CP. METHODS We analyzed all 13 PNLIP exons in 429 nonalcoholic patients with CP and 600 control subjects from Germany, in 632 patients and 957 controls from France, and in 223 patients and 1,070 controls from Japan by DNA sequencing. Additionally, we analyzed selected exons in further 545 patients with CP and 1,849 controls originating from Germany, United States, and India. We assessed the cellular secretion, lipase activity, and proteolytic stability of recombinant PNLIP variants. RESULTS In the German discovery cohort, 8/429 (1.9%) patients and 2/600 (0.3%) controls carried a PNLIP missense variant (P = 0.02, odds ratio [OR] = 5.7, 95% confidence interval [CI] = 1.1-38.9). Variants detected in patients were prone to proteolytic degradation by trypsin and chymotrypsin. In the French replication cohort, protease-sensitive variants were also enriched in patients with early-onset CP (5/632 [0.8%]) vs controls (1/957 [0.1%]) (P = 0.04, OR = 7.6, 95% CI = 0.9-172.9). In contrast, we detected no protease-sensitive variants in the non-European populations. In the combined European data, protease-sensitive variants were found in 13/1,163 cases (1.1%) and in 3/3,000 controls (0.1%) (OR = 11.3, 95% CI = 3.0-49.9, P < 0.0001). CONCLUSIONS Our data indicate that protease-sensitive PNLIP variants are novel genetic risk factors for the development of CP.
Collapse
|
79
|
Hirth M, Weiss C, Hardt P, Gubergrits N, Härtel N, Ebert MP, Schneider A. Analysis of the Course of Chronic Pancreatitis: Pancreatic Burnout Rates Are Only Increased in a Subgroup of Patients With Alcoholic Chronic Pancreatitis. Pancreas 2019; 48:726-733. [PMID: 31091222 DOI: 10.1097/mpa.0000000000001302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The pancreatic burnout hypothesis postulated an increasing absence of pain with simultaneous functional insufficiency in advanced stages of chronic pancreatitis (CP). However, the underlying data remain scarce and contradictory. We aimed to analyze, first, the frequency of a pancreatic burnout in CP, and, second, its association with etiological risk factors. METHODS We performed a multicenter, retrospective, cross-sectional study with 741 patients with CP categorized according to the M-ANNHEIM classification. Pancreatic burnout was defined by different combinations of exocrine or endocrine insufficiency with partial or complete absence of abdominal pain. RESULTS The frequency of a pancreatic burnout increased with prolonged disease duration and was observed in a maximum of 38% of patients after 20 years. Development of a pancreatic burnout was significantly associated with alcohol consumption (P < 0.05, Mann-Whitney U test), but not with other etiological risk factors. After a disease duration of more than 10 years, the likelihood of a burnout was 8 times higher in alcoholic CP than in nonalcoholic CP (95% confidence interval, 1.5-42.0; P = 0.015, logistic regression analysis). CONCLUSIONS A pancreatic burnout does not regularly occur in CP. Increased burnout rates are only observed in patients with alcoholic CP.
Collapse
Affiliation(s)
| | - Christel Weiss
- Medical Statistics, Mannheim Medical Faculty of the University Heidelberg, Mannheim
| | - Philip Hardt
- Department of Gastroenterology, University Hospital Giessen, Giessen, Germany
| | - Natalia Gubergrits
- Department of Internal Medicine, Donetsk National Medical University, Lyman, Ukraine
| | | | | | | |
Collapse
|
80
|
Jayasekara H, English DR, Hodge AM, Room R, Hopper JL, Milne RL, Giles GG, MacInnis RJ. Lifetime alcohol intake and pancreatic cancer incidence and survival: findings from the Melbourne Collaborative Cohort Study. Cancer Causes Control 2019; 30:323-331. [PMID: 30798509 DOI: 10.1007/s10552-019-01146-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Pancreatic cancer has one of the worst prognoses with 5-year survival below 10%. There is some evidence that alcohol consumption might increase the risk of pancreatic cancer. We examined associations of pre-diagnostic alcohol intake with (i) incidence of pancreatic cancer, and (ii) overall survival following pancreatic cancer. METHODS Usual alcohol intake was estimated at recruitment in 1990-1994 for 38,472 participants in the Melbourne Collaborative Cohort Study using recalled frequency and quantity of beverage-specific intake for 10-year periods from age 20. Pancreatic cancer incidence (C25 according to International Classification of Diseases for Oncology) and vital status were ascertained through to 30 September 2015. Cox regression was performed to estimate multivariable-adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for associations with lifetime, age 20-29, and baseline alcohol intakes. RESULTS By the end of follow-up (average 20.2 years), 239 incident cases of pancreatic cancer were diagnosed, of which 228 had died. No evidence of an association was observed between alcohol intake and risk of pancreatic cancer. Higher lifetime alcohol intake was associated with lower overall survival following a diagnosis of pancreatic cancer (mortality HR 1.09 per 10 g/day increment, 95% CI 1.00-1.19; p value = 0.04). A similar finding was observed for age 20-29 intake (HR 1.09 per 10 g/day increment, 95% CI 1.02-1.18; p value = 0.01) but not with baseline intake. CONCLUSIONS We observed an association between lower alcohol use from an early age and improved survival following pancreatic cancer, but this finding needs to be confirmed by other studies.
Collapse
Affiliation(s)
- Harindra Jayasekara
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia.
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.
- Centre for Alcohol Policy Research, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| | - Dallas R English
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| | - Allison M Hodge
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| | - Robin Room
- Centre for Alcohol Policy Research, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia
- Centre for Health Equity, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
- Centre for Social Research on Alcohol and Drugs, Stockholm University, 106 91, Stockholm, Sweden
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| | - Roger L Milne
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| | - Robert J MacInnis
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, 207 Bouverie Street, Melbourne, VIC, 3010, Australia
| |
Collapse
|
81
|
Boumezrag M, Harounzadeh S, Ijaz H, Johny A, Richards L, Ma Y, Le Saux MA, Kulie P, Davis C, Meltzer AC. Assessing the CT findings and clinical course of ED patients with first-time versus recurrent acute pancreatitis. Am J Emerg Med 2019; 37:304-307. [DOI: 10.1016/j.ajem.2018.10.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 10/25/2018] [Accepted: 10/31/2018] [Indexed: 12/11/2022] Open
|
82
|
Cerulein-induced chronic pancreatitis in Swiss albino mice: An improved short-term model for pharmacological screening. J Pharmacol Toxicol Methods 2019; 96:46-55. [PMID: 30684670 DOI: 10.1016/j.vascn.2019.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/13/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022]
Abstract
There is a need for short-term, reliable and reproducible animal model of chronic pancreatitis (CP) in small animals like mice. This study was aimed to establish the 9 exposures of cerulein-induced CP in mice. Repeated intraperitoneal cerulein injections were performed at 6 consecutive doses (50 μg/kg)/day, 3 days a week for 3 weeks to induce chronic pancreatitis in Swiss albino mice. The severity of damage was assessed by biochemical assays and histopathology. The expression of pro-inflammatory cytokine and fibrotic proteins was assessed by IHC and western blotting. The cerulein treated mice showed significantly elevated plasma amylase (p < .0285) and lipase levels (p < .0022) and resulted in significantly increased pancreatic oxidative (p < .0022) and nitrosative (p < .0022) stress. The hydroxyproline levels were 3.06 fold increased in the cerulein treated mice. The expressions of fibrotic cytokine TGF-β1 by 1.8 folds and pro-inflammatory cytokines TNF-α by 2.3 fold, IL-6 by 2.2 fold and IL-1β by 3.7 fold were markedly increased in cerulein treated mice. The histological evaluations indicated increased inflammatory cells infiltration and deposition of collagen. Moreover, the expression of fibrotic markers such as α-SMA increased by 2.5 folds (p < .00014), collagen1a by 1.3 folds (p < .0258) and fibronectin by 3.5 folds (p < .00014) were significantly increased. Our study demonstrates the superiority of 9 exposures of cerulein-induced CP model in mice with the reduction of duration, cerulein exposure, more economical and mortality rate of mice over the available models. Therefore, our model may be suitable to evaluate the pharmacological effects of new drugs in chronic pancreatitis.
Collapse
|
83
|
Manohar M, Kandikattu HK, Verma AK, Mishra A. IL-15 regulates fibrosis and inflammation in a mouse model of chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2018; 315:G954-G965. [PMID: 30212254 PMCID: PMC6336943 DOI: 10.1152/ajpgi.00139.2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pancreatitis is an inflammatory disease characterized by the induction of several proinflammatory cytokines like interleukin (IL)-6, IL-8, IL-1β, and IL-1. Recently, the multifunctional innate cytokine IL-15 has been implicated in the protection of several diseases, including cancer. Tissue fibrosis is one of the major problems in successfully treating chronic pancreatitis pathogenesis. Therefore, we tested the hypothesis that recombinant IL-15 (rIL-15) treatment may induce innate tissue responses and its overexpression will improve the pathogenesis of cerulein-induced chronic pancreatitis, associated remodeling, and fibrosis. We observed atrophy of acinar cells, increased inflammation, and increased deposition of perivascular collagen, the upregulated protein level of transforming growth factor (TGF)-β1, α-smooth muscle actin (α-SMA), and collagen-1 in cerulein-induced chronic pancreatitis in mice. Furthermore, we reported that rIL-15 treatment protects mice from the cerulein-induced chronic pancreatitis pathogenesis, including acinar cell atrophy, and perivascular accumulation of tissue collagen followed by downregulation of profibrotic genes such as TGF-β1, α-SMA, collagen-1, collagen-3, and fibronectin in cerulein-induced chronic pancreatitis in mice. Mechanistically, we show that IL-15-mediated increase of interferon-γ-responsive invariant natural killer T (iNKT) cells in the blood and tissue protects cerulein-induced pancreatic pathogenesis in mice. Of note, a reduction in iNKT cells was also observed in human chronic pancreatitis compared with normal individuals. Taken together, these data suggest that IL-15 treatment may be a novel therapeutic strategy for treating chronic pancreatitis pathogenesis. NEW & NOTEWORTHY Pancreatic fibrosis is a major concern for the successful treatment of chronic pancreatitis and pancreatic cancer. Therefore, restriction in the progression of fibrosis is the promising approach to manage the pancreatitis pathogenesis. Herein, we present in vivo evidences that pharmacological treatment of recombinant interleukin-15 improves remodeling and fibrosis in cerulein-induced chronic pancreatitis in mice. Our observations indicate that interleukin-15 immunotherapy may be a possible and potential strategy for restricting the progression of fibrosis in chronic pancreatitis.
Collapse
Affiliation(s)
- Murli Manohar
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Hemanth Kumar Kandikattu
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Alok Kumar Verma
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Anil Mishra
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
84
|
Dos Santos ALM, de Melo Santos H, Nogueira MB, Távora HTO, de Lourdes Jaborandy Paim da Cunha M, de Melo Seixas RBP, de Freitas Velloso Monte L, de Carvalho E. Cystic Fibrosis: Clinical Phenotypes in Children and Adolescents. Pediatr Gastroenterol Hepatol Nutr 2018; 21:306-314. [PMID: 30345244 PMCID: PMC6182489 DOI: 10.5223/pghn.2018.21.4.306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/26/2018] [Accepted: 07/22/2018] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The objective of this study was to describe the clinical phenotypes of children and adolescents with cystic fibrosis (CF); and to assess the role of pancreatic insufficiency and neonatal screening in diagnosis. METHODS A cross-sectional study was conducted, which included 77 patients attending a reference center of CF between 2014 and 2016. Epidemiological data, anthropometric measurements, and the presence of pulmonary, pancreatic, gastrointestinal and hepatobiliary manifestations were evaluated based on clinical data and complementary examinations. RESULTS Of the 77 patients, 51.9% were male, with a median age of 147 months (7.0-297.0 months), and the majority showed adequate nutritional status. The most common phenotype was pulmonary (92.2%), followed by pancreatic (87.0%), with pancreatic insufficiency in most cases. Gastrointestinal manifestation occurred in 46.8%, with constipation being the more common factor. Hepatobiliary disease occurred in 62.3% of patients. The group with pancreatic insufficiency was diagnosed earlier (5.0 months) when compared to the group with sufficiency (84.0 months) (p=0.01). The age of diagnosis was reduced following implementation of neonatal screening protocols for CF (6.0 months before vs. 3.0 months after, p=0.02). CONCLUSION The pulmonary phenotype was the most common, although extrapulmonary manifestations were frequent and clinically relevant, and should mandate early detection and treatment. Neonatal screening for CF led to earlier diagnosis in patients with pancreatic failure, and therefore, should be adopted universally.
Collapse
Affiliation(s)
- Ana Luiza Melo Dos Santos
- Department of Pediatric Gastroenterology, Brasília José Alencar Children's Hospital, Brasília, Brazil.,Department of Pediatric Gastroenterology, Distrito Federal Base Hospital, Brasília, Brazil
| | - Helen de Melo Santos
- Department of Pediatric Gastroenterology, Distrito Federal Base Hospital, Brasília, Brazil
| | - Marina Bettiol Nogueira
- Department of Pediatric Gastroenterology, Brasília José Alencar Children's Hospital, Brasília, Brazil
| | | | - Maria de Lourdes Jaborandy Paim da Cunha
- Department of Pediatric Gastroenterology, Brasília José Alencar Children's Hospital, Brasília, Brazil.,Department of Pediatric Gastroenterology, Distrito Federal Base Hospital, Brasília, Brazil
| | - Renata Belém Pessoa de Melo Seixas
- Department of Pediatric Gastroenterology, Brasília José Alencar Children's Hospital, Brasília, Brazil.,Department of Pediatric Gastroenterology, Distrito Federal Base Hospital, Brasília, Brazil
| | | | - Elisa de Carvalho
- Department of Pediatric Gastroenterology, Brasília José Alencar Children's Hospital, Brasília, Brazil.,Department of Pediatric Gastroenterology, Distrito Federal Base Hospital, Brasília, Brazil
| |
Collapse
|
85
|
Rosendahl J, Kirsten H, Hegyi E, Kovacs P, Weiss FU, Laumen H, Lichtner P, Ruffert C, Chen JM, Masson E, Beer S, Zimmer C, Seltsam K, Algül H, Bühler F, Bruno MJ, Bugert P, Burkhardt R, Cavestro GM, Cichoz-Lach H, Farré A, Frank J, Gambaro G, Gimpfl S, Grallert H, Griesmann H, Grützmann R, Hellerbrand C, Hegyi P, Hollenbach M, Iordache S, Jurkowska G, Keim V, Kiefer F, Krug S, Landt O, Leo MD, Lerch MM, Lévy P, Löffler M, Löhr M, Ludwig M, Macek M, Malats N, Malecka-Panas E, Malerba G, Mann K, Mayerle J, Mohr S, te Morsche RHM, Motyka M, Mueller S, Müller T, Nöthen MM, Pedrazzoli S, Pereira SP, Peters A, Pfützer R, Real FX, Rebours V, Ridinger M, Rietschel M, Rösmann E, Saftoiu A, Schneider A, Schulz HU, Soranzo N, Soyka M, Simon P, Skipworth J, Stickel F, Strauch K, Stumvoll M, Testoni PA, Tönjes A, Werner L, Werner J, Wodarz N, Ziegler M, Masamune A, Mössner J, Férec C, Michl P, P H Drenth J, Witt H, Scholz M, Sahin-Tóth M. Genome-wide association study identifies inversion in the CTRB1-CTRB2 locus to modify risk for alcoholic and non-alcoholic chronic pancreatitis. Gut 2018; 67:1855-1863. [PMID: 28754779 PMCID: PMC6145291 DOI: 10.1136/gutjnl-2017-314454] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/16/2017] [Accepted: 06/24/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Alcohol-related pancreatitis is associated with a disproportionately large number of hospitalisations among GI disorders. Despite its clinical importance, genetic susceptibility to alcoholic chronic pancreatitis (CP) is poorly characterised. To identify risk genes for alcoholic CP and to evaluate their relevance in non-alcoholic CP, we performed a genome-wide association study and functional characterisation of a new pancreatitis locus. DESIGN 1959 European alcoholic CP patients and population-based controls from the KORA, LIFE and INCIPE studies (n=4708) as well as chronic alcoholics from the GESGA consortium (n=1332) were screened with Illumina technology. For replication, three European cohorts comprising 1650 patients with non-alcoholic CP and 6695 controls originating from the same countries were used. RESULTS We replicated previously reported risk loci CLDN2-MORC4, CTRC, PRSS1-PRSS2 and SPINK1 in alcoholic CP patients. We identified CTRB1-CTRB2 (chymotrypsin B1 and B2) as a new risk locus with lead single-nucleotide polymorphism (SNP) rs8055167 (OR 1.35, 95% CI 1.23 to 1.6). We found that a 16.6 kb inversion in the CTRB1-CTRB2 locus was in linkage disequilibrium with the CP-associated SNPs and was best tagged by rs8048956. The association was replicated in three independent European non-alcoholic CP cohorts of 1650 patients and 6695 controls (OR 1.62, 95% CI 1.42 to 1.86). The inversion changes the expression ratio of the CTRB1 and CTRB2 isoforms and thereby affects protective trypsinogen degradation and ultimately pancreatitis risk. CONCLUSION An inversion in the CTRB1-CTRB2 locus modifies risk for alcoholic and non-alcoholic CP indicating that common pathomechanisms are involved in these inflammatory disorders.
Collapse
Affiliation(s)
- Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Eszter Hegyi
- Department of Molecular and Cell Biology, Center for Exocrine Disorders, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Frank Ulrich Weiss
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Helmut Laumen
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Claudia Ruffert
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1078; Etablissement Français du Sang (EFS) – Bretagne; Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale; Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France
| | - Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1078; Etablissement Français du Sang (EFS) – Bretagne; Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale; Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France
| | - Sebastian Beer
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Constantin Zimmer
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Katharina Seltsam
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Hana Algül
- Department of Gastroenterology, Technische Universität München (TUM), Munich, Germany
| | - Florence Bühler
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus Medical Centre, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg, Mannheim, Germany
| | - Ralph Burkhardt
- LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Giulia Martina Cavestro
- Division of Gastroenterology and Gastrointestinal Endoscopy, Vita Salute San Raffaele University - San Raffaele Scientific Institute, Milan, Italy
| | - Halina Cichoz-Lach
- Department of Gastroenterology, Medical University of Lublin, Lublin, Poland
| | - Antoni Farré
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Institute of Internal Medicine, Renal Program, Columbus-Gemelli University Hospital, Catholic University, Rome, Italy
| | - Sebastian Gimpfl
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Heidi Griesmann
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Robert Grützmann
- Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Chirurgische Klinik, Erlangen, Germany
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Péter Hegyi
- Institute for Translational Medicine and First Department of Internal Medicine, University of Pécs, Pécs, Hungary
- HAS-SZTE, Momentum Gastroenterology Multidisciplinary Research Group, Szeged, Hungary
| | - Marcus Hollenbach
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Sevastitia Iordache
- Department of Internal Medicine and Gastroenterology, University of Medicine and Pharmacy, Craiova, Romania
| | - Grazyna Jurkowska
- Department of Gastroenterology and Internal Medicine, Medical University Bialystok, Bialystok, Poland
| | - Volker Keim
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Sebastian Krug
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | | | - Milena Di Leo
- Division of Gastroenterology and Gastrointestinal Endoscopy, Vita Salute San Raffaele University - San Raffaele Scientific Institute, Milan, Italy
| | - Markus M Lerch
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Philippe Lévy
- Pôle des Maladies de l’Appareil Digestif, Service de Gastroentérologie-Pancréatologie, Hôpital Beaujon, AP-HP, Clichy, France
| | - Markus Löffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Matthias Löhr
- Gastrocentrum, Karolinska Institutet CLINTEC, Stockholm, Sweden
| | - Maren Ludwig
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Milan Macek
- Department of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nuria Malats
- Grupo de Epidemiología Genética y Molecular Programa de Genética del Cáncer Humano Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBERONC, Spain
| | - Ewa Malecka-Panas
- Department of Digestive Tract Diseases, Medical University of Łódź, Łódź, Poland
| | - Giovanni Malerba
- Biology and Genetics, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Karl Mann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sonja Mohr
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Rene H M te Morsche
- Department of Gastroenterology and Hepatology, Radboud umc, Nijmegen, The Netherlands
| | - Marie Motyka
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Sebastian Mueller
- Department of Internal Medicine, Salem Medical Centre and Centre for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Thomas Müller
- Department of Pediatrics I, Medical University, Innsbruck, Austria
| | - Markus M Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Sergio Pedrazzoli
- Department of Medical and Surgical Sciences, IV Surgical Clinic, University of Padua, Padua, Italy
| | - Stephen P Pereira
- Division of Medicine, UCL Institute for Liver and Digestive Health, University College London, London, UK
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Roland Pfützer
- Clinic for Internal Medicine, Hospital Döbeln, Döbeln, Germany
| | - Francisco X Real
- CIBERONC, Spain
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vinciane Rebours
- Pôle des Maladies de l’Appareil Digestif, Service de Gastroentérologie-Pancréatologie, Hôpital Beaujon, AP-HP, Clichy, France
| | - Monika Ridinger
- Department of Psychiatry, University of Regensburg, Regensburg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Eva Rösmann
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Adrian Saftoiu
- Department of Internal Medicine and Gastroenterology, University of Medicine and Pharmacy, Craiova, Romania
| | - Alexander Schneider
- Department of Gastroenterology, Hepatology, Infectious Diseases, Medical Faculty of Mannheim University of Heidelberg, Mannheim, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Nicole Soranzo
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Michael Soyka
- Psychiatric Hospital, University of Munich, Munich, Germany
| | - Peter Simon
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - James Skipworth
- Department of Surgery and Interventional Science, University College London, London, UK
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital of Zürich, Zürich, Switzerland
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Michael Stumvoll
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Pier Alberto Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, Vita Salute San Raffaele University - San Raffaele Scientific Institute, Milan, Italy
| | - Anke Tönjes
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Lena Werner
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig Maximilian University, Munich, Germany
| | - Norbert Wodarz
- Department of Psychiatry, University of Regensburg, Regensburg, Germany
| | - Martin Ziegler
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, SendaiMiyagi, Japan
| | - Joachim Mössner
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1078; Etablissement Français du Sang (EFS) – Bretagne; Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale; Laboratoire de Génétique Moléculaire et d’Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France
| | - Patrick Michl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud umc, Nijmegen, The Netherlands
| | - Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Center for Exocrine Disorders, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
86
|
Halbrook CJ, Pasca di Magliano M, Lyssiotis CA. Tumor cross-talk networks promote growth and support immune evasion in pancreatic cancer. Am J Physiol Gastrointest Liver Physiol 2018; 315. [PMID: 29543507 PMCID: PMC6109710 DOI: 10.1152/ajpgi.00416.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In the event of an injury, normal tissues exit quiescent homeostasis and rapidly engage a complex stromal and immune program. These tissue repair responses are hijacked and become dysregulated in carcinogenesis to form a growth-supportive tumor microenvironment. In pancreatic ductal adenocarcinoma (PDA), which remains one of the deadliest major cancers, the microenvironment is a key driver of tumor maintenance that impedes many avenues of therapy. In this review, we outline recent efforts made to uncover the microenvironmental cross-talk mechanisms that support pancreatic cancer cells, and we detail the strategies that have been undertaken to help overcome these barriers.
Collapse
Affiliation(s)
- Christopher J. Halbrook
- 1Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Marina Pasca di Magliano
- 2Department of Surgery, University of Michigan, Ann Arbor, Michigan,3Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Costas A. Lyssiotis
- 1Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan,3Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan,4Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
87
|
Weiss FU, Hesselbarth N, Párniczky A, Mosztbacher D, Lämmerhirt F, Ruffert C, Kovacs P, Beer S, Seltsam K, Griesmann H, Böhme R, Kaune T, Hollenbach M, Schulz HU, Simon P, Mayerle J, Lerch MM, Cavestro GM, Zuppardo RA, Di Leo M, Testoni PA, Malecka-Panas E, Gasirowska A, Głuszek S, Bugert P, Szentesi A, Mössner J, Witt H, Michl P, Hégyi P, Scholz M, Rosendahl J. Common variants in the CLDN2-MORC4 and PRSS1-PRSS2 loci confer susceptibility to acute pancreatitis. Pancreatology 2018; 18:477-481. [PMID: 29884332 DOI: 10.1016/j.pan.2018.05.486] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Acute pancreatitis (AP) is one of the most common gastrointestinal disorders often requiring hospitalization. Frequent aetiologies are gallstones and alcohol abuse. In contrast to chronic pancreatitis (CP) few robust genetic associations have been described. Here we analysed whether common variants in the CLDN2-MORC4 and the PRSS1-PRSS2 locus that increase recurrent AP and CP risk associate with AP. METHODS We screened 1462 AP patients and 3999 controls with melting curve analysis for SNPs rs10273639 (PRSS1-PRSS2), rs7057398 (RIPPLY), and rs12688220 (MORC4). Calculations were performed for the overall group, aetiology, and gender sub-groups. To examine genotype-phenotype relationships we performed several meta-analyses. RESULTS Meta-analyses of all AP patients depicted significant (p-value < 0.05) associations for rs10273639 (odds ratio (OR) 0.88, 95% confidence interval (CI) 0.81-0.97, p-value 0.01), rs7057398 (OR 1.27, 95% CI 1.07-1.5, p-value 0.005), and rs12688220 (OR 1.32, 95% CI 1.12-1.56, p-value 0.001). For the different aetiology groups a significant association was shown for rs10273639 (OR 0.76, 95% CI 0.63-0.92, p-value 0.005), rs7057398 (OR 1.43, 95% CI 1.07-1.92, p-value 0.02), and rs12688220 (OR 1.44, 95% CI 1.07-1.93, p-value 0.02) in the alcoholic sub-group only. CONCLUSIONS The association of CP risk variants with different AP aetiologies, which is strongest in the alcoholic AP group, might implicate common pathomechanisms most likely between alcoholic AP and CP.
Collapse
Affiliation(s)
- Frank Ulrich Weiss
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Nico Hesselbarth
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Andrea Párniczky
- Heim Pál Children's Hospital, Budapest, Hungary; Institute for Translational Medicine and First Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Dora Mosztbacher
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Felix Lämmerhirt
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Claudia Ruffert
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Sebastian Beer
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Katharina Seltsam
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Heidi Griesmann
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Richard Böhme
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Tom Kaune
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Marcus Hollenbach
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter Simon
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Julia Mayerle
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany; Department of Medicine II, University Hospital, Ludwig-Maximilians-University Munich, Germany
| | - Markus M Lerch
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Alessia Zuppardo
- Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Milena Di Leo
- Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Pier Alberto Testoni
- Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Ewa Malecka-Panas
- Department of Digestive Tract Diseases, Medical University of Łódź, Łódź, Poland
| | - Anita Gasirowska
- Department of Digestive Tract Diseases, Medical University of Łódź, Łódź, Poland
| | - Stanislaw Głuszek
- Faculty of Medicine and Health Sciences, Jan Kochanowski University, Kielce, Poland
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg, Mannheim, Germany
| | - Andrea Szentesi
- Institute for Translational Medicine and First Department of Internal Medicine, University of Pécs, Pécs, Hungary; First Department of Medicine, University of Szeged, Hungary
| | - Joachim Mössner
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany
| | - Patrick Michl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Peter Hégyi
- Institute for Translational Medicine and First Department of Internal Medicine, University of Pécs, Pécs, Hungary; HAS-SZTE, Momentum Gastroenterology Multidisciplinary Research Group, Szeged, Hungary
| | - Markus Scholz
- LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany.
| |
Collapse
|
88
|
Liu DM. Effect of comprehensive nursing intervention on self-management ability and quality of life in acute pancreatitis patients with diabetes mellitus. Shijie Huaren Xiaohua Zazhi 2018; 26:999-1004. [DOI: 10.11569/wcjd.v26.i16.999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To explore if comprehensive nursing intervention can improve self-management ability and quality of life in patients with acute pancreatitis and diabetes mellitus (DM).
METHODS One hundred and fourteen acute pancreatitis patients with DM were divided into a control group and a study group, with 57 cases in each group. All patients received the same treatments. The control group was given routine nursing intervention, and the study group received comprehensive nursing intervention. Fasting blood glucose (FBG), 2 h blood glucose (PG), and glycosylated hemoglobin (HbA1c) were measured before and after nursing intervention. The self-management ability and quality of life before and after intervention were compared between the two groups.
RESULTS The levels of FBG, 2 h PG, and HbA1c at weeks 12 and 24 after nursing intervention in the study group were significantly lower than those at corresponding time points in the control group (P < 0.05). The levels of FBG, 2 h PG, and HbA1c were different among different time points in each group (P < 0.05). The scores of self-management ability and quality of life were significantly higher in the study group than in the control group (P < 0.05).
CONCLUSION Comprehensive nursing intervention can effectively control blood sugar level in acute pancreatitis patients with DM, which is of great significance for improving patients' self-management ability and quality of life.
Collapse
Affiliation(s)
- Dong-Mei Liu
- Department of Hematology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
89
|
Abstract
RATIONALE Pancreatic pseudocyst can present single or multiple, inside or outside the pancreas. Pancreatic panniculitis is a rare skin lesion in pancreatic disease patients. The purpose of this study is to report a case of chronic pancreatitis coexisting with multiple pseudocysts and pancreatic panniculitis. PATIENT CONCERNS A 46-year-old man with chronic pancreatitis presented multiple small cystic lesions inside the head of the pancreas and two large cystic lesions adjacent to the tail of the pancreas. The patient also developed subcutaneous nodules involving upper and lower limbs, hands, and lower abdomen bilaterally. DIAGNOSIS The patient was diagnosed with pancreatic pseudocyst and pancreatic panniculitis resulted from chronic pancreatitis. INTERVENTIONS Bile duct stent and pancreatic duct stent placement was performed endoscopicly. OUTCOMES Panniculitis faded three weeks later and the pancreatic pseudocysts disappeared six weeks later. LESSONS Clinicians should be aware of the manifestation of multiple pancreatic pseudocyst and pancreatic panniculitis, and endoscopic transpapillary drainage may be a effective way in this scenario.
Collapse
|
90
|
Ní Chonchubhair HM, Bashir Y, Dobson M, Ryan BM, Duggan SN, Conlon KC. The prevalence of small intestinal bacterial overgrowth in non-surgical patients with chronic pancreatitis and pancreatic exocrine insufficiency (PEI). Pancreatology 2018; 18:379-385. [PMID: 29502987 DOI: 10.1016/j.pan.2018.02.010] [Citation(s) in RCA: 42] [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] [Received: 12/18/2017] [Revised: 02/13/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Small intestinal bacterial overgrowth (SIBO) is a condition characterised by symptoms similar to pancreatic exocrine insufficiency (PEI) in chronic pancreatitis patients. SIBO is thought to complicate chronic pancreatitis in up to 92% of cases; however, studies are heterogeneous and protocols non-standardised. SIBO may be determined by measuring lung air-expiration of either hydrogen or methane which are by-products of small bowel bacterial fermentation of intraluminal substrates such as carbohydrates. We evaluated the prevalence of SIBO among a defined cohort of non-surgical chronic pancreatitics with mild to severe PEI compared with matched healthy controls. METHODS Thirty-five patients and 31 age-, gender- and smoking status-matched healthy controls were evaluated for SIBO by means of a fasting glucose hydrogen breath test (GHBT). The relationship between SIBO and clinical symptoms in chronic pancreatitis was evaluated. RESULTS SIBO was present in 15% of chronic pancreatitis patients, while no healthy controls tested positive (P = 0.029). SIBO was more prevalent in those taking pancreatic enzyme replacement therapy (PERT) (P = 0.016), with proton pump inhibitor use (PPI) (P = 0.022) and in those with alcohol aetiology (P = 0.023). Patients with concurrent diabetes were more often SIBO-positive and this was statistically significant (P = 0.009). There were no statistically significant differences in reported symptoms between patients with and without SIBO, with the exception of 'weight loss', with patients reporting weight loss more likely to have SIBO (P = 0.047). CONCLUSION The prevalence of SIBO in this study was almost 15% and consistent with other studies of SIBO in non-surgical chronic pancreatitis patients. These data support the testing of patients with clinically-relevant PEI unresolved by adequate doses of PERT, particularly in those patients with concurrent diabetes. SIBO can be easily diagnosed therefore allowing more specific and more targeted symptom treatment.
Collapse
Affiliation(s)
- Hazel M Ní Chonchubhair
- Professorial Surgical Unit, Department of Surgery, The University of Dublin Trinity College, Tallaght Hospital, Dublin 24, Ireland.
| | - Yasir Bashir
- Professorial Surgical Unit, Department of Surgery, The University of Dublin Trinity College, Tallaght Hospital, Dublin 24, Ireland
| | - Mark Dobson
- Department of Gastroenterology, Tallaght Hospital, Dublin 24, Ireland
| | - Barbara M Ryan
- Department of Gastroenterology, Tallaght Hospital, Dublin 24, Ireland
| | - Sinead N Duggan
- Professorial Surgical Unit, Department of Surgery, The University of Dublin Trinity College, Tallaght Hospital, Dublin 24, Ireland
| | - Kevin C Conlon
- Professorial Surgical Unit, Department of Surgery, The University of Dublin Trinity College, Tallaght Hospital, Dublin 24, Ireland
| |
Collapse
|
91
|
Abstract
Chronic pancreatitis is a syndrome involving inflammation, fibrosis, and loss of acinar and islet cells which can manifest in unrelenting abdominal pain, malnutrition, and exocrine and endocrine insufficiency. The Toxic-Metabolic, Idiopathic, Genetic, Autoimmune, Recurrent and Severe Acute Pancreatitis, Obstructive (TIGAR-O) classification system categorizes known causes and factors that contribute to chronic pancreatitis. Although determining disease etiology provides a framework for focused and specific treatments, chronic pancreatitis remains a challenging condition to treat owing to the often refractory, centrally mediated pain and the lack of consensus regarding when endoscopic therapy and surgery are indicated. Further complications incurred include both exocrine and endocrine pancreatic insufficiency, pseudocyst formation, bile duct obstruction, and pancreatic cancer. Medical treatment of chronic pancreatitis involves controlling pain, addressing malnutrition via the treatment of vitamin and mineral deficiencies and recognizing the risk of osteoporosis, and administering appropriate pancreatic enzyme supplementation and diabetic agents. Cornerstones in treatment include the recognition of pancreatic exocrine insufficiency and administration of pancreatic enzyme replacement therapy, support to cease smoking and alcohol consumption, consultation with a dietitian, and a systematic follow-up to assure optimal treatment effect.
Collapse
Affiliation(s)
- Angela Pham
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, Florida, USA
| | - Christopher Forsmark
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
92
|
Wang M, Gao F, Wang X, Liu Y, Ji R, Cang L, Shi Y. Magnetic resonance elastography and T 1 mapping for early diagnosis and classification of chronic pancreatitis. J Magn Reson Imaging 2018; 48:10.1002/jmri.26008. [PMID: 29537715 PMCID: PMC6138575 DOI: 10.1002/jmri.26008] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/24/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early detection and classification of chronic pancreatitis (CP) are both important and challenging. PURPOSE To investigate the diagnostic performance of MR elastography (MRE) and T1 mapping of the pancreas for different stages of CP. STUDY TYPE Retrospective. SUBJECTS Clinical and imaging records of 81 patients (from 5/2015 to 7/2017) with suspected CP were analyzed. Patients were categorized into the normal control (n = 35), mild CP (n = 30), and moderate/severe CP groups (n = 16) according to the Cambridge Classification based on concordant endoscopic retrograde cholangiopancreatography or ultrasound endoscopy findings. FIELD STRENGTH/SEQUENCE 3T pancreatic MRI, which included MRE and T1 mapping. ASSESSMENT T1 relaxation times, pancreatic stiffness values, the main pancreatic duct (MPD) diameter, and pancreatic thickness were measured in all patients. Statistical Tests: Cutoff values of T1 relaxation times and pancreatic stiffness values for diagnosis of CP were calculated using receiver operating characteristic analysis. Associations of imaging parameters with different stages of CP were assessed using logistic regression analysis. RESULTS Both T1 relaxation times (865 ± 220 msec vs. 1075 ± 221 msec vs. 1350 ± 139 msec) and pancreatic stiffness (1.21 ± 0.13 kPa vs. 1.50 ± 0.15 kPa vs. 1.90 ± 0.16 kPa) differed significantly (P < 0.001) among the control, mild CP, and moderate/severe CP groups. Pancreatic stiffness (>1.34 kPa) achieved significantly higher area under the curve (AUC) than T1 relaxation time (>908.4 msec) for detection of mild CP (AUC: 0.928 vs. 0.751, P = 0.011). Pancreatic stiffness values (>1.61 kPa) also achieved significantly higher AUC than T1 relaxation time (>1131.6 msec) (AUC: 0.981 vs. 0.910, P = 0.033) for diagnosing moderate/severe CP from the other two groups. Multiple regression analysis showed that T1 relaxation time and stiffness were the independent factors associated with mild CP (P = 0.025 and <0.001, respectively). DATA CONCLUSION Both MRE and T1 mapping are promising quantitative imaging methods for evaluation of CP; MRE slightly outperformed T1 mapping. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 1 J. Magn. Reson. Imaging 2018.
Collapse
Affiliation(s)
- Min Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Feng Gao
- Department of Pancreato-thyroidic Surgery, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | | | - Yanqing Liu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Ruoyun Ji
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Lizhuo Cang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
93
|
Shi Y, Gao F, Li Y, Tao S, Yu B, Liu Z, Liu Y, Glaser KJ, Ehman RL, Guo Q. Differentiation of benign and malignant solid pancreatic masses using magnetic resonance elastography with spin-echo echo planar imaging and three-dimensional inversion reconstruction: a prospective study. Eur Radiol 2018; 28:936-945. [PMID: 28986646 PMCID: PMC5812826 DOI: 10.1007/s00330-017-5062-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 08/11/2017] [Accepted: 09/06/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To determine the diagnostic performance of MR elastography (MRE) and compare it with serum CA19-9 in differentiating malignant from benign pancreatic masses, with emphasis on differentiating between pancreatic ductal adenocarcinoma (PDAC) and mass-forming pancreatitis (MFP). METHODS We performed a prospective, consecutive, 24-month study in 85 patients with solid pancreatic masses confirmed by histopathologic examinations. The mass stiffness and stiffness ratio (calculated as the ratio of mass stiffness to the parenchymal stiffness) were assessed. The diagnostic accuracy was analysed by calculating the area under the ROC curve (AUROC). RESULTS The final diagnosis included 54 malignant tumours (43 patients with PDAC) and 31 benign masses (24 patients with MFP). The stiffness ratio showed better diagnostic performance than the mass stiffness and serum CA19-9 for the differentiation between malignant and benign masses (AUC: 0.912 vs. 0.845 vs. 0.702; P = 0.026, P < 0.001) and, specifically, between PDAC and MFP (AUC: 0.955 vs. 0.882 vs. 0.745; P = 0.026, P = 0.003). The sensitivity, specificity, and accuracy of stiffness ratio for the differentiation of PDAC and MFP were all higher than 0.9. CONCLUSIONS MRE presents an effective and quantitative strategy for non-invasive differentiation between PDAC and MFP based on their mechanical properties. KEY POINTS • 3D MRE is useful for calculating stiffness of solid pancreatic tumours. • Stiffness ratio outperformed stiffness and CA19-9 for differentiating PDAC from MFP. • Incorporation of 3D MRE into a standard MRI protocol is recommended.
Collapse
Affiliation(s)
- Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Feng Gao
- Department of Hepato-Pancreato-Biliary Tumour Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yue Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Shengzhen Tao
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Bing Yu
- Department of Radiology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong, People's Republic of China
| | - Yanqing Liu
- Department of Radiology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Kevin J Glaser
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Qiyong Guo
- Department of Radiology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
94
|
Pancreatic inflammation and atrophy are not associated with pancreatic cancer concomitant with intraductal papillary mucinous neoplasm. Pancreatology 2018; 18:54-60. [PMID: 29269290 DOI: 10.1016/j.pan.2017.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 11/29/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammation-induced carcinogenesis in pancreatic ductal adenocarcinoma (PDAC) has been reported; however, its involvement in PDAC with intraductal papillary mucinous neoplasm (IPMN) remains unclear. We herein investigated the relationship between pancreatic atrophy and inflammation and the incidence of PDAC concomitant with IPMN. METHODS This study included 178 consecutive patients who underwent surgical resection for PDAC with IPMN (N = 21) and IPMN (N = 157) between April 2001 and October 2016. A multivariable logistic regression analysis was conducted to assess the relationship between pancreatic inflammation and atrophy and the incidence of PDAC concomitant with IPMN, with adjustments for clinical characteristics and imaging features. Pathological pancreatic inflammation and atrophy were evaluated in resected specimens. RESULTS High degrees of pancreatic inflammation and atrophy were not associated with the incidence of PDAC with IPMN (multivariable odds ratio [OR] = 0.5, 95% confidence interval [CI] = 0.07 to 3.33, P = .52, adjusted by clinical characteristics, OR = 0.9, 95% CI = 0.10 to 5.86, P = .91, adjusted by imaging studies; OR = 0.2, 95% CI = 0.009 to 1.31, P = .10, adjusted by clinical characteristics, OR = 0.2, 95% CI = 0.01 to 1.43, P = .12, adjusted by imaging studies, respectively). CONCLUSIONS Pancreatic inflammation and atrophy were not associated with pancreatic cancer concomitant with IPMN.
Collapse
|
95
|
Bansal RK, Patil GK, Puri R, Choudhary NS, Patle SR, Sharma ZD, Sud R. Endoscopic pancreatic balloon sphincteroplasty for difficult to treat pancreatic stones and strictures: experience in 80 patients. Endosc Int Open 2017; 5:E1229-E1234. [PMID: 29218314 PMCID: PMC5718908 DOI: 10.1055/s-0043-119752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/24/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIM There is paucity of data about endoscopic pancreatic sphincteroplasty (EPS) after endoscopic pancreatic sphincterotomy (EPST) in the treatment of chronic pancreatitis. The aim of this study was to establish the indications for EPS, complications related to it, and to examine its effectiveness in managing chronic pancreatitis after a year of follow-up. METHODS We evaluated the safety and efficacy of pancreatic balloon dilation coupled with sphincterotomy for the treatment of chronic pancreatitis. The technical success rate of balloon dilation, stone clearance, frequency of pancreatic stenting, and procedure-related adverse events were recorded. RESULTS Out of 580 patients who underwent pancreatic endotherapy between July 2014 and February 2016, 80 patients underwent EPS. The mean age of these 80 patients was 34 ± 11 years, and 80 % (n = 64) were males. The common indications were removal of large radiolucent stones in 31 patients; unyielding radiopaque stones post extracorporeal shock wave lithotripsy (ESWL) in 20 patients, and pancreatic duct stricture combined with stones in 29 patients. EPS could be successfully completed in 98.75 % of patients. Complete ductal clearance in a single session was achieved in only 25 patients, while 26 patients required two sessions. There were two adverse events of pain requiring admission for more than 24 hours and one procedure related bleeding, all of which were managed conservatively. The patients had an average follow-up of 8 months (6 - 12 months) and all the patients were pain free. CONCLUSIONS Endoscopic pancreatic sphincteroplasty is a relatively safe procedure with a low incidence of complications and a high rate of treatment success.
Collapse
Affiliation(s)
- Rinkesh Kumar Bansal
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| | - Gaurav Kumar Patil
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| | - Rajesh Puri
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India,Corresponding author Rajesh Puri, MD DNB Institute of Digestive and Hepatobiliary SciencesMedanta – The MedicitySector 38GurugramDelhi NCRIndiaPIN 122001+91-12-44834445
| | - Narendra S. Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| | - Saurabh R. Patle
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| | - Zubin D. Sharma
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| | - Randhir Sud
- Institute of Digestive and Hepatobiliary Sciences, Medanta – The Medicity, Gurugram, Haryana, India
| |
Collapse
|
96
|
Hu Y, Teng C, Yu S, Wang X, Liang J, Bai X, Dong L, Song T, Yu M, Qu J. Inonotus obliquus polysaccharide regulates gut microbiota of chronic pancreatitis in mice. AMB Express 2017; 7:39. [PMID: 28197985 PMCID: PMC5309192 DOI: 10.1186/s13568-017-0341-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
Polysaccharide is efficient in attenuation of metabolic ailments and modulation of gut microbiota as prebiotics. The therapeutic effect of Inonotus obliquus polysaccharide (IOP) on chronic pancreatitis (CP) in mice has been validated in our previous study. However, it is not clear whether IOP is conducive to maintaining the homeostasis between gut microbiota and host. The aim of this study is to testify the potential effects of IOP on gut microbiota composition and diversity in mice with CP. The changes in glutathione peroxidase (GSH-PX), total antioxidant capacity (TAOC), tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β), lipase and trypsin levels were measured by commercial assay kits, meanwhile the gut microbiota composition and diversity were analyzed by high throughput sequencing. The IOP treatment increased GSH-PX and TAOC levels, and decreased TNF-α, TGF-β, lipase and trypsin levels in CP mice. It was also observed that gut microbiota in IOP treated groups were less diverse than others in terms of lower Shannon diversity index and Chao 1 estimator. IOP increased the proportion of Bacteroidetes and decreased that of Firmicutes at phylum level. Bacteroidetes was found positively correlated with GSH-PX and TAOC, and Firmicutes correlated with TNF-α, TGF-β, and lipase. In conclusion, administration of IOP could regulate gut microbiota composition and diversity to a healthy profile in mice with CP, and some bacterial phylum significantly correlated with characteristic parameters.
Collapse
|
97
|
Chymotrypsinogen C Genetic Variants, Including c.180TT, Are Strongly Associated With Chronic Pancreatitis in Pediatric Patients. J Pediatr Gastroenterol Nutr 2017; 65:652-657. [PMID: 28968289 DOI: 10.1097/mpg.0000000000001767] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Genetic studies in adults/adolescent patients with chronic pancreatitis (CP) identified chymotrypsinogen C (CTRC) genetic variants but their association with CP risk has been difficult to replicate. To evaluate the risk of CP associated with CTRC variants in CP pediatric patients-control study. METHODS The distribution of CTRC variants in CP pediatric cohort (n = 136, median age at CP onset 8 years) with no history of alcohol/smoking abuse was compared with controls (n = 401, median age 45). RESULTS We showed that p.Arg254Trp (4.6%) and p.Lys247_Arg254del (5.3%) heterozygous mutations are frequent and significantly associated with CP risk in pediatric patients (odds ratio [OR] = 19.1; 95% CI 2.8-160; P = 0.001 and OR = 5.5; 95% CI 1.6-19.4; P = 0.001, respectively). For the first time, we demonstrated that the c.180TT genotype of common p.Gly60Gly variant is strong, an independent CP risk factor (OR = 23; 95% CI 7.7-70; P < 0.001) with effect size comparable to p.Arg254Trp mutation. The other novel observation is that common c.493+51C>A variant, both CA and AA genotype, is significantly underrepresented in CP compared with controls (15% vs 35%; OR = 0.33; 95% CI 0.19-0.59; P < 0.001 and 2.8% vs 11%; OR = 0.24; 95% CI 0.06-0.85; P = 0.027, respectively). CONCLUSIONS Our study provides evidence that CTRC variants, including c.180TT (p.Gly60Gly) are strong CP risk factors. The c.493+51C>A variant may play a protective role against CP development.
Collapse
|
98
|
Hao L, Zeng XP, Xin L, Wang D, Pan J, Bi YW, Ji JT, Du TT, Lin JH, Zhang D, Ye B, Zou WB, Chen H, Xie T, Li BR, Zheng ZH, Wang T, Guo HL, Liao Z, Li ZS, Hu LH. Incidence of and risk factors for pancreatic cancer in chronic pancreatitis: A cohort of 1656 patients. Dig Liver Dis 2017; 49:1249-1256. [PMID: 28756974 DOI: 10.1016/j.dld.2017.07.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Risk of pancreatic cancer may increase in chronic pancreatitis patients. AIMS This study aimed to identify the incidence of and risk factors for pancreatic cancer in chronic pancreatitis patients. METHODS Chronic pancreatitis patients admitted to our center from January 2000 to December 2013 were enrolled. Cumulative rates of pancreatic cancer and survival rates were calculated. The standardized incidence ratio was calculated based on the pancreatic cancer incidence in general population of China. Risk factors for pancreatic cancer were identified. RESULTS In a total of 1656 patients, the median follow-up duration was 8.0 years. Pancreatic cancer was detected in 21 patients (1.3%). The expected number of cases of pancreatic cancer was 1.039, yielding a standardized incidence ratio of 20.22. The standardized incidence ratios for patients with a >60 pack-year smoking history were much higher (145.82). Two risk factors for pancreatic cancer were identified: age at the onset of chronic pancreatitis (hazard ratio, 1.05) and a >60 pack-year smoking history (hazard ratio, 11.83). CONCLUSION The risk of pancreatic cancer is markedly increased in chronic pancreatitis patients compared with the general population, especially in patients with an older age at onset and a >60 pack-year smoking history. The high-risk populations were suggested to be followed up closely.
Collapse
Affiliation(s)
- Lu Hao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiang-Peng Zeng
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Dan Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Jun Pan
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ya-Wei Bi
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Jun-Tao Ji
- Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ting-Ting Du
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Jin-Huan Lin
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Di Zhang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Bo Ye
- Department of surgery, Jinling Hospital, Nanjing, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Hui Chen
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ting Xie
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Bai-Rong Li
- Department of Gastroenterology, Air Force General Hospital, Beijing, China
| | - Zhao-Hong Zheng
- Department of Traditional Chinese Medicine, Jiangdu Hospital, Yangzhou, China
| | - Teng Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Hong-Lei Guo
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Liang-Hao Hu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China; Digestive Endoscopy Center, Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
99
|
Kamat N, Pai G, Mallayasamy SR, Kamath A, S R. Direct costs for nonsurgical management of Chronic Pancreatitis in a tertiary care teaching hospital. Expert Rev Pharmacoecon Outcomes Res 2017; 18:315-320. [DOI: 10.1080/14737167.2018.1386560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nagesh Kamat
- Department of Gastroenterology and Hepatology, Kasturba Medical College, Manipal University, Manipal, India
| | - Ganesh Pai
- Department of Gastroenterology and Hepatology, Kasturba Medical College, Manipal University, Manipal, India
| | - Surulivel Rajan Mallayasamy
- Department of Pharmacotherapy, UNTS College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, India
| | - Asha Kamath
- Department of Community Medicine, Kasturba Medical College, Manipal University, Manipal, India
| | - Rajasulochana S
- Department of Finance and Economics, T. A. Pai Management Institute, Manipal, India
| |
Collapse
|
100
|
The Differential Role of Human Cationic Trypsinogen ( PRSS1) p.R122H Mutation in Hereditary and Nonhereditary Chronic Pancreatitis: A Systematic Review and Meta-Analysis. Gastroenterol Res Pract 2017; 2017:9505460. [PMID: 29118810 PMCID: PMC5651130 DOI: 10.1155/2017/9505460] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/22/2017] [Accepted: 09/07/2017] [Indexed: 02/05/2023] Open
Abstract
Background Environmental factors and genetic mutations have been increasingly recognized as risk factors for chronic pancreatitis (CP). The PRSS1 p.R122H mutation was the first discovered to affect hereditary CP, with 80% penetrance. We performed here a systematic review and meta-analysis to evaluate the associations of PRSS1 p.R122H mutation with CP of diverse etiology. Methods The PubMed, EMBASE, and MEDLINE database were reviewed. The pooled odds ratio (OR) with 95% confidence intervals was used to evaluate the association of p.R122H mutation with CP. Initial analysis was conducted with all etiologies of CP, followed by a subgroup analysis for hereditary and nonhereditary CP, including alcoholic or idiopathic CP. Results A total of eight case-control studies (1733 cases and 2415 controls) were identified and included. Overall, PRSS1 p.R122H mutation was significantly associated with an increased risk of CP (OR = 4.78[1.13-20.20]). Further analysis showed p.R122H mutation strongly associated with the increased risk of hereditary CP (OR = 65.52[9.09-472.48]) but not with nonhereditary CP, both alcoholic and idiopathic CP. Conclusions Our study showing the differential role of p.R122H mutation in various etiologies of CP indicates that this complex disorder is likely influenced by multiple genetic factors as well as environmental factors.
Collapse
|