151
|
Tanhehco YC, Weisberg S, Schwartz J. Pancreatic islet autotransplantation for nonmalignant and malignant indications. Transfusion 2015; 56:761-70. [DOI: 10.1111/trf.13417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/02/2015] [Accepted: 10/08/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Yvette C. Tanhehco
- Department of Pathology and Cell Biology; Columbia University Medical Center; New York New York
| | - Stuart Weisberg
- Department of Pathology and Cell Biology; Columbia University Medical Center; New York New York
| | - Joseph Schwartz
- Department of Pathology and Cell Biology; Columbia University Medical Center; New York New York
| |
Collapse
|
152
|
Morvaridi S, Dhall D, Greene MI, Pandol SJ, Wang Q. Role of YAP and TAZ in pancreatic ductal adenocarcinoma and in stellate cells associated with cancer and chronic pancreatitis. Sci Rep 2015; 5:16759. [PMID: 26567630 PMCID: PMC4645184 DOI: 10.1038/srep16759] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/16/2015] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibrotic and inflammatory microenvironment that is formed primarily by activated, myofibroblast-like, stellate cells. Although the stellate cells are thought to contribute to tumorigenesis, metastasis and drug resistance of PDAC, the signaling events involved in activation of the stellate cells are not well defined. Functioning as transcription co-factors, Yes-associated protein (YAP) and its homolog transcriptional co-activator with PDZ-binding motif (TAZ) modulate the expression of genes involved in various aspects of cellular functions, such as proliferation and mobility. Using human tissues we show that YAP and TAZ expression is restricted to the centroacinar and ductal cells of normal pancreas, but is elevated in cancer cells. In particular, YAP and TAZ are expressed at high levels in the activated stellate cells of both chronic pancreatitis and PDAC patients as well as in the islets of Langerhans in chronic pancreatitis tissues. Of note, YAP is up regulated in both acinar and ductal cells following induction of acute and chronic pancreatitis in mice. These findings indicate that YAP and TAZ may play a critical role in modulating pancreatic tissue regeneration, neoplastic transformation, and stellate cell functions in both PDAC and pancreatitis.
Collapse
Affiliation(s)
- Susan Morvaridi
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Pancreatic Research Program; Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Deepti Dhall
- Department of Pathology and Laboratory Medicine; Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Stephen J. Pandol
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Pancreatic Research Program; Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Qiang Wang
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Pancreatic Research Program; Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
153
|
Samokhvalov AV, Rehm J, Roerecke M. Alcohol Consumption as a Risk Factor for Acute and Chronic Pancreatitis: A Systematic Review and a Series of Meta-analyses. EBioMedicine 2015; 2:1996-2002. [PMID: 26844279 PMCID: PMC4703772 DOI: 10.1016/j.ebiom.2015.11.023] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 12/11/2022] Open
Abstract
Background Pancreatitis is a highly prevalent medical condition associated with a spectrum of endocrine and exocrine pancreatic insufficiencies. While high alcohol consumption is an established risk factor for pancreatitis, its relationship with specific types of pancreatitis and a potential threshold have not been systematically examined. Methods We conducted a systematic literature search for studies on the association between alcohol consumption and pancreatitis based on PRISMA guidelines. Non-linear and linear random-effect dose–response meta-analyses using restricted cubic spline meta-regressions and categorical meta-analyses in relation to abstainers were conducted. Findings Seven studies with 157,026 participants and 3618 cases of pancreatitis were included into analyses. The dose–response relationship between average volume of alcohol consumption and risk of pancreatitis was monotonic with no evidence of non-linearity for chronic pancreatitis (CP) for both sexes (p = 0.091) and acute pancreatitis (AP) in men (p = 0.396); it was non-linear for AP in women (p = 0.008). Compared to abstention, there was a significant decrease in risk (RR = 0.76, 95%CI: 0.60–0.97) of AP in women below the threshold of 40 g/day. No such association was found in men (RR = 1.1, 95%CI: 0.69–1.74). The RR for CP at 100 g/day was 6.29 (95%CI: 3.04–13.02). Interpretation The dose–response relationships between alcohol consumption and risk of pancreatitis were monotonic for CP and AP in men, and non-linear for AP in women. Alcohol consumption below 40 g/day was associated with reduced risk of AP in women. Alcohol consumption beyond this level was increasingly detrimental for any type of pancreatitis. Funding The work was financially supported by a grant from the National Institute on Alcohol Abuse and Alcoholism (R21AA023521) to the last author. The dose–response relationships between alcohol use and different types of pancreatitis in men and women are different. The relationship was linear for chronic and acute pancreatitis in men, but non-linear for acute pancreatitis in women. There is a threshold effect for acute pancreatitis in women at the level of up to 40 g/day. The risk of pancreatitis was higher than previously thought beyond the level of 40 g of pure alcohol/day.
The article updates existing knowledge on the relationship between average alcohol consumption and the risk of pancreatitis. Specifically, there are differences between acute and chronic pancreatitis and different sexes. For women there is a threshold of 40 g of ethanol per day — below this level alcohol use is not increasing the risk of acute pancreatitis and might even be beneficial. Above this threshold alcohol use is detrimental. Beyond this threshold the risk of pancreatitis, acute and chronic, in both sexes is greater than previously thought, and increases with increases of average consumption.
Collapse
Affiliation(s)
- Andriy V Samokhvalov
- Centre for Addiction and Mental Health, Social and Epidemiological Research Department, 33 Russell Street, Toronto, Ontario M5S 2S1, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Jürgen Rehm
- Centre for Addiction and Mental Health, Social and Epidemiological Research Department, 33 Russell Street, Toronto, Ontario M5S 2S1, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Dalla Lana School of Public Health (DLSPH), University of Toronto, Toronto, Canada; Institute for Clinical Psychology and Psychotherapy, TU Dresden, Dresden, Germany; WHO Collaborating Centre on Mental Health and Addiction, Toronto, Canada
| | - Michael Roerecke
- Centre for Addiction and Mental Health, Social and Epidemiological Research Department, 33 Russell Street, Toronto, Ontario M5S 2S1, Canada; Dalla Lana School of Public Health (DLSPH), University of Toronto, Toronto, Canada; WHO Collaborating Centre on Mental Health and Addiction, Toronto, Canada
| |
Collapse
|
154
|
Nakano E, Geisz A, Masamune A, Niihori T, Hamada S, Kume K, Kakuta Y, Aoki Y, Matsubara Y, Ebert K, Ludwig M, Braun M, Groneberg DA, Shimosegawa T, Sahin-Tóth M, Witt H. Variants in pancreatic carboxypeptidase genes CPA2 and CPB1 are not associated with chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G688-94. [PMID: 26316592 PMCID: PMC4609930 DOI: 10.1152/ajpgi.00241.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/21/2015] [Indexed: 01/31/2023]
Abstract
Genetic alterations in the carboxypeptidase A1 gene (CPA1) are associated with early onset chronic pancreatitis (CP). Besides CPA1, there are two other human pancreatic carboxypeptidases (CPA2 and CPB1). Here we examined whether CPA2 and CPB1 alterations are associated with CP in Japan and Germany. All exons and flanking introns of CPA2 and CPB1 were sequenced in 477 Japanese patients with CP (234 alcoholic, 243 nonalcoholic) and in 497 German patients with nonalcoholic CP by targeted next-generation sequencing and/or Sanger sequencing. Secretion and enzymatic activity of CPA2 and CPB1 variants were determined after transfection into HEK 293T cells. We identified six nonsynonymous CPA2 variants (p.V67I, p.G166R, p.D168E, p.D173H, p.R237W, and p.G388S), eight nonsynonymous CPB1 alterations (p.S65G, p.N120S, p.D172E, p.R195H, p.D208N, p.F232L, p.A317V, and p.D364Y), and one splice-site variant (c.687+1G>T) in CPB1. Functional analysis revealed essentially complete loss of function in CPA2 variants p.R237W and p.G388S and CPB1 variants p.R110H and p.D364Y. None of the CPA2 or CPB1 variants, including those resulting in a marked loss of function, were overrepresented in patients with CP. In conclusion, CPA2 and CPB1 variants are not associated with CP.
Collapse
Affiliation(s)
- Eriko Nakano
- 1Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Andrea Geisz
- 2Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts;
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Tetsuya Niihori
- 3Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Shin Hamada
- 1Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Kiyoshi Kume
- 1Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Yoichi Kakuta
- 1Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Yoko Aoki
- 3Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Yoichi Matsubara
- 4National Research Institute for Child Health and Development, Tokyo, Japan;
| | - Karolin Ebert
- 5Pediatric Nutritional Medicine, Klinikum rechts der Isar; Else Kröner-Fresenius-Zentrum für Ernährungsmedizin & Zentralinstitut für Ernährungs-und Lebensmittelforschung, Technische Universität München, Munich, Germany; and
| | - Maren Ludwig
- 5Pediatric Nutritional Medicine, Klinikum rechts der Isar; Else Kröner-Fresenius-Zentrum für Ernährungsmedizin & Zentralinstitut für Ernährungs-und Lebensmittelforschung, Technische Universität München, Munich, Germany; and
| | - Markus Braun
- 6Institute of Occupational Medicine, Social Medicine and Environmental Medicine, Goethe-University, Frankfurt am Main, Germany
| | - David A. Groneberg
- 6Institute of Occupational Medicine, Social Medicine and Environmental Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Tooru Shimosegawa
- 1Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan;
| | - Miklós Sahin-Tóth
- 2Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts;
| | - Heiko Witt
- 5Pediatric Nutritional Medicine, Klinikum rechts der Isar; Else Kröner-Fresenius-Zentrum für Ernährungsmedizin & Zentralinstitut für Ernährungs-und Lebensmittelforschung, Technische Universität München, Munich, Germany; and
| |
Collapse
|
155
|
Rastellini C, Han S, Bhatia V, Cao Y, Liu K, Gao X, Ko TC, Greeley GH, Falzon M. Induction of chronic pancreatitis by pancreatic duct ligation activates BMP2, apelin, and PTHrP expression in mice. Am J Physiol Gastrointest Liver Physiol 2015; 309:G554-65. [PMID: 26229008 DOI: 10.1152/ajpgi.00076.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/15/2015] [Indexed: 01/31/2023]
Abstract
Chronic pancreatitis (CP) is a devastating disease with no treatments. Experimental models have been developed to reproduce the parenchyma and inflammatory responses typical of human CP. For the present study, one objective was to assess and compare the effects of pancreatic duct ligation (PDL) to those of repetitive cerulein (Cer)-induced CP in mice on pancreatic production of bone morphogenetic protein-2 (BMP2), apelin, and parathyroid hormone-related protein (PTHrP). A second objective was to determine the extent of cross talk among pancreatic BMP2, apelin, and PTHrP signaling systems. We focused on BMP2, apelin, and PTHrP since these factors regulate the inflammation-fibrosis cascade during pancreatitis. Findings showed that PDL- and Cer-induced CP resulted in significant elevations in expression and peptide/protein levels of pancreatic BMP2, apelin, and PTHrP. In vivo mouse and in vitro pancreatic cell culture experiments demonstrated that BMP2 stimulated pancreatic apelin expression whereas apelin expression was inhibited by PTHrP exposure. Apelin or BMP2 exposure inhibited PTHrP expression, and PTHrP stimulated upregulation of gremlin, an endogenous inhibitor of BMP2 activity. Transforming growth factor-β (TGF-β) stimulated PTHrP expression. Together, findings demonstrated that PDL- and Cer-induced CP resulted in increased production of the pancreatic BMP2, apelin, and PTHrP signaling systems and that significant cross talk occurred among pancreatic BMP2, apelin, and PTHrP. These results together with previous findings imply that these factors interact via a pancreatic network to regulate the inflammation-fibrosis cascade during CP. More importantly, this network communicated with TGF-β, a key effector of pancreatic pathophysiology. This novel network may be amenable to pharmacologic manipulations during CP in humans.
Collapse
Affiliation(s)
- Cristiana Rastellini
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; and
| | - Song Han
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; and
| | - Vandanajay Bhatia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas; and
| | - Yanna Cao
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ka Liu
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Xuxia Gao
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Tien C Ko
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - George H Greeley
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; and
| | - Miriam Falzon
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas; and
| |
Collapse
|
156
|
Watson P. Pancreatitis in dogs and cats: definitions and pathophysiology. J Small Anim Pract 2015; 56:3-12. [PMID: 25586802 DOI: 10.1111/jsap.12293] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 12/10/2013] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
Pancreatitis, or inflammation of the pancreas, is commonly seen in dogs and cats and presents a spectrum of disease severities from acute to chronic and mild to severe. It is usually sterile, but the causes and pathophysiology remain poorly understood. The acute end of the disease spectrum is associated with a high mortality but the potential for complete recovery of organ structure and function if the animal survives. At the other end of the spectrum, chronic pancreatitis in either species can cause refractory pain and reduce quality of life. It may also result in progressive exocrine and endocrine functional impairment. There is confusion in the veterinary literature about definitions of acute and chronic pancreatitis and there are very few studies on the pathophysiology of naturally occurring pancreatitis in dogs and cats. This article reviews histological and clinical definitions and current understanding of the pathophysiology and causes in small animals by comparison with the much more extensive literature in humans, and suggests many areas that need further study in dogs and cats.
Collapse
Affiliation(s)
- P Watson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES
| |
Collapse
|
157
|
Steele CW, Karim SA, Foth M, Rishi L, Leach JDG, Porter RJ, Nixon C, Jeffry Evans TR, Carter CR, Nibbs RJB, Sansom OJ, Morton JP. CXCR2 inhibition suppresses acute and chronic pancreatic inflammation. J Pathol 2015; 237:85-97. [PMID: 25950520 PMCID: PMC4833178 DOI: 10.1002/path.4555] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/01/2015] [Accepted: 04/28/2015] [Indexed: 12/18/2022]
Abstract
Pancreatitis is a significant clinical problem and the lack of effective therapeutic options means that treatment is often palliative rather than curative. A deeper understanding of the pathogenesis of both acute and chronic pancreatitis is necessary to develop new therapies. Pathological changes in pancreatitis are dependent on innate immune cell recruitment to the site of initial tissue damage, and on the coordination of downstream inflammatory pathways. The chemokine receptor CXCR2 drives neutrophil recruitment during inflammation, and to investigate its role in pancreatic inflammation, we induced acute and chronic pancreatitis in wild-type and Cxcr2(-/-) mice. Strikingly, Cxcr2(-/-) mice were strongly protected from tissue damage in models of acute pancreatitis, and this could be recapitulated by neutrophil depletion or by the specific deletion of Cxcr2 from myeloid cells. The pancreata of Cxcr2(-/-) mice were also substantially protected from damage during chronic pancreatitis. Neutrophil depletion was less effective in this model, suggesting that CXCR2 on non-neutrophils contributes to the development of chronic pancreatitis. Importantly, pharmacological inhibition of CXCR2 in wild-type mice replicated the protection seen in Cxcr2(-/-) mice in acute and chronic models of pancreatitis. Moreover, acute pancreatic inflammation was reversible by inhibition of CXCR2. Thus, CXCR2 is critically involved in the development of acute and chronic pancreatitis in mice, and its inhibition or loss protects against pancreatic damage. CXCR2 may therefore be a viable therapeutic target in the treatment of pancreatitis.
Collapse
MESH Headings
- Acute Disease
- Animals
- Anti-Inflammatory Agents/pharmacology
- Ceruletide
- Cytoprotection
- Disease Models, Animal
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Infiltration/drug effects
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Pancreas/drug effects
- Pancreas/immunology
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatitis/chemically induced
- Pancreatitis/genetics
- Pancreatitis/immunology
- Pancreatitis/metabolism
- Pancreatitis/pathology
- Pancreatitis/prevention & control
- Pancreatitis, Chronic/chemically induced
- Pancreatitis, Chronic/genetics
- Pancreatitis, Chronic/immunology
- Pancreatitis, Chronic/metabolism
- Pancreatitis, Chronic/pathology
- Pancreatitis, Chronic/prevention & control
- Peptides/pharmacology
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/deficiency
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/immunology
- Signal Transduction/drug effects
- Time Factors
Collapse
Affiliation(s)
- Colin W Steele
- Cancer Research UK Beatson Institute, Glasgow, UK
- Department of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | | | - Mona Foth
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Loveena Rishi
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Joshua D G Leach
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - T R Jeffry Evans
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - C Ross Carter
- Department of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | - Robert J B Nibbs
- Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
158
|
Abstract
PURPOSE OF REVIEW This report reviews recent animal model and human studies associated with inflammatory responses in acute and chronic pancreatitis. RECENT FINDINGS Animal model and limited human acute and chronic pancreatitis studies unravel the dynamic nature of the inflammatory processes and the ability of the immune cells to sense danger and environmental signals. In acute pancreatitis, such molecules include pathogen-associated molecular pattern recognition receptors such as toll-like receptors, and the more recently appreciated damage-associated molecular pattern molecules or 'alarmin' high mobility group box 1 and IL-33. In chronic pancreatitis, a recent understanding of a critical role for macrophage-pancreatic stellate cell interaction offers a potential targetable pathway that can alter fibrogenesis. Microbiome research in pancreatitis is a new field gaining interest but will require further investigation. SUMMARY Immune cell contribution to the pathogenesis of acute and chronic pancreatitis is gaining more appreciation and further understanding in immune signaling presents potential therapeutic targets that can alter disease progression.
Collapse
|
159
|
Derikx MH, Kovacs P, Scholz M, Masson E, Chen JM, Ruffert C, Lichtner P, Te Morsche RHM, Cavestro GM, Férec C, Drenth JPH, Witt H, Rosendahl J. Polymorphisms at PRSS1-PRSS2 and CLDN2-MORC4 loci associate with alcoholic and non-alcoholic chronic pancreatitis in a European replication study. Gut 2015; 64:1426-1433. [PMID: 25253127 DOI: 10.1136/gutjnl-2014-307453] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Several genetic risk factors have been identified for non-alcoholic chronic pancreatitis (NACP). A genome-wide association study reported an association of chronic pancreatitis (CP) with variants in PRSS1-PRSS2 (rs10273639; near the gene encoding cationic trypsinogen) and CLDN2-MORC4 loci (rs7057398 in RIPPLY1 and rs12688220 in MORC4). We aimed to refine these findings in a large European cohort. DESIGN We studied 3062 patients with alcohol-related CP (ACP) or NACP and 5107 controls. Also, 1559 German patients with alcohol-associated cirrhosis or alcohol dependence were included for comparison. We performed several meta-analyses to examine genotype-phenotype relationships. RESULTS Association with ACP was found for rs10273639 (OR, 0.63; 95% CI 0.55 to 0.72). ACP was also associated with variants rs7057398 and rs12688220 in men (OR, 2.26; 95% CI 1.94 to 2.63 and OR, 2.66; 95% CI 2.21 to 3.21, respectively) and in women (OR, 1.57; 95% CI 1.14 to 2.18 and OR 1.71; 95% CI 1.41 to 2.07, respectively). Similar results were obtained when German patients with ACP were compared with those with alcohol-associated cirrhosis or alcohol dependence. In the overall population of patients with NACP, association with rs10273639 was absent (OR, 0.93; 95% CI 0.79 to 1.01), whereas rs7057398 of the X chromosomal single nucleotide polymorphisms was associated with NACP in women only (OR, 1.32; 95% CI 1.15 to 1.51). CONCLUSIONS The single-nucleotide polymorphisms rs10273639 at the PRSS1-PRSS2 locus and rs7057398 and rs12688220 at the CLDN2-MORC4 locus are associated with CP and strongly associate with ACP, but only rs7057398 with NACP in female patients.
Collapse
Affiliation(s)
- Monique H Derikx
- Department of Gastroenterology and Hepatology, Radboud UMC, Nijmegen, The Netherlands
| | - Peter Kovacs
- Integrated Research and Treatment Centre (IFB) Adiposity Diseases, University of Leipzig, Leipzig, 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
| | - Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France Etablissement Français du Sang (EFS)-Bretagne, Brest, France Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, France Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Universitaire (CHU) Brest, Hôpital Morvan, Brest, France
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France Etablissement Français du Sang (EFS)-Bretagne, Brest, France Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, France Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Universitaire (CHU) Brest, Hôpital Morvan, Brest, France
| | - Claudia Ruffert
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Rene H M Te Morsche
- Department of Gastroenterology and Hepatology, Radboud UMC, Nijmegen, The Netherlands
| | - Giulia Martina Cavestro
- Unità Operativa di Gastroenterologia ed Endoscopia Digestiva, Università Vita Salute San Raffaele e IRCCS Ospedale San Raffaele, Milan, Italy
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France Etablissement Français du Sang (EFS)-Bretagne, Brest, France Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, France Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Universitaire (CHU) Brest, Hôpital Morvan, Brest, France
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud UMC, Nijmegen, The Netherlands
| | - Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL) & Paediatric Nutritional Medicine, Technische Universität München (TUM), Munich, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
160
|
Yu T, Qing Q, Deng N, Min XH, Zhao LN, Li JY, Xia ZS, Chen QK. CXCR4 positive cell-derived Pdx1-high/Shh-low cells originated from embryonic stem cells improve the repair of pancreatic injury in mice. Cell Biol Int 2015; 39:995-1006. [PMID: 25820869 DOI: 10.1002/cbin.10470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/14/2015] [Accepted: 03/18/2015] [Indexed: 01/05/2023]
Abstract
Treatments for pancreatic injuries have been significantly improved recently, but full recovery of pancreatic function remains difficult. Embryonic stem cells have great potentialities for self-renewal and multiple differentiations. In this study, we explored an approach to induce the differentiation of pancreatic progenitor cells from embryonic stem cells in vitro. Male mouse embryonic stem cells were cultured by the hanging-drop method to form embryoid bodies. The definitive endoderm marked by CXCR4 in embryoid bodies was sorted by magnetic activated cell sorting and subsequently administrated with b-FGF, exendin-4, and cyclopamine to induce the differentiation of putative pancreatic progenitor cells, which was monitored by Pdx1, and Shh expressions. The putative pancreatic progenitor cells were transplanted into female BALB/c mice with pancreatitis induced by L-Arginine. Male donor cells were located by detecting sex-determining region of Y-chromosome DNA. Definitive endoderm cells (CXCR4(+) cells) were sorted from 5-day embryoid bodies. After 3-day administration with b-FGF, exendin-4, and cyclopamine, Pdx1-high/Shh-low cells were differentiated from CXCR4(+) cells. These cells developed into more amylase-secreted cells in vitro and could specifically reside in the damaged pancreas acinar area in mice with acute pancreatitis to enhance the regeneration. The putative pancreatic progenitor cells (Pdx1-high/Shh-low cells) derived from mouse embryonic stem cells through the administration of b-FGF, exendin-4, and cyclopamine on the CXCR4(+) cells in vitro could improve the regeneration of injured pancreatic acini in vivo.
Collapse
Affiliation(s)
- Tao Yu
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qing Qing
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Na Deng
- Department of Gastroenterology, Yuebei People's Hospital, Shaoguan, Guangdong, People's Republic of China
| | - Xiao-Hui Min
- Department of Infectious Disease, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Li-Na Zhao
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jie-Yao Li
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhong-Sheng Xia
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qi-Kui Chen
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
161
|
|
162
|
Vallance AE, Wilson CH, Dennison A, Manas DM, White SA. Total pancreatectomy and islet autotransplantation for chronic pancreatitis. Hippokratia 2015. [DOI: 10.1002/14651858.cd011799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Abigail E Vallance
- The Freeman Hospital; Institute of Transplantation; Freeman Road High Heaton Newcastle upon Tyne UK NE7 7DN
| | - Colin H Wilson
- The Freeman Hospital; Institute of Transplantation; Freeman Road High Heaton Newcastle upon Tyne UK NE7 7DN
| | - Ashley Dennison
- Leicester General Hospital; Department of Hepatobiliary Surgery; Gwendolen Road Leicester UK LE5 4PW
| | - Derek M Manas
- The Freeman Hospital; Institute of Transplantation; Freeman Road High Heaton Newcastle upon Tyne UK NE7 7DN
| | - Steven A White
- Institute of Cellular Medicine, Newcastle University; Newcastle upon Tyne UK NE2 4HH
| |
Collapse
|
163
|
Hillson R. Diabetes and alcohol. PRACTICAL DIABETES 2015. [DOI: 10.1002/pdi.1955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
164
|
Hou XJ, Jin ZD, Jiang F, Zhu JW, Li ZS. Expression of Smad7 and Smad ubiquitin regulatory factor 2 in a rat model of chronic pancreatitis. J Dig Dis 2015; 16:408-415. [PMID: 25943897 DOI: 10.1111/1751-2980.12253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To quantify the expressions of Smad7 and Smad ubiquitin regulatory factor 2 (Smurf2) in the pancreas in rats with chronic pancreatitis (CP). METHODS A total of 16 male Wistar rats were randomly divided into the control group and the CP group, with 8 rats in each group. CP was induced in vivo with dibutyltin dichloride (DBTC). Four weeks after DBTC administration, histological assessment and the measurement of hydroxyproline content in the pancreatic tissues were performed to assess the inflammation and fibrosis of the pancreas. Immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR) for transforming growth factor (TGF)-β1 and α-smooth muscle actin (α-SMA) were applied to assess activated pancreatic stellate cells (PSC) and TGF-β1 expression. Smad7 and Smurf2 expressions in the pancreas were measured using Western blot and RT-PCR. RESULTS Typical histopathological characteristics of DBTC-induced CP in the rats with extensively activated PSC. Compared with the control group, the expressions of TGF-β1, α-SMA and hydroxyproline content in the pancreatic tissues in the CP group were significantly increased. Meanwhile, the mRNA and protein expressions of Smad7 and Smurf2 were significant increased in the fibrotic pancreas, in which the expressions of Smad7 proteins showed an obvious reduction compared with controls. CONCLUSION The dysregulation of Smad7 and Smurf2 may be associated with the pathogenesis of pancreatic fibrosis through the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Xiao Jia Hou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhen Dong Jin
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fei Jiang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jian Wei Zhu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhao Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
165
|
Antioxidant therapy for pain reduction in patients with chronic pancreatitis: a systematic review and meta-analysis. Pancreas 2015; 44:812-8. [PMID: 25882696 DOI: 10.1097/mpa.0000000000000327] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Oxidative stress is strongly implicated in the pathogenesis of chronic pancreatitis (CP); however, clinical studies evaluating antioxidant therapy (AOT) in the management of CP pain have yielded conflicting results. The aim of this meta-analysis was to evaluate the efficacy of AOT in pain reduction in CP patients. METHODS Two authors independently conducted a comprehensive literature search from inception to December 2013. Relative risk (RR) and 95% confidence intervals (CIs) were calculated using the Mantel-Haenszel and DerSimonian-Laird methods. RESULTS Eight studies involving 446 patients (234 in AOT and 212 in control group) were included. Overall, CP patients who received AOT had significant reduction in pain compared with the control group (RR, 0.73; 95% CI, 0.58-0.91; P = 0.006). In the subgroup analysis, studies with predominantly alcoholic CP patients (RR, 0.60; 95% CI, 0.47-0.77; P < 0.0001) and studies with mean age of patients 42 years or older (RR, 0.66; 95% CI, 0.49-0.89; P = 0.006) showed significant benefit of AOT over placebo. CONCLUSIONS This is the first meta-analysis to summarize all the available evidence and show benefit of AOT in pain reduction in CP patients. There is a pressing need for well-designed larger studies with longer follow-up to better define the patient and disease factors favoring response, the optimal formulation, and duration of AOT.
Collapse
|
166
|
Falzon M, Bhatia V. Role of Parathyroid Hormone-Related Protein Signaling in Chronic Pancreatitis. Cancers (Basel) 2015; 7:1091-108. [PMID: 26095761 PMCID: PMC4491701 DOI: 10.3390/cancers7020826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/05/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic pancreatitis (CP), a progressive inflammatory disease where acini are destroyed and replaced by fibrous tissue, increases the risk for pancreatic cancer. Risk factors include alcohol, smoking, and obesity. The effects of these risk factors are exacerbated in patients with mutations in genes that predispose to CP. The different environmental and genetic factors produce the same clinical phenotype; once CP develops, disease course is the same regardless of etiology. Critical questions still need to be answered to understand what modifies predisposition to develop CP in persons exposed to risk factors. We postulate that risk factors modulate endogenous pathways, with parathyroid hormone-related protein (PTHrP) signaling being one such pathway. In support, PTHrP levels are elevated in mice treated with alcohol, and in mouse models of cerulein- and pancreatic duct ligation-induced CP. Disrupting the Pthrp gene in acinar cells exerts protective effects (decreased edema, histological damage, amylase and cytokine release, and fibrosis) in these CP models. PTHrP levels are elevated in human CP. Currently, CP care lacks specific pharmacological interventions. Targeting PTHrP signaling may present a novel therapeutic strategy that inhibits pancreatic inflammation and fibrosis, especially since the risk of developing pancreatic cancer is strongly associated with duration of chronic inflammation.
Collapse
Affiliation(s)
- Miriam Falzon
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Vandanajay Bhatia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
167
|
Xue J, Sharma V, Hsieh MH, Chawla A, Murali R, Pandol SJ, Habtezion A. Alternatively activated macrophages promote pancreatic fibrosis in chronic pancreatitis. Nat Commun 2015; 6:7158. [PMID: 25981357 PMCID: PMC4632846 DOI: 10.1038/ncomms8158] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/10/2015] [Indexed: 12/17/2022] Open
Abstract
Chronic pancreatitis (CP) is a progressive and irreversible inflammatory and fibrotic disease with no cure. Unlike acute pancreatitis (AP), we find that alternatively activated macrophages (AAMs) are dominant in mouse and human CP. AAMs are dependent on interleukin (IL)-4 and IL-13 signalling, and we show that mice lacking IL-4Rα, myeloid-specific IL-4Rα and IL-4/IL-13 were less susceptible to pancreatic fibrosis. Furthermore, we demonstrate that mouse and human pancreatic stellate cells (PSCs) are a source of IL-4/IL-13. Notably, we show that pharmacologic inhibition of IL-4/IL-13 in human ex vivo studies as well as in established mouse CP decreases pancreatic AAMs and fibrosis. We identify a critical role for macrophages in pancreatic fibrosis and in turn PSCs as important inducers of macrophage-alternative activation. Our study challenges and identifies pathways involved in crosstalk between macrophages and PSCs that can be targeted to reverse or halt pancreatic fibrosis progression.
Collapse
Affiliation(s)
- Jing Xue
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Vishal Sharma
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Michael H Hsieh
- Department of Urology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA
| | - Ajay Chawla
- Department of Physiology and Medicine, Cardiovascular Research Institute, University of California, San Francisco, California 94158, USA
| | - Ramachandran Murali
- Research division of immunology, Department of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Stephen J Pandol
- Research division of immunology, Department of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
168
|
Derikx MHM, Geisz A, Kereszturi É, Sahin-Tóth M. Functional significance of SPINK1 promoter variants in chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2015; 308:G779-84. [PMID: 25792561 PMCID: PMC4421017 DOI: 10.1152/ajpgi.00022.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/11/2015] [Indexed: 01/31/2023]
Abstract
Chronic pancreatitis is a progressive inflammatory disorder of the pancreas, which often develops as a result of genetic predisposition. Some of the most frequently identified risk factors affect the serine protease inhibitor Kazal type 1 (SPINK1) gene, which encodes a trypsin inhibitor responsible for protecting the pancreas from premature trypsinogen activation. Recent genetic and functional studies indicated that promoter variants in the SPINK1 gene might contribute to disease risk in carriers. Here, we investigated the functional effects of 17 SPINK1 promoter variants using luciferase reporter gene expression assay in four different cell lines, including three pancreatic acinar cell lines (rat AR42J with or without dexamethasone-induced differentiation and mouse 266-6) and human embryonic kidney 293T cells. We found that most variants caused relatively small changes in promoter activity. Surprisingly, however, we observed significant variations in the effects of the promoter variants in the different cell lines. Only four variants exhibited consistently reduced promoter activity in all acinar cell lines, confirming previous reports that variants c.-108G>T, c.-142T>C, and c.-147A>G are risk factors for chronic pancreatitis and identifying c.-52G>T as a novel risk variant. In contrast, variant c.-215G>A, which is linked with the disease-associated splice-site mutation c.194 + 2T>C, caused increased promoter activity, which may mitigate the overall effect of the pathogenic haplotype. Our study lends further support to the notion that sequence evaluation of the SPINK1 promoter region in patients with chronic pancreatitis is justified as part of the etiological investigation.
Collapse
Affiliation(s)
- Monique H. M. Derikx
- 1Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts; ,2Department of Gastroenterology and Hepatology, Radboud UMC, Nijmegen, The Netherlands
| | - Andrea Geisz
- 1Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts;
| | - Éva Kereszturi
- 1Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts;
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts;
| |
Collapse
|
169
|
Nakano E, Masamune A, Niihori T, Kume K, Hamada S, Aoki Y, Matsubara Y, Shimosegawa T. Targeted next-generation sequencing effectively analyzed the cystic fibrosis transmembrane conductance regulator gene in pancreatitis. Dig Dis Sci 2015; 60:1297-307. [PMID: 25492507 DOI: 10.1007/s10620-014-3476-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/28/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The cystic fibrosis transmembrane conductance regulator (CFTR) gene, responsible for the development of cystic fibrosis, is known as a pancreatitis susceptibility gene. Direct DNA sequencing of PCR-amplified CFTR gene segments is a first-line method to detect unknown mutations, but it is a tedious and labor-intensive endeavor given the large size of the gene (27 exons, 1,480 amino acids). Next-generation sequencing (NGS) is becoming standardized, reducing the cost of DNA sequencing, and enabling the generation of millions of reads per run. We here report a comprehensive analysis of CFTR variants in Japanese patients with chronic pancreatitis using NGS coupling with target capture. METHODS Exon sequences of the CFTR gene from 193 patients with chronic pancreatitis (121 idiopathic, 46 alcoholic, 17 hereditary, and nine familial) were captured by HaloPlex target enrichment technology, followed by NGS. RESULTS The sequencing data covered 91.6 % of the coding regions of the CFTR gene by ≥ 20 reads with a mean read depth of 449. We could identify 12 non-synonymous variants including three novel ones [c.A1231G (p.K411E), c.1753G>T (p.E585X) and c.2869delC (p.L957fs)] and seven synonymous variants including three novel ones in the exonic regions. The frequencies of the c.4056G>C (p.Q1352H) and the c.3468G>T (p.L1156F) variants were higher in patients with chronic pancreatitis than those in controls. CONCLUSIONS Target sequence capture combined with NGS is an effective method for the analysis of pancreatitis susceptibility genes.
Collapse
Affiliation(s)
- Eriko Nakano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
170
|
Cystic fibrosis transmembrane conductance regulator gene variants are associated with autoimmune pancreatitis and slow response to steroid treatment. J Cyst Fibros 2015; 14:661-7. [PMID: 25869325 DOI: 10.1016/j.jcf.2015.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/22/2015] [Accepted: 03/22/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Autoimmune pancreatitis (AIP) is a distinct type of chronic pancreatitis. To date, the association of CFTR gene variants with AIP has not been studied. METHODS The entire coding and intronic regions of the CFTR gene were examined using next-generation sequencing in 89 AIP patients. Clinical features, including imaging, histology, serology, steroid treatment response and extra-pancreatic involvement, were compared between AIP patients with and without CFTR gene variants. RESULTS A total of 28.1% (25/89) of the AIP patients carried 26 CFTR variants, including nine with I556V, seven with 5T, four with S42F, two with I125T, and one each with R31C, R553X, S895N, and G1069R. The presence of CFTR variants and age was independent predictors of the response to steroid treatment, as shown by multivariate analysis. CONCLUSIONS CFTR variants are associated with AIP. Because AIP patients with CFTR variants show slower and reduced steroid treatment responses, different treatments should be considered in AIP patients with CFTR variants.
Collapse
|
171
|
Jiménez-Zamora V, Bellacetín-Figueroa O, García-Guerrero VA, Zarate-Guzmán ÁM, Corral-Medina A, Bernal-Sahagún F, Valdés-Lías R, Espino-Cortés H, Vasquéz-Bustamante F. Rendimiento diagnóstico del ultrasonido endoscópico en la pancreatitis crónica. ENDOSCOPIA 2015. [DOI: 10.1016/j.endomx.2015.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
172
|
Ahmed Ali U, Pahlplatz JM, Nealon WH, van Goor H, Gooszen HG, Boermeester MA, Cochrane Upper GI and Pancreatic Diseases Group. Endoscopic or surgical intervention for painful obstructive chronic pancreatitis. Cochrane Database Syst Rev 2015; 2015:CD007884. [PMID: 25790326 PMCID: PMC10710281 DOI: 10.1002/14651858.cd007884.pub3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Endoscopy and surgery are the treatment modalities of choice for patients with chronic pancreatitis and dilated pancreatic duct (obstructive chronic pancreatitis). Physicians face, without clear consensus, the choice between endoscopy or surgery for this group of patients. OBJECTIVES To assess and compare the effects and complications of surgical and endoscopic interventions in the management of pain for obstructive chronic pancreatitis. SEARCH METHODS We searched the following databases in The Cochrane Library: CENTRAL (2014, Issue 2), the Cochrane Database of Systematic Reviews (2014, Issue 2), and DARE (2014, Issue 2). We also searched the following databases up to 25 March 2014: MEDLINE (from 1950), Embase (from 1980), and the Conference Proceedings Citation Index - Science (CPCI-S) (from 1990). We performed a cross-reference search. Two review authors independently performed the selection of trials. SELECTION CRITERIA All randomised controlled trials (RCTs) of endoscopic or surgical interventions in obstructive chronic pancreatitis. We included trials comparing endoscopic versus surgical interventions as well as trials comparing either endoscopic or surgical interventions to conservative treatment (i.e. non-invasive treatment modalities). We included relevant trials irrespective of blinding, the number of participants randomised, and the language of the article. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by The Cochrane Collaboration. Two authors independently extracted data from the articles. We evaluated the methodological quality of the included trials and requested additional information from study authors in the case of missing data. MAIN RESULTS We identified three eligible trials. Two trials compared endoscopic intervention with surgical intervention and included a total of 111 participants: 55 in the endoscopic group and 56 in the surgical group. Compared with the endoscopic group, the surgical group had a higher proportion of participants with pain relief, both at middle/long-term follow-up (two to five years: risk ratio (RR) 1.62, 95% confidence interval (CI) 1.22 to 2.15) and long-term follow-up (≥ five years, RR 1.56, 95% CI 1.18 to 2.05). Surgical intervention resulted in improved quality of life and improved preservation of exocrine pancreatic function at middle/long-term follow-up (two to five years), but not at long-term follow-up (≥ 5 years). No differences were found in terms of major post-interventional complications or mortality, although the number of participants did not allow for this to be reliably evaluated. One trial, including 32 participants, compared surgical intervention with conservative treatment: 17 in the surgical group and 15 in the conservative group. The trial showed that surgical intervention resulted in a higher percentage of participants with pain relief and better preservation of pancreatic function. The trial had methodological limitations, and the number of participants was relatively small. AUTHORS' CONCLUSIONS For patients with obstructive chronic pancreatitis and dilated pancreatic duct, this review shows that surgery is superior to endoscopy in terms of pain relief. Morbidity and mortality seem not to differ between the two intervention modalities, but the small trials identified do not provide sufficient power to detect the small differences expected in this outcome.Regarding the comparison of surgical intervention versus conservative treatment, this review has shown that surgical intervention in an early stage of chronic pancreatitis is a promising approach in terms of pain relief and pancreatic function. Other trials need to confirm these results because of the methodological limitations and limited number of participants assessed in the present evidence.
Collapse
Affiliation(s)
- Usama Ahmed Ali
- University Medical Center UtrechtDepartment of SurgeryHeidelberglaan 100P.O. Box 85500UtrechtUtrechtNetherlands3508 GA
| | - Johanna M Pahlplatz
- University Medical Center UtrechtDepartment of SurgeryHeidelberglaan 100P.O. Box 85500UtrechtUtrechtNetherlands3508 GA
| | - Wiliam H Nealon
- Vanderbilt University Medical CenterDepartment of SurgeryD‐4314 Medical Center North1161 21st Avenue SouthNashvilleTennesseeUSA37232‐2730
| | - Harry van Goor
- Radboud University Nijmegen Medical CentreDepartment of SurgeryPO Box 9101NijmegenNetherlands6500 HB
| | - Hein G Gooszen
- Radboud University Nijmegen Medical CenterCentre of Evidence‐based SurgeryPO Box 9101Huispost 630, route 631NijmegenNetherlands6500 HB
| | - Marja A Boermeester
- University of AmsterdamDepartment of Surgery, Academic Medical CenterG4‐132.1Meibergdreef 9, Postbus 22660AmsterdamNoord‐HollandNetherlands1100 DD Amsterdam
| | | |
Collapse
|
173
|
Fjeld K, Weiss FU, Lasher D, Rosendahl J, Chen JM, Johansson BB, Kirsten H, Ruffert C, Masson E, Steine SJ, Bugert P, Cnop M, Grützmann R, Mayerle J, Mössner J, Ringdal M, Schulz HU, Sendler M, Simon P, Sztromwasser P, Torsvik J, Scholz M, Tjora E, Férec C, Witt H, Lerch MM, Njølstad PR, Johansson S, Molven A. A recombined allele of the lipase gene CEL and its pseudogene CELP confers susceptibility to chronic pancreatitis. Nat Genet 2015; 47:518-522. [PMID: 25774637 PMCID: PMC5321495 DOI: 10.1038/ng.3249] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/19/2015] [Indexed: 12/13/2022]
Abstract
Carboxyl-ester lipase is a digestive pancreatic enzyme encoded by the highly polymorphic CEL gene1. Mutations in CEL cause maturity-onset diabetes of the young (MODY) with pancreatic exocrine dysfunction2. Here we identified a hybrid allele (CEL-HYB), originating from a crossover between CEL and its neighboring pseudogene CELP. In a discovery cohort of familial chronic pancreatitis cases, the carrier frequency of CEL-HYB was 14.1% (10/71) compared with 1.0% (5/478) in controls (odds ratio [OR] = 15.5, 95% confidence interval [CI] = 5.1-46.9, P = 1.3 × 10−6). Three replication studies in non-alcoholic chronic pancreatitis cohorts identified CEL-HYB in a total of 3.7% (42/1,122) cases and 0.7% (30/4,152) controls (OR = 5.2, 95% CI = 3.2-8.5, P = 1.2 × 10−11; formal meta-analysis). The allele was also enriched in alcoholic chronic pancreatitis. Expression of CEL-HYB in cellular models revealed reduced lipolytic activity, impaired secretion, prominent intracellular accumulation and induced autophagy. The hybrid variant of CEL is the first chronic pancreatitis gene identified outside the protease/antiprotease system of pancreatic acinar cells.
Collapse
Affiliation(s)
- Karianne Fjeld
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Frank Ulrich Weiss
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Denise Lasher
- Pediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany.,Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Freising, Germany
| | - Jonas Rosendahl
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France.,Etablissement Français du Sang (EFS)-Bretagne, Brest, France.,Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France
| | - Bente B Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Holger Kirsten
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,LIFE-Leipzig Research Center for Civilization Diseases, Universität Leipzig, Leipzig, Germany.,Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Universität Leipzig, Leipzig, Germany
| | - Claudia Ruffert
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany.,Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany.,Integrated Research and Treatment Centre (IFB) Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France.,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Universitaire (CHU) Brest, Hôpital Morvan, Brest, France
| | - Solrun J Steine
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg-Hessen, Mannheim, Germany
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Brussels, Belgium
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Dresden, Dresden, Germany
| | - Julia Mayerle
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Joachim Mössner
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Leipzig, Germany
| | - Monika Ringdal
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Matthias Sendler
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Peter Simon
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Paweł Sztromwasser
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway.,Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Janniche Torsvik
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Markus Scholz
- LIFE-Leipzig Research Center for Civilization Diseases, Universität Leipzig, Leipzig, Germany.,Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Universität Leipzig, Leipzig, Germany
| | - Erling Tjora
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France.,Etablissement Français du Sang (EFS)-Bretagne, Brest, France.,Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France.,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Universitaire (CHU) Brest, Hôpital Morvan, Brest, France
| | - Heiko Witt
- Pediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany.,Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Freising, Germany
| | - Markus M Lerch
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Stefan Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anders Molven
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
174
|
Kanika G, Khan S, Jena G. Sodium Butyrate Ameliorates L-Arginine-Induced Pancreatitis and Associated Fibrosis in Wistar Rat: Role of Inflammation and Nitrosative Stress. J Biochem Mol Toxicol 2015; 29:349-59. [PMID: 25774002 DOI: 10.1002/jbt.21698] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/30/2015] [Accepted: 02/04/2015] [Indexed: 01/29/2023]
Abstract
Several reports indicated that histone deacetylases (HDACs) play a crucial role in inflammation and fibrogenesis. Sodium butyrate (SB) is a short-chain fatty acid having HDAC inhibition potential. The present study aimed to evaluate the protective effect of SB against L-arginine (L-Arg)-induced pancreatic fibrosis in Wistar rats. Pancreatic fibrosis was induced by twice intraperitoneal (i.p.) injections of 20% L-Arg (250 mg/100 g) at 2-h interval on day 1, 4, 7, and 10, whereas SB (800 mg/kg/day) was administrated for 10 days. At the end of the study, biochemical estimations, histological alterations, DNA damage, and the expression of various proteins were evaluated. Posttreatment of SB decreased L-Arg-induced oxidative and nitrosative stress, DNA damage, histological alterations, and fibrosis. Interestingly, posttreatment of SB significantly decreased the expression of α-smooth muscle actin, interleukin-1β, inducible nitric oxide synthase, and 3-nitrotyrosine. The present study demonstrated that posttreatment of SB alleviates L-Arg-induced pancreatic damage and fibrosis in rat.
Collapse
Affiliation(s)
- Gayathri Kanika
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, 160062, India
| | - Sabbir Khan
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, 160062, India. ,
| |
Collapse
|
175
|
Abstract
The myriad of presentations of pancreatitis can cause confusion and controversy among clinicians affecting the diagnosis, treatment, and research of patients with these disorders. Although the disease is best thought of as a spectrum with classic presentations, the underlying pathophysiologic reasons for the differences in manifestations remains unknown. In this issue of the Journal, LaRusch and colleagues provide an elegant study combining epidemiology and molecular biology to explain why some patients with pancreatitis develop fibrosis chronic pancreatitis. The implications of the findings add to the growing request to support large multidisciplinary, combined genetic, and epidemiologic studies in pancreatic disease.
Collapse
Affiliation(s)
- Scott Tenner
- Department of Medicine, State University of New York Health Sciences Center, Brooklyn, New York, USA
| |
Collapse
|
176
|
Prognostic value of purinergic P2X7 receptor expression in patients with hepatocellular carcinoma after curative resection. Tumour Biol 2015; 36:5039-49. [PMID: 25722111 DOI: 10.1007/s13277-015-3155-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/26/2015] [Indexed: 01/24/2023] Open
Abstract
The family of type 2 purinergic (P2) receptors, especially P2X7, is responsible for the direct tumor-killing functions of extracellular adenosine triphosphate (ATP), but the precise role of P2X7 in the progression of hepatocellular carcinoma (HCC) remains elusive. This study aims to evaluate prognostic value of P2X7 expression in HCC patients after surgical resection. Expression of P2X7 was assessed by immunohistochemistry in tissue microarrays containing paired tumor and peritumoral liver tissues from 273 patients with HCC who had undergone hepatectomy between 2006 and 2007. Prognostic value of P2X7 expression and clinical outcomes were evaluated. Peritumoral P2X7 expression was significantly higher than intratumoral P2X7 expression. No significant prognostic difference was observed for overall survival for intratumoral P2X7 density, whereas peritumoral P2X7 density indicates unfavorable overall survival in training set and BCLC stage 0-A subset. Besides, peritumoral P2X7 density, which correlated with tumor size, venous invasion, and BCLC stage, was identified as an independent poor prognosticator for overall survival and recurrence-free survival. The association was further validated in validation set. Peritumoral P2X7 is a potential unfavorable prognosticator for overall survival and recurrence free survival in HCC patients after surgical resection. Further external validation and functional analysis should be pursued to evaluate its potential prognostic value and therapeutic significance for HCC patients.
Collapse
|
177
|
Mrazek AA, Porro LJ, Bhatia V, Falzon M, Spratt H, Zhou J, Chao C, Hellmich MR. Apigenin inhibits pancreatic stellate cell activity in pancreatitis. J Surg Res 2015; 196:8-16. [PMID: 25799526 DOI: 10.1016/j.jss.2015.02.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/04/2015] [Accepted: 02/13/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic pancreatitis (CP) is characterized by recurrent pancreatic injury, resulting in inflammation, necrosis, and fibrosis. There are currently no drugs limiting pancreatic fibrosis associated with CP, and there is a definite need to fill this void in patient care. MATERIALS AND METHODS Pancreatitis was induced in C57/BL6 mice using supraphysiologic doses of cerulein, and apigenin treatment (once daily, 50 μg per mouse by oral gavage) was initiated 1 wk into the recurrent acute pancreatitis (RAP) protocol. Pancreata were harvested after 4 wk of RAP. Immunostaining with fibronectin antibody was used to quantify the extent of pancreatic fibrosis. To assess how apigenin may decrease organ fibrosis, we evaluated the effect of apigenin on the proliferation and apoptosis of human pancreatic stellate cells (PSCs) in vitro. Finally, we assessed apigenin's effect on the gene expression in PSCs stimulated with parathyroid hormone-related protein, a profibrotic and proinflammatory mediator of pancreatitis, using reverse transcription-polymerase chain reaction. RESULTS After 4 wk of RAP, apigenin significantly reduced the fibrotic response to injury while preserving acinar units. Apigenin inhibited viability and induced apoptosis of PSCs in a time- and dose-dependent manner. Finally, apigenin reduced parathyroid hormone-related protein-stimulated increases in the PSC messenger RNA expression levels of extracellular matrix proteins collagen 1A1 and fibronectin, proliferating cell nuclear antigen, transforming growth factor-beta, and interleukin-6. CONCLUSIONS These in vivo and in vitro studies provide novel insights regarding apigenin's mechanism(s) of action in reducing the severity of RAP. Additional preclinical testing of apigenin analogs is warranted to develop a therapeutic agent for patients at risk for CP.
Collapse
Affiliation(s)
- Amy A Mrazek
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Laura J Porro
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Vandanajay Bhatia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Miriam Falzon
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Heidi Spratt
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, Texas
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Celia Chao
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
178
|
Shi Y, Glaser KJ, Sudhakar VK, Ben-Abraham EI, Ehman RL. Feasibility of using 3D MR elastography to determine pancreatic stiffness in healthy volunteers. J Magn Reson Imaging 2015; 41:369-75. [PMID: 24497052 PMCID: PMC4122650 DOI: 10.1002/jmri.24572] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/02/2014] [Indexed: 01/02/2023] Open
Abstract
PURPOSE To evaluate the feasibility of using three-dimensional (3D) MR elastography (MRE) to determine the stiffness of the pancreas in healthy volunteers. MATERIALS AND METHODS Twenty healthy volunteers underwent 1.5 Tesla MRE exams using an accelerated echo planar imaging (EPI) pulse sequence with low-frequency vibrations (40 and 60 Hz). Stiffness was calculated with a 3D direct inversion algorithm. The mean shear stiffness in five pancreatic subregions (uncinate, head, neck, body, and tail) and the corresponding liver stiffness were calculated. The intrasubject coefficient of variation (CV) was calculated as a measure of the reproducibility for each volunteer. RESULTS The mean shear stiffness (average of values obtained in different pancreatic subregions) was (1.15 ± 0.17) kPa at 40 Hz, and (2.09 ± 0.33) kPa at 60 Hz. The corresponding liver stiffness was higher than the pancreas stiffness at 40 Hz ([1.60 ± 0.21] kPa, mean pancreas-to-liver stiffness ratio: 0.72), but similar at 60Hz ([2.12 ± 0.23) kPa, mean ratio: 0.95). The mean intrasubject CV for each pancreatic subregion was lower at 40 Hz than 60 Hz (P < 0.05 for all subregions, range: 11.9-15.7% at 40 Hz and 16.5-19.6% at 60 Hz). CONCLUSION The 3D pancreatic MRE can provide promising and reproducible stiffness measurements throughout the pancreas, with more consistent data acquired at 40 Hz.
Collapse
Affiliation(s)
- Yu Shi
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
- Department of Radiology, Shengjing Hospital, China Medical University, No.36, Sanhao Street, Heping District, Shenyang, 110004, P.R. China
| | - Kevin J. Glaser
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | | | | | - Richard L. Ehman
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| |
Collapse
|
179
|
The Common Chymotrypsinogen C (CTRC) Variant G60G (C.180T) Increases Risk of Chronic Pancreatitis But Not Recurrent Acute Pancreatitis in a North American Population. Clin Transl Gastroenterol 2015; 6:e68. [PMID: 25569187 PMCID: PMC4418406 DOI: 10.1038/ctg.2014.13] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/30/2014] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES: Recurrent acute pancreatitis (RAP) is a complex inflammatory disorder that may progress to fibrosis and other irreversible features recognized as chronic pancreatitis (CP). Chymotrypsinogen C (CTRC) protects the pancreas by degrading prematurely activated trypsinogen. Rare mutations are associated with CP in Europe and Asia. We evaluated the occurrence of CTRC variants in subjects with RAP, CP, and controls from the North American Pancreatitis Study II cohort. METHODS: CP (n=694), RAP (n=448), and controls (n=1017) of European ancestry were evaluated. Subgroup analysis included CFTR and SPINK1 variants, alcohol, and smoking. RESULTS: We identified previously reported rare pathogenic CTRC A73T, R254W, and K247_R254del variants, intronic variants, and G60G (c.180 C>T; rs497078). Compared with controls (minor allele frequency (MAF)=10.8%), c.180T was associated with CP (MAF=16.8%, P<0.00001) but not RAP (MAF=11.9% P=NS). Trend test indicated co-dominant risk for CP (CT odds ratio (OR)=1.36, 95% confidence interval (CI)=1.13–1.64, P=0.0014; TT OR=3.98, 95% CI=2.10–7.56, P<0.0001). The T allele was significantly more frequent with concurrent pathogenic CFTR variants and/or SPINK1 N34S (combined 22.9% vs. 16.1%, OR 1.92, 95% C.I. 1.26–2.94, P=0.0023) and with alcoholic vs. non-alcoholic CP etiologies (20.8% vs. 12.4%, OR=1.9, 95% CI=1.30–2.79, P=0.0009). Alcohol and smoking generally occurred together, but the frequency of CTRC c.180 T in CP, but not RAP, was higher among never drinkers–ever smokers (22.2%) than ever drinker–never smokers (10.8%), suggesting that smoking rather than alcohol may be the driving factor in this association. CONCLUSIONS: The common CTRC variant c.180T acts as disease modifier that promotes progression from RAP to CP, especially in patients with CFTR or SPINK1 variants, alcohol, or smoking.
Collapse
|
180
|
Feng WM, Guo HH, Xue T, Wang X, Tang CW, Ying B, Gong H, Cui G. Anti-inflammation and anti-fibrosis with PEGylated, apigenin loaded PLGA nanoparticles in chronic pancreatitis disease. RSC Adv 2015. [DOI: 10.1039/c5ra17686g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In this work, apigenin, a drug that can inhibit pancreatic stellate cell fibrosis, is loaded into PEGylated PLGA nanoparticles to treat the inflammation and fibrosis associated with chronic pancreatitis (CP).
Collapse
Affiliation(s)
- Wen-ming Feng
- Department of General Surgery
- The first affiliated hospital
- Huzhou University
- China
| | - Hui-hui Guo
- Research Center
- The first affiliated hospital
- Huzhou University
- China
| | - Tao Xue
- Research Center
- The first affiliated hospital
- Huzhou University
- China
| | - Xiang Wang
- Research Center
- The first affiliated hospital
- Huzhou University
- China
| | - Cheng-wu Tang
- Department of General Surgery
- The first affiliated hospital
- Huzhou University
- China
| | - Bao Ying
- Department of General Surgery
- The first affiliated hospital
- Huzhou University
- China
| | - Hui Gong
- Research Center
- The first affiliated hospital
- Huzhou University
- China
| | - Ge Cui
- Department of Pathology
- The first affiliated hospital
- Huzhou University
- Huzhou
- China
| |
Collapse
|
181
|
Ahmed Ali U, Issa Y, van Goor H, van Eijck CH, Nieuwenhuijs VB, Keulemans Y, Fockens P, Busch OR, Drenth JP, Dejong CH, van Dullemen HM, van Hooft JE, Siersema PD, Spanier BWM, Poley JW, Poen AC, Timmer R, Seerden T, Tan AC, Thijs WJ, Witteman BJM, Romkens TEH, Roeterdink AJ, Gooszen HG, van Santvoort HC, Bruno MJ, Boermeester MA. Dutch Chronic Pancreatitis Registry (CARE): design and rationale of a nationwide prospective evaluation and follow-up. Pancreatology 2014; 15:46-52. [PMID: 25511908 DOI: 10.1016/j.pan.2014.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 10/30/2014] [Accepted: 11/14/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic pancreatitis is a complex disease with many unanswered questions regarding the natural history and therapy. Prospective longitudinal studies with long-term follow-up are warranted. METHODS The Dutch Chronic Pancreatitis Registry (CARE) is a nationwide registry aimed at prospective evaluation and follow-up of patients with chronic pancreatitis. All patients with (suspected) chronic or recurrent pancreatitis are eligible for CARE. Patients are followed-up by yearly questionnaires and review of medical records. Study outcomes are pain, disease complications, quality of life, and pancreatic function. The target sample size was set at 500 for the first year and 1000 patients within 3 years. RESULTS A total of 1218 patients were included from February 2010 until June 2013 by 76 participating surgeons and gastroenterologist from 33 hospitals. Participation rate was 90% of eligible patients. Eight academic centers included 761 (62%) patients, while 25 community hospitals included 457 (38%). Patient centered outcomes were assessed by yearly questionnaires, which had a response rate of 85 and 82% for year 1 and 2, respectively. The median age of patients was 58 years, 814 (67%) were male, and 38% had symptoms for less than 5 years. DISCUSSION The CARE registry has successfully recruited over 1200 patients with chronic and recurrent pancreatitis in about 3 years. The defined inclusion criteria ensure patients are included at an early disease stage. Participation and compliance rates are high. CARE offers a unique opportunity with sufficient power to investigate many clinical questions regarding natural course, complications, and efficacy and timing of treatment strategies.
Collapse
Affiliation(s)
- U Ahmed Ali
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht, Netherlands.
| | - Y Issa
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - H van Goor
- Department of Surgery, RadboudUMC, Nijmegen, Netherlands
| | - C H van Eijck
- Department of Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Y Keulemans
- Department of Gastroenterology, Maastricht University Medical Center, Maastricht, Netherlands
| | - P Fockens
- Department of Gastroenterology, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - O R Busch
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - J P Drenth
- Department of Gastroenterology, RadboudUMC, Nijmegen, Netherlands
| | - C H Dejong
- Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - H M van Dullemen
- Department of Gastroenterology, University Medical Center Groningen, Groningen, Netherlands
| | - J E van Hooft
- Department of Gastroenterology, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - P D Siersema
- Department of Gastroenterology, University Medical Center Utrecht, Utrecht, Netherlands
| | - B W M Spanier
- Department of Gastroenterology, Rijnstate Hospital, Arnhem, Netherlands
| | - J W Poley
- Department of Gastroenterology, Erasmus Medical Center, Rotterdam, Netherlands
| | - A C Poen
- Department of Gastroenterology, Isala Clinics, Zwolle, Netherlands
| | - R Timmer
- Department of Gastroenterology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - T Seerden
- Department of Gastroenterology, Amphia Hospital, Breda, Netherlands
| | - A C Tan
- Department of Gastroenterology, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | - W J Thijs
- Department of Gastroenterology, Martini Hospital, Groningen, Netherlands
| | - B J M Witteman
- Department of Gastroenterology, Gelderse Vallei Hospital, Ede, Netherlands
| | - T E H Romkens
- Department of Gastroenterology, Jeroen Bosch Hospital, 's-Hertogenbosch, Netherlands
| | - A J Roeterdink
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - H G Gooszen
- Department of OR and Evidence Based Surgery, RadboudUMC, Nijmegen, Netherlands
| | - H C van Santvoort
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - M J Bruno
- Department of Gastroenterology, Erasmus Medical Center, Rotterdam, Netherlands
| | - M A Boermeester
- Department of Surgery, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
182
|
Park JM, Lee S, Chung MK, Kwon SH, Kim EH, Ko KH, Kwon CI, Hahm KB. Antioxidative phytoceuticals to ameliorate pancreatitis in animal models: An answer from nature. World J Gastroenterol 2014; 20:16570-16581. [PMID: 25469025 PMCID: PMC4248200 DOI: 10.3748/wjg.v20.i44.16570] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/10/2014] [Accepted: 05/14/2014] [Indexed: 02/06/2023] Open
Abstract
Despite enthusiastic efforts directed at elucidating critical underlying mechanisms towards the identification of novel therapeutic targets for severe acute pancreatitis (SAP), the disease remains without a specific therapy to be executed within the first hours to days after onset of symptoms. Although earlier management for SAP should aim to either treat organ failure or reduce infectious complications, the current standard of care for the general management of AP in the first hours to days after onset of symptoms include intravenous fluid replacement, nutritional changes, and the use of analgesics with a close monitoring of vital signs. Furthermore, repeated evaluation of severity is very important, as the condition is particularly unstable in the early stages. In cases where biliary pancreatitis is accompanied by acute cholangitis or in cases where biliary stasis is suspected, an early endoscopic retrograde cholangiopancreatography is recommended. However, practice guidelines regarding the treatment of pancreatitis are suboptimal. In chronic pancreatitis, conservative management strategies include lifestyle modifications and dietary changes followed by analgesics and pancreatic enzyme supplementation. Recently, attention has been focused on phytoceuticals or antioxidants as agents that could surpass the limitations associated with currently available therapies. Because oxidative stress has been shown to play an important role in the pathogenesis of pancreatitis, antioxidants alone or combined with conventional therapy may improve oxidative-stress-induced organ damage. Interest in phytoceuticals stems from their potential use as simple, accurate tools for pancreatitis prognostication that could replace older and more tedious methods. Therefore, the use of antioxidative nutrition or phytoceuticals may represent a new direction for clinical research in pancreatitis. In this review article, recent advances in the understanding of the pathogenesis of pancreatitis are discussed and the paradigm shift underway to develop phytoceuticals and antioxidants to treat it is introduced. Despite the promise of studies evaluating the effects of antioxidants/phytoceuticals in pancreatitis, translation to the clinic has thus far been disappointing. However, it is expected that continued research will provide solid evidence to justify the use of antioxidative phytoceuticals in the treatment of pancreatitis.
Collapse
|
183
|
Abstract
Chronic pancreatitis is a progressive inflammatory disease in which pancreatic secretory parenchyma is destroyed and replaced by fibrous tissue, eventually leading to malnutrition and diabetes. Alcohol is the leading cause in Western countries, but genetic factors are also implicated. Since the identification of mutations in the cationic trypsinogen (PRSS1) gene as a cause of hereditary pancreatitis in 1996, we have seen great progress in our understanding of the genetics of pancreatitis. It has been established that mutations in the genes related to the activation and inactivation of trypsin(ogen) such as PRSS1, serine protease inhibitor Kazal type 1 (SPINK1) and chymotrypsin C (CTRC) genes are associated with pancreatitis. In 2013, carboxypeptidase A1 (CPA1) was identified as a novel pancreatitis susceptibility gene. Endoplasmic reticulum stress in pancreatic acinar cells resulting from the mis-folding of mutated pancreatic enzymes has been shown to act as a novel mechanism underlying the susceptibility to pancreatitis. In Japan, the nationwide survey revealed 171 patients (96 males and 75 females) with hereditary pancreatitis in 59 families based on the European Registry of Hereditary Pancreatitis and Familial Pancreatic Cancer criteria. Because about 30% of families with hereditary pancreatitis do not carry mutations in any of the known pancreatitis susceptibility genes, other yet unidentified genes might be involved. Next generation sequencers can perform billions of sequencing reactions with a read length of 150-250 nucleotides. Comprehensive analysis using next generation sequencers will be a promising strategy to identify novel pancreatitis-associated genes and further clarify the pathogenesis of pancreatitis.
Collapse
Affiliation(s)
- Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| |
Collapse
|
184
|
50 years ago in The Journal of Pediatrics: Pancreatic lithiasis due to malnutrition and alcoholism in a child. J Pediatr 2014; 165:743. [PMID: 25256051 DOI: 10.1016/j.jpeds.2014.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
185
|
Sakata K, Ohmuraya M, Araki K, Suzuki C, Ida S, Hashimoto D, Wang J, Uchiyama Y, Baba H, Yamamura KI. Generation and analysis of serine protease inhibitor kazal type 3-cre driver mice. Exp Anim 2014; 63:45-53. [PMID: 24521862 PMCID: PMC4160937 DOI: 10.1538/expanim.63.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Serine protease inhibitor Kazal type 1 (SPINK1; mouse homologue Spink3) was initially discovered as a trypsin-specific inhibitor in the pancreas. However, previous studies have suggested that SPINK1/Spink3 is expressed in a wide range of normal tissues and tumors, although precise characterization of its gene expression has not been described in adulthood. To further analyze Spink3 expression, we generated two mouse lines in which either lacZ or Cre recombinase genes were inserted into the Spink3 locus by Cre-loxP technology. In Spink3(lacZ) mice, β-galactosidase activity was found in acinar cells of the pancreas and kidney, as well as epithelial cells of the bronchus in the lung, but not in the gastrointestinal tract or liver. Spink3(cre) knock-in mice were crossed with Rosa26 reporter (R26R) mice to monitor Spink3 promoter activity. In Spink3(cre);R26R mice, β-galactosidase activity was found in acinar cells of the pancreas, kidney, lung, and a small proportion of cells in the gastrointestinal tract and liver. These data suggest that Spink3 is widely expressed in endoderm-derived tissues, and that Spink3(cre) knock-in mice are a useful tool for establishment of a conditional knockout mice to analyze Spink3 function not only in normal tissues, but also in tumors that express SPINK1/Spink3.
Collapse
Affiliation(s)
- Kazuya Sakata
- Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Abstract
This Review covers the latest developments in the treatment options for chronic pancreatitis. Pain is the most frequent and dominant symptom in patients with chronic pancreatitis, which ranges from severe disabling continuous pain to mild pain attacks and pain-free periods. Conventional treatment strategies and recent changes in the treatment of pain in patients with chronic pancreatitis are outlined. The different treatment options for pain consist of medical therapy, endoscopy or surgery. Their related merits and drawbacks are discussed. Finally, novel insights in the field of genetics and microbiota are summarized, and future perspectives are discussed.
Collapse
|
187
|
Bhatia V, Rastellini C, Han S, Aronson JF, Greeley GH, Falzon M. Acinar cell-specific knockout of the PTHrP gene decreases the proinflammatory and profibrotic responses in pancreatitis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G533-49. [PMID: 25035110 PMCID: PMC4154118 DOI: 10.1152/ajpgi.00428.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pancreatitis is a necroinflammatory disease with acute and chronic manifestations. Accumulated damage incurred during repeated bouts of acute pancreatitis (AP) can lead to chronic pancreatitis (CP). Pancreatic parathyroid hormone-related protein (PTHrP) levels are elevated in a mouse model of cerulein-induced AP. Here, we show elevated PTHrP levels in mouse models of pancreatitis induced by chronic cerulein administration and pancreatic duct ligation. Because acinar cells play a major role in the pathophysiology of pancreatitis, mice with acinar cell-specific targeted disruption of the Pthrp gene (PTHrP(Δacinar)) were generated to assess the role of acinar cell-secreted PTHrP in pancreatitis. These mice were generated using Cre-LoxP technology and the acinar cell-specific elastase promoter. PTHrP(Δacinar) exerted protective effects in cerulein and pancreatic duct ligation models, evident as decreased edema, histological damage, amylase secretion, pancreatic stellate cell (PSC) activation, and extracellular matrix deposition. Treating acinar cells in vitro with cerulein increased IL-6 expression and NF-κB activity; these effects were attenuated in PTHrP(Δacinar) cells, as were the cerulein- and carbachol-induced elevations in amylase secretion. The cerulein-induced upregulation of procollagen I expression was lost in PSCs from PTHrP(Δacinar) mice. PTHrP immunostaining was elevated in human CP sections. The cerulein-induced upregulation of IL-6 and ICAM-1 (human acinar cells) and procollagen I (human PSCs) was suppressed by pretreatment with the PTH1R antagonist, PTHrP (7-34). These findings establish PTHrP as a novel mediator of inflammation and fibrosis associated with CP. Acinar cell-secreted PTHrP modulates acinar cell function via its effects on proinflammatory cytokine release and functions via a paracrine pathway to activate PSCs.
Collapse
Affiliation(s)
- Vandanajay Bhatia
- 1Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas;
| | | | - Song Han
- 2Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| | - Judith F. Aronson
- 3Department of Pathology, University of Texas Medical Branch, Galveston, Texas; and
| | - George H. Greeley
- 2Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| | - Miriam Falzon
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
188
|
Ahmed Ali U, Jens S, Busch ORC, Keus F, van Goor H, Gooszen HG, Boermeester MA, Cochrane Upper GI and Pancreatic Diseases Group. Antioxidants for pain in chronic pancreatitis. Cochrane Database Syst Rev 2014; 2014:CD008945. [PMID: 25144441 PMCID: PMC10114264 DOI: 10.1002/14651858.cd008945.pub2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Reduced intake and absorption of antioxidants due to pain and malabsorption are probable causes of the lower levels of antioxidants observed in patients with chronic pancreatitis (CP). Improving the status of antioxidants might be effective in slowing the disease process and reducing pain in CP. OBJECTIVES To assess the benefits and harms of antioxidants for the treatment of pain in patients with CP. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE and the Conference Proceedings Citation Index from inception to October 2012. Two review authors performed the selection of trials independently. SELECTION CRITERIA We included all randomised controlled trials (RCTs) evaluating antioxidants for treatment of pain in CP. All trials were included irrespective of blinding, numbers of participants randomly assigned and language of the article. DATA COLLECTION AND ANALYSIS Two review authors extracted data independently. The risk of bias of included trials was assessed. Study authors were asked for additional information in the case of missing data. MAIN RESULTS Twelve RCTs with a total of 585 participants were included. Six trials were double-blinded, placebo-controlled studies, and the other six trials were of less adequate methodology. Most trials were small and had high rates of dropout. Eleven of the 12 included trials described the effects of antioxidants on chronic abdominal pain in chronic pancreatitis. Pain as measured on a visual analogue scale (VAS, scale range 0 to 10) after one to six months was less in the antioxidant group than in the control group (mean difference (MD) -0.33, 95% confidence interval (CI) -0.64 to -0.02, P value 0.04, moderate-quality evidence). The number of pain-free participants was not statistically significantly different (risk ratio (RR) 1.73, 95% CI 0.95 to 3.15, P value 0.07, low-quality evidence). More adverse events were observed in the antioxidant group, both in the parallel trials (RR 4.43, 95% CI 1.60 to 12.29, P value 0.0004, moderate-quality evidence) and in the cross-over trials (RR 5.80, 95% CI 1.56 to 21.53, P value 0.0009, moderate-quality evidence). Adverse events occurred in 16% of participants and were mostly mild (e.g. headache, gastrointestinal complaints), but were sufficient to make participants stop antioxidant use. Other important outcomes such as use of analgesics, exacerbation of pancreatitis and quality of life were rarely reported. One trial from 1991 evaluated the effects of antioxidants on acute pain during exacerbation of chronic pancreatitis and found that a significantly higher proportion of participants in the antioxidant group experienced pain relief. This trial was conducted more than 25 years ago and has not been reproduced since that time. Therefore, additional trials are needed before reliable conclusions can be drawn. AUTHORS' CONCLUSIONS Current evidence shows that antioxidants can reduce pain slightly in patients with chronic pancreatitis. The clinical relevance of this small reduction is uncertain, and more evidence is needed. Adverse events in one of six patients may prevent the use of antioxidants. Effects of antioxidants on other outcome measures, such as use of analgesics, exacerbation of pancreatitis and quality of life remain uncertain because reliable data are not available.
Collapse
Affiliation(s)
- Usama Ahmed Ali
- University Medical Center UtrechtDepartment of SurgeryHeidelberglaan 100P.O. Box 85500UtrechtUtrechtNetherlands3508 GA
| | - Sjoerd Jens
- Academic Medical Centre, University of AmsterdamDepartment of RadiologyAmsterdamNetherlands
| | - Olivier RC Busch
- University of AmsterdamDepartment of Surgery, Academic Medical CenterAmsterdamNetherlands
| | - Frederik Keus
- University of Groningen, University Medical Center GroningenDepartment of Critical CareHanzeplein 1GroningenNetherlands9713 GZ
| | - Harry van Goor
- Radboud University Nijmegen Medical CentreDepartment of SurgeryPO Box 9101NijmegenNetherlands6500 HB
| | - Hein G Gooszen
- Radboud University Nijmegen Medical CenterCentre of Evidence‐based SurgeryPO Box 9101Huispost 630, route 631NijmegenNetherlands6500 HB
| | - Marja A Boermeester
- University of AmsterdamDepartment of Surgery, Academic Medical CenterAmsterdamNetherlands
| | | |
Collapse
|
189
|
D'Haese JG, Ceyhan GO, Demir IE, Tieftrunk E, Friess H. Treatment options in painful chronic pancreatitis: a systematic review. HPB (Oxford) 2014; 16:512-21. [PMID: 24033614 PMCID: PMC4048072 DOI: 10.1111/hpb.12173] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/05/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Longlasting and unbearable pain is the most common and striking symptom of chronic pancreatitis. Accordingly, pain relief and improvement in patients' quality of life are the primary goals in the treatment of this disease. This systematic review aims to summarize the available data on treatment options. METHODS A systematic search of MEDLINE/PubMed and the Cochrane Library was performed according to the PRISMA statement for reporting systematic reviews and meta-analysis. The search was limited to randomized controlled trials and meta-analyses. Reference lists were then hand-searched for additional relevant titles. The results obtained were examined individually by two independent investigators for further selection and data extraction. RESULTS A total of 416 abstracts were reviewed, of which 367 were excluded because they were obviously irrelevant or represented overlapping studies. Consequently, 49 full-text articles were systematically reviewed. CONCLUSIONS First-line medical options include the provision of pain medication, adjunctive agents and pancreatic enzymes, and abstinence from alcohol and tobacco. If medical treatment fails, endoscopic treatment offers pain relief in the majority of patients in the short term. However, current data suggest that surgical treatment seems to be superior to endoscopic intervention because it is significantly more effective and, especially, lasts longer.
Collapse
Affiliation(s)
- Jan G D'Haese
- Department of Surgery, Rechts der Isar Clinic, Technical University of Munich, Munich, Germany
| | | | | | | | | |
Collapse
|
190
|
Yang J, Huang Q, Lin XS, Liu CH, Xie F, Li RY. Endoscopic versus surgical treatment of chronic pancreatitis: A systematic review. Shijie Huaren Xiaohua Zazhi 2014; 22:2183-2189. [DOI: 10.11569/wcjd.v22.i15.2183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare the long-term clinical outcomes and complications between endoscopic and surgical treatment of chronic pancreatitis.
METHODS: An electronic search of Medline, EMBASE, Science Direct, Springer link, CBM, CNKI, Wan fang and VIP databases (before December 21, 2013) was performed to retrieve the articles reporting endoscopic versus surgical treatment of chronic pancreatitis. The quality of the included trials was assessed according to the inclusive and exclusive criteria, and the data were extracted and analyzed using RevMan 5.2.7 software.
RESULTS: A total of 5 articles involving 393 chronic pancreatitis patients were included in the analysis, including 4 English articles and 1 Chinese article. The meta-analysis showed that the patients undergoing surgical treatment had higher rates of pain relief and complete pain relief (P < 0.05), but there were no significant differences in the rate of partial relief, incidence of diabetes at 1, 3, and 5 years after treatment, the incidence of complications, or endocrine and exocrine functions of the pancreas (P > 0.05).
CONCLUSION: Surgical treatment is associated with better quality of life than endoscopic treatment. Due to the traumatic risk of surgery, endoscopic treatment is safe, and can be the first choice for treatment of chronic pancreatitis.
Collapse
|
191
|
Chen H, Mrazek AA, Wang X, Ding C, Ding Y, Porro LJ, Liu H, Chao C, Hellmich MR, Zhou J. Design, synthesis, and characterization of novel apigenin analogues that suppress pancreatic stellate cell proliferation in vitro and associated pancreatic fibrosis in vivo. Bioorg Med Chem 2014; 22:3393-404. [PMID: 24837156 DOI: 10.1016/j.bmc.2014.04.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/14/2014] [Accepted: 04/22/2014] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests that activated pancreatic stellate cells (PSC) play an important role in chronic pancreatitis (CP), and inhibition of the activated PSC is considered as a potential strategy for the treatment and prevention of CP. Herein, we disclose our findings that apigenin and its novel analogues suppress the proliferation and induce apoptosis in PSC, which reduce the PSC-mediated fibrosis in CP. Chemical modifications of apigenin have been directed to build a focused library of O-alkylamino-tethered apigenin derivatives at 4'-O position of the ring C with the attempt to enhance the potency and drug-like properties including aqueous solubility. A number of compounds such as 14, 16, and 24 exhibited potent antiproliferative effects as well as improved aqueous solubility. Intriguingly, apigenin, new analogues 23 and 24 displayed significant efficacy to reduce pancreatic fibrosis even at a low dose of 0.5mg/kg in our proof-of-concept study using a preclinical in vivo mouse model of CP.
Collapse
Affiliation(s)
- Haijun Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Amy A Mrazek
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Xiaofu Wang
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Chunyong Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Ye Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Laura J Porro
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Huiling Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Celia Chao
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
192
|
Glass LM, Whitcomb DC, Yadav D, Romagnuolo J, Kennard E, Slivka AA, Brand RE, Anderson MA, Banks PA, Lewis MD, Baillie J, Sherman S, Alkaade S, Amann ST, Disario JA, O'Connell M, Gelrud A, Forsmark CE, Gardner TB. Spectrum of use and effectiveness of endoscopic and surgical therapies for chronic pancreatitis in the United States. Pancreas 2014; 43:539-43. [PMID: 24717802 PMCID: PMC4122518 DOI: 10.1097/mpa.0000000000000122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This study aims to describe the frequency of use and reported effectiveness of endoscopic and surgical therapies in patients with chronic pancreatitis treated at US referral centers. METHODS Five hundred fifteen patients were enrolled prospectively in the North American Pancreatitis Study 2, where patients and treating physicians reported previous therapeutic interventions and their perceived effectiveness. We evaluated the frequency and effectiveness of endoscopic (biliary or pancreatic sphincterotomy, biliary or pancreatic stent placement) and surgical (pancreatic cyst removal, pancreatic drainage procedure, pancreatic resection, surgical sphincterotomy) therapies. RESULTS Biliary and/or pancreatic sphincterotomy (42%) were the most common endoscopic procedure (biliary stent, 14%; pancreatic stent, 36%; P < 0.001). Endoscopic procedures were equally effective (biliary sphincterotomy, 40.0%; biliary stent, 40.8%; pancreatic stent, 47.0%; P = 0.34). On multivariable analysis, the presence of abdominal pain (odds ratio, 1.82; 95% confidence interval, 1.15-2.88) predicted endoscopy, whereas exocrine insufficiency (odds ratio, 0.63; 95% confidence interval, 0.42-0.94) deterred endoscopy. Surgical therapies were attempted equally (cyst removal, 7%; drainage procedure, 10%; resection procedure, 12%) except for surgical sphincteroplasty (4%; P < 0.001). Surgical sphincteroplasty was the least effective (46%; P < 0.001) versus cyst removal (76% drainage [71%] and resection [73%]). CONCLUSIONS Although surgical therapies were performed less frequently than endoscopic therapies, they were more often reported to be effective.
Collapse
Affiliation(s)
- Lisa M Glass
- From the *Section of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, NH; †Department of Medicine, University of Pittsburgh, Pittsburgh, PA; ‡Digestive Disease Center, Medical University of South Carolina, Charleston, SC; §University of Michigan, Ann Arbor, MI; ∥Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA; ¶Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL; #Department of Medicine, Wake Forest University Medical Center, Winston-Salem, NC; **Department of Medicine, Indiana University Medical Center, Indianapolis, IN; ††Department of Internal Medicine, St Louis University School of Medicine, St Louis, MO; ‡‡North Mississippi Medical Center, Tupelo, MS; §§Monterey Gastroenterology, Monterey, CA; and ∥∥Department of Medicine, University of Florida, Gainesville, FL
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Hamada S, Masamune A, Shimosegawa T. Inflammation and pancreatic cancer: disease promoter and new therapeutic target. J Gastroenterol 2014; 49:605-17. [PMID: 24292163 DOI: 10.1007/s00535-013-0915-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 11/13/2013] [Indexed: 02/04/2023]
Abstract
Chronic inflammation has a certain impact on the carcinogenesis of the digestive organs. The characteristic tissue structure of pancreatic cancer, desmoplasia, results from inflammatory processes induced by cancer cells and stromal cells. Concerning the progression of pancreatic cancer, recent research has clarified the pivotal role of tumor-stromal interaction, which promotes the development of an invasive phenotype of cancer and provides survival advantages against chemotherapeutic agents or immune surveillance. Tumor stromal cells such as pancreatic stellate cells and immune cells establish a microenvironment that protects cancer cells through complex interactions. The microenvironment of pancreatic cancer acts as a niche for pancreatic cancer stem cells from which therapy-resistance and disease recurrence develop. Inhibition of the stromal functions or restoration of the immune reaction against cancer cells has therapeutic benefits that enhance the efficacy of conventional therapies. Some of the recent advances in this field are now under evaluation in clinical settings, but many problems must be overcome to establish a radical therapy for pancreatic cancer. This review summarizes current knowledge about the tumor-promoting stromal functions, immune system modulation and therapeutic strategies targeting tumor-stromal interactions in pancreatic cancer.
Collapse
Affiliation(s)
- Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi Aobaku, Sendai, Miyagi, 980-8574, Japan,
| | | | | |
Collapse
|
194
|
Löhr JM, Haas S. Can a polymorphism in the thalassemia gene and a heterozygote CFTR mutation cause acute pancreatitis? World J Clin Cases 2014; 2:62-66. [PMID: 24653987 PMCID: PMC3955802 DOI: 10.12998/wjcc.v2.i3.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/10/2013] [Accepted: 01/15/2014] [Indexed: 02/05/2023] Open
Abstract
The case of a 32-year-old black woman of African descent who suffered from repeated episodes of acute pancreatitis, initially triggered when flying on airplanes, is reported. She did not drink alcohol or smoke. Genetic analysis was negative for cationic trypsinogen, serine protease inhibitor Kazal type 1 and chymotrypsin C. However, hemoglobin F was elevated. Sequencing of the thalassemia gene revealed a novel alteration in the 5' region indicative of a functional abnormality of the molecule. Sequencing the cystic fibrosis transmembrane conductance regulator (CFTR) gene revealed a heterozygote sequence variant. The combination of a hemoglobin gene mutation known for thalassemia in conjunction with the hitherto undescribed CFTR mutation is suggested to pave the road for initial and repetitive pancreatitis attacks. This will be discussed.
Collapse
|
195
|
Lesina M, Wörmann SM, Neuhöfer P, Song L, Algül H. Interleukin-6 in inflammatory and malignant diseases of the pancreas. Semin Immunol 2014; 26:80-7. [DOI: 10.1016/j.smim.2014.01.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023]
|
196
|
Bellin MD, Freeman ML, Gelrud A, Slivka A, Clavel A, Humar A, Schwarzenberg SJ, Lowe ME, Rickels MR, Whitcomb DC, Matthews JB, Amann S, Andersen DK, Anderson MA, Baillie J, Block G, Brand R, Chari S, Cook M, Cote GA, Dunn T, Frulloni L, Greer JB, Hollingsworth MA, Kim KM, Larson A, Lerch MM, Lin T, Muniraj T, Robertson RP, Sclair S, Singh S, Stopczynski R, Toledo FGS, Wilcox CM, Windsor J, Yadav D. Total pancreatectomy and islet autotransplantation in chronic pancreatitis: recommendations from PancreasFest. Pancreatology 2014; 14:27-35. [PMID: 24555976 PMCID: PMC4058640 DOI: 10.1016/j.pan.2013.10.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/23/2013] [Accepted: 10/25/2013] [Indexed: 12/11/2022]
Abstract
DESCRIPTION Total pancreatectomy with islet autotransplantation (TPIAT) is a surgical procedure used to treat severe complications of chronic pancreatitis or very high risk of pancreatic cancer while reducing the risk of severe diabetes mellitus. However, clear guidance on indications, contraindications, evaluation, timing, and follow-up are lacking. METHODS A working group reviewed the medical, psychological, and surgical options and supporting literature related to TPIAT for a consensus meeting during PancreasFest. RESULTS Five major areas requiring clinical evaluation and management were addressed: These included: 1) indications for TPIAT; 2) contraindications for TPIAT; 3) optimal timing of the procedure; 4) need for a multi-disciplinary team and the roles of the members; 5) life-long management issues following TPIAP including diabetes monitoring and nutrition evaluation. CONCLUSIONS TPIAT is an effective method of managing the disabling complications of chronic pancreatitis and risk of pancreatic cancer in very high risk patients. Careful evaluation and long-term management of candidate patients by qualified multidisciplinary teams is required. Multiple recommendations for further research were also identified.
Collapse
Affiliation(s)
- Melena D. Bellin
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Martin L. Freeman
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andres Gelrud
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Adam Slivka
- Department of Medicine, University of Pittsburgh, Pennsylvania, USA
| | - Alfred Clavel
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Abhinav Humar
- Department of Surgery, University of Pittsburgh, Pennsylvania, USA
| | | | - Mark E. Lowe
- Department of Pediatrics, University of Pittsburgh, Pennsylvania, USA,Children’s Hospital of Pittsburgh, Pennsylvania, USA
| | - Michael R. Rickels
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David C Whitcomb
- Department of Medicine, University of Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Acquired immunity plays an important role in the development of murine experimental pancreatitis induced by alcohol and lipopolysaccharide. Pancreas 2014; 43:28-36. [PMID: 24201778 DOI: 10.1097/mpa.0b013e3182a7c76b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Although chronic alcohol ingestion is the major cause of chronic pancreatitis, less than 10% of alcohol abusers develop this disease. To address this issue, we created a murine model of pancreatitis induced by alcohol and lipopolysaccharide (LPS) and analyzed its immune responses. METHODS C57BL/6 mice were administered 20% ethanol (AL) in their drinking water and then injected intraperitoneally with LPS twice weekly for 4 weeks. Severe combined immunodeficient mice were reconstituted with splenocytes, CD4 cells, or CD8 T cells isolated from wild-type mice and then treated similarly. The severity of pancreatitis was graded histologically, and serum cytokine levels were measured. RESULTS Ethanol alone did not cause pancreatitis. However, the administration of AL+LPS or LPS alone induced pancreatitis. The histological scores were higher in the mice treated with AL+LPS than in those treated with LPS. Serum levels of interleukin 1β, interferon-γ, and tumor necrosis factor α were elevated in the AL+LPS-treated mice. The severe combined immunodeficient mice developed pancreatitis only after their reconstitution with splenocytes, CD4 cells, or CD8 T cells. CONCLUSIONS Repeated stimulation of the innate immune system is necessary, but not sufficient, to cause pancreatitis. The participation of the acquired immune response is essential for the development of the disease.
Collapse
|
198
|
Olesen SS, Juel J, Graversen C, Kolesnikov Y, Wilder-Smith OHG, Drewes AM. Pharmacological pain management in chronic pancreatitis. World J Gastroenterol 2013; 19:7292-7301. [PMID: 24259960 PMCID: PMC3831211 DOI: 10.3748/wjg.v19.i42.7292] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 08/05/2013] [Accepted: 08/20/2013] [Indexed: 02/06/2023] Open
Abstract
Intense abdominal pain is a prominent feature of chronic pancreatitis and its treatment remains a major clinical challenge. Basic studies of pancreatic nerves and experimental human pain research have provided evidence that pain processing is abnormal in these patients and in many cases resembles that seen in neuropathic and chronic pain disorders. An important ultimate outcome of such aberrant pain processing is that once the disease has advanced and the pathophysiological processes are firmly established, the generation of pain can become self-perpetuating and independent of the initial peripheral nociceptive drive. Consequently, the management of pain by traditional methods based on nociceptive deafferentation (e.g., surgery and visceral nerve blockade) becomes difficult and often ineffective. This novel and improved understanding of pain aetiology requires a paradigm shift in pain management of chronic pancreatitis. Modern mechanism based pain treatments taking into account altered pain processing are likely to increasingly replace invasive therapies targeting the nociceptive source, which should be reserved for special and carefully selected cases. In this review, we offer an overview of the current available pharmacological options for pain management in chronic pancreatitis. In addition, future options for pain management are discussed with special emphasis on personalized pain medicine and multidisciplinarity.
Collapse
|
199
|
Reduction of fibrosis in dibutyltin dichloride-induced chronic pancreatitis using rat umbilical mesenchymal stem cells from Wharton's jelly. Pancreas 2013; 42:1291-302. [PMID: 24152954 DOI: 10.1097/mpa.0b013e318296924e] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The objective of this study was to investigate the effects of rat umbilical cord mesenchymal stem cells (UCMSCs) from Wharton's jelly on dibutyltin dichloride (DBTC)-induced chronic pancreatitis (CP) and subsequent pancreatic fibrosis in rats. METHODS A rat model of CP induced by DBTC was used. Male Sprague-Dawley rats were randomly divided into 4 groups: the control, DBTC, DBTC + UCMSCs, and control + UCMSC groups. Umbilical cord mesenchymal stem cells were administered intravenously on day 5 after the administration of DBTC. On days 14 and 28, the rats were evaluated morphologically and biochemically. The expression levels of inflammatory cytokines and chemokines in the pancreatic tissues of different groups were evaluated using quantitative real-time polymerase chain reaction. The activation of pancreatic stellate cells was estimated by immunochemistry and Western blot analysis of α-smooth muscle actin. RESULTS Umbilical cord mesenchymal stem cells were detected in inflamed pancreatic tissues. Umbilical cord mesenchymal stem cell treatment improved the histological scores and alleviated the fibrosis of pancreas samples, The expression of cytokines in the DBTC + UCMSC group was significantly lower than that in the DBTC group. Also, pancreatic stellate cell activation was inhibited by UCMSC treatment. CONCLUSIONS Xenogeneic transplantation of UCMSCs is a novel approach for the treatment of CP and subsequent fibrosis. Umbilical cord mesenchymal stem cells may be a promising therapeutic intervention for human CP in the future.
Collapse
|
200
|
Witt H, Beer S, Rosendahl J, Chen JM, Chandak GR, Masamune A, Bence M, Szmola R, Oracz G, Macek M, Bhatia E, Steigenberger S, Lasher D, Bühler F, Delaporte C, Tebbing J, Ludwig M, Pilsak C, Saum K, Bugert P, Masson E, Paliwal S, Bhaskar S, Sobczynska-Tomaszewska A, Bak D, Balascak I, Choudhuri G, Reddy DN, Rao GV, Thomas V, Kume K, Nakano E, Kakuta Y, Shimosegawa T, Durko L, Szabó A, Schnúr A, Hegyi P, Rakonczay Z, Pfützer R, Schneider A, Groneberg DA, Braun M, Schmidt H, Witt U, Friess H, Algül H, Landt O, Schuelke M, Krüger R, Wiedenmann B, Schmidt F, Zimmer KP, Kovacs P, Stumvoll M, Blüher M, Müller T, Janecke A, Teich N, Grützmann R, Schulz HU, Mössner J, Keim V, Löhr M, Férec C, Sahin-Tóth M. Variants in CPA1 are strongly associated with early onset chronic pancreatitis. Nat Genet 2013; 45:1216-1220. [PMID: 23955596 PMCID: PMC3909499 DOI: 10.1038/ng.2730] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/23/2013] [Indexed: 12/12/2022]
Abstract
Chronic pancreatitis is an inflammatory disorder of the pancreas. We analyzed CPA1, encoding carboxypeptidase A1, in subjects with nonalcoholic chronic pancreatitis (cases) and controls in a German discovery set and three replication sets. Functionally impaired variants were present in 29/944 (3.1%) German cases and 5/3,938 (0.1%) controls (odds ratio (OR) = 24.9, P = 1.5 × 10(-16)). The association was strongest in subjects aged ≤ 10 years (9.7%; OR = 84.0, P = 4.1 × 10(-24)). In the replication sets, defective CPA1 variants were present in 8/600 (1.3%) cases and 9/2,432 (0.4%) controls from Europe (P = 0.01), 5/230 (2.2%) cases and 0/264 controls from India (P = 0.02) and 5/247 (2.0%) cases and 0/341 controls from Japan (P = 0.013). The mechanism by which CPA1 variants confer increased pancreatitis risk may involve misfolding-induced endoplasmic reticulum stress rather than elevated trypsin activity, as is seen with other genetic risk factors for this disease.
Collapse
Affiliation(s)
- Heiko Witt
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Department of Pediatrics, Klinikum rechts der Isar (MRI), Technische Universität München (TUM), Parzivalstr. 16, 80804 Munich, Germany
| | - Sebastian Beer
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Jonas Rosendahl
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Jian-Min Chen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Giriraj Ratan Chandak
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Melinda Bence
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Richárd Szmola
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
- 2nd Department of Medicine, Semmelweis University, Szentkirályi u. 46, H-1088, Budapest, Hungary
| | - Grzegorz Oracz
- Department of Gastroenterology, Hepatology and Immunology, The Children’s Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Milan Macek
- Department of Biology and Medical Genetics, University Hospital Motol & 2nd Faculty of Medicine of Charles University Prague, V Uvalu 84, Prague, CZ 150 06, Czech Republic
| | - Eesh Bhatia
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Sandra Steigenberger
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Denise Lasher
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Florence Bühler
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Catherine Delaporte
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Johanna Tebbing
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Maren Ludwig
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Claudia Pilsak
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Karolin Saum
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München (TUM), Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg - Hessen, Friedrich-Ebert-Str. 107, 68167 Mannheim, Germany
| | - Emmanuelle Masson
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Sumit Paliwal
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Seema Bhaskar
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research (CSIR), Uppal Road, Hyderabad 500 007, India
| | - Agnieszka Sobczynska-Tomaszewska
- MedGen Health Care Centre, Orzycka 27, 02-695 Warsaw, Poland
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Daniel Bak
- Department of Medical Genetics, Institute of Mother and Child, ul. Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Ivan Balascak
- Clinic of Obstetrics and Gynecology, Neonatology Department, University Hospital Motol & 2 Medical School, Charles University Prague, V Uvalu 84, 103 Prague 5, Czech Republic
| | - Gourdas Choudhuri
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - D. Nageshwar Reddy
- Asian Institute of Gastroenterology, 6-3-661, Somajiguda, Hyderabad 500 082, India
| | - G. Venkat Rao
- Asian Institute of Gastroenterology, 6-3-661, Somajiguda, Hyderabad 500 082, India
| | - Varghese Thomas
- Department of Gastroenterology, Medical College Hospital, Calicut 673008, India
| | - Kiyoshi Kume
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Eriko Nakano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Yoichi Kakuta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Lukasz Durko
- Department of Digestive Tract Diseases, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland
| | - András Szabó
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| | - Andrea Schnúr
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Péter Hegyi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Zoltán Rakonczay
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720, Szeged, Hungary
| | - Roland Pfützer
- Department of Internal Medicine, Klinikum Döbeln, Sörmitzer Str. 10, 04720 Döbeln, Germany
| | - Alexander Schneider
- Department of Medicine II, Universitätsmedizin Mannheim, Ruprecht-Karls-Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - David Alexander Groneberg
- Institute for Occupational Medicine, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, Haus 9B, 60590 Frankfurt, Germany
| | - Markus Braun
- Institute for Occupational Medicine, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, Haus 9B, 60590 Frankfurt, Germany
| | - Hartmut Schmidt
- Department for Transplant Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Ulrike Witt
- Department of Surgery, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Hana Algül
- Department of Gastroenterology, Technische Universität München (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Olfert Landt
- TIB MOLBIOL, Eresburgstr. 22-23, 12103 Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
- NeuroCure Clinical Research Center, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Renate Krüger
- Department of Pediatrics, Division of Pediatric Pulmonology, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Bertram Wiedenmann
- Department of Internal Medicine, Division of Hepatology and Gastroenterology, Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Frank Schmidt
- Department of Pediatrics, University of Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle/Saale, Germany
| | - Klaus-Peter Zimmer
- Department of Pediatrics, Justus-Liebig-Universität, Feulgenstr. 12, 35385 Gießen, Germany
| | - Peter Kovacs
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Michael Stumvoll
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Matthias Blüher
- Department for Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | - Thomas Müller
- Universitätsklinik für Pädiatrie I, Department für Kinder-und Jugendheilkunde, Medizinische Universität Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Andreas Janecke
- Universitätsklinik für Pädiatrie I, Department für Kinder-und Jugendheilkunde, Medizinische Universität Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
- Sektion für Humangenetik, Medizinische Universität Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Niels Teich
- Practice for Digestive and Metabolic Diseases, Nordstr. 21, 04105 Leipzig, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Joachim Mössner
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Volker Keim
- Department for Internal Medicine, Neurology and Dermatology, Division of Gastroenterology, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078; Etablissement Français du Sang (EFS) – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 46 rue Félix Le Dantec, 29218 Brest, France
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Evans-433, Boston, MA 02118, USA
| |
Collapse
|