1
|
Buriko Y, Tinsley A. Controversies of and Indications for Use of Glucocorticoids in the Intensive Care Unit and the Emergency Room. Vet Clin North Am Small Anim Pract 2025; 55:427-442. [PMID: 40316370 DOI: 10.1016/j.cvsm.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Corticosteroids are ubiquitous endogenous compounds that are essential for most body functions. Exogenous steroids are routinely used for a plethora of conditions associated with substantial and sometimes detrimental inflammation or immune-mediated tissue destruction. In this article, we will review the relevant physiology of steroids, pharmacology of the common exogenously administered steroids, common side effects of glucocorticoid administration, as well as some of the more controversial and less researched indications for steroid therapy in veterinary medicine. Relevant human literature as well as available information on veterinary species will be presented to augment the discussion.
Collapse
Affiliation(s)
- Yekaterina Buriko
- Cornell University Veterinary Specialists, Stamford, Connecticut, USA.
| | - Ashlei Tinsley
- Cornell University Veterinary Specialists, Stamford, Connecticut, USA
| |
Collapse
|
2
|
Wei ZX, Jiang SH, Qi XY, Cheng YM, Liu Q, Hou XY, He J. scRNA-seq of the intestine reveals the key role of mast cells in early gut dysfunction associated with acute pancreatitis. World J Gastroenterol 2025; 31:103094. [PMID: 40182603 PMCID: PMC11962851 DOI: 10.3748/wjg.v31.i12.103094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Intestinal barrier dysfunction is a prevalent and varied manifestation of acute pancreatitis (AP). Molecular mechanisms underlying the early intestinal barrier in AP remain poorly understood. AIM To explore the biological processes and mechanisms of intestinal injury associated with AP, and to find potential targets for early prevention or treatment of intestinal barrier injury. METHODS This study utilized single-cell RNA sequencing of the small intestine, alongside in vitro and in vivo experiments, to examine intestinal barrier function homeostasis during the early stages of AP and explore involved biological processes and potential mechanisms. RESULTS Seventeen major cell types and 33232 cells were identified across all samples, including normal, AP1 (4x caerulein injections, animals sacrificed 2 h after the last injection), and AP2 (8x caerulein injections, animals sacrificed 4 h after the last injection). An average of 980 genes per cell was found in the normal intestine, compared to 927 in the AP1 intestine and 1382 in the AP2 intestine. B cells, dendritic cells, mast cells (MCs), and monocytes in AP1 and AP2 showed reduced numbers compared to the normal intestine. Enterocytes, brush cells, enteroendocrine cells, and goblet cells maintained numbers similar to the normal intestine, while cytotoxic T cells and natural killer (NK) cells increased. Enterocytes in early AP exhibited elevated programmed cell death and intestinal barrier dysfunction but retained absorption capabilities. Cytotoxic T cells and NK cells showed enhanced pathogen-fighting abilities. Activated MCs, secreted chemokine (C-C motif) ligand 5 (CCL5), promoted neutrophil and macrophage infiltration and contributed to barrier dysfunction. CONCLUSION These findings enrich our understanding of biological processes and mechanisms in AP-associated intestinal injury, suggesting that CCL5 from MCs is a potential target for addressing dysfunction.
Collapse
Affiliation(s)
- Zu-Xing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shi-He Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Yan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yi-Miao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Qiong Liu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xu-Yang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
3
|
Wu L, Hu J, Yi X, Lv J, Yao J, Tang W, Zhang S, Wan M. Gut microbiota interacts with inflammatory responses in acute pancreatitis. Therap Adv Gastroenterol 2023; 16:17562848231202133. [PMID: 37829561 PMCID: PMC10566291 DOI: 10.1177/17562848231202133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common acute abdominal conditions, and its incidence has been increasing for years. Approximately 15-20% of patients develop severe AP (SAP), which is complicated by critical inflammatory injury and intestinal dysfunction. AP-associated inflammation can lead to the gut barrier and function damage, causing dysbacteriosis and facilitating intestinal microbiota migration. Pancreatic exocrine deficiency and decreased levels of antimicrobial peptides in AP can also lead to abnormal growth of intestinal bacteria. Meanwhile, intestinal microbiota migration influences the pancreatic microenvironment and affects the severity of AP, which, in turn, exacerbates the systemic inflammatory response. Thus, the interaction between the gut microbiota (GM) and the inflammatory response may be a key pathogenic feature of SAP. Treating either of these factors or breaking their interaction may offer some benefits for SAP treatment. In this review, we discuss the mechanisms of interaction of the GM and inflammation in AP and factors that can deteriorate or even cure both, including some traditional Chinese medicine treatments, to provide new methods for studying AP pathogenesis and developing therapies.
Collapse
Affiliation(s)
- Linjun Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Jing Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Intensive Care Unit, Suining Municipal Hospital of TCM, Suining, China
| | - Jianqin Lv
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, Sichuan, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, China
| |
Collapse
|
4
|
Ling X, Nie C, Sheng LP, Han CQ, Ding Z. Disulfiram relieves severe acute pancreatitis by inhibiting GSDMD-dependent NETs formation. J Dig Dis 2023; 24:359-368. [PMID: 37503822 DOI: 10.1111/1751-2980.13211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/18/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023]
Abstract
OBJECTIVES Severe acute pancreatitis (SAP) is characterized by pancreatic and systemic inflammation and persistent organ failure. Neutrophil extracellular traps (NETs) are considered to play an important role in the development of SAP. We aimed to explore the effect of disulfiram (DSL), a newly developed anti-inflammatory drug, on NETs in SAP. METHODS A mouse model of SAP was induced by caerulein and lipopolysaccharide, and the mice were divided into the normal control group, the DSL group, the SAP group, and the SAP treated with 50 mg/kg (50DSLSAP) and 100 mg/kg DSL (100DSLSAP) groups, respectively. The severity of SAP was evaluated based on the morphological and biochemical changes. Gasdermin D (GSDMD) expression was evaluated in vivo and in vitro to verify the effect of DSL. Additionally, the expressions of NETs were also evaluated in vivo and in vitro in SAP with and without DSL treatment to explore the possible mechanism of DSL on SAP. RESULTS Pancreatic inflammatory injury increased in the SAP group, which was alleviated by DSL. GSDMD, a protein related to the formation of NETs, increased in SAP. Expressions of NETs were also promoted in the in vivo SAP model and by phorbol myristate acetate (PMA) in vitro. Moreover, DSL inhibited the expressions of GSDMD and NETs in vivo. The results were further confirmed in the in vitro experiment. CONCLUSIONS NETs are highly associated with inflammatory injury in SAP. DSL inhibits NETs formation by downregulating GSDMD, which in turn relieves the inflammation of SAP. Our study may provide a possible therapeutic target for SAP.
Collapse
Affiliation(s)
- Xin Ling
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chi Nie
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Li Ping Sheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chao Qun Han
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhen Ding
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
Cilingir S, Acikel-Elmas M, Arbak S, Kolgazi M. Ferulic acid attenuates pancreaticobiliary duct occlusion-induced inflammation in both pancreas and liver. Inflammopharmacology 2023; 31:997-1008. [PMID: 36752934 DOI: 10.1007/s10787-023-01150-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Acute pancreatitis is a systemic inflammatory disorder characterized by the hyperactivation of digestion enzymes and the release of proinflammatory cytokines. Ferulic acid (FA) is a hydroxycinnamic acid derivative that has recently been shown to have antioxidant and anti-inflammatory properties. AIM The anti-inflammatory effects of FA were investigated in the pancreaticobiliary duct ligation (PBDL)-induced pancreatitis model. METHODS Wistar albino rats (250-300 g; female = male) were divided into sham operation and PBDL groups. Some PBDL-performed animals were given intragastric saline or 250 mg/kg FA or 500 mg/kg FA 30 min before the PBDL and for 3 consecutive days. Moreover, the control group received saline. Blood samples are collected at the 24th, 48th, and 72nd hours to measure serum tumor necrosis factor (TNF)-α, liver, and pancreatic enzymes. At the 72nd hour, rats were euthanized; pancreas, lung, and liver samples were collected, scored microscopically, and analyzed for myeloperoxidase activity, malondialdehyde, and glutathione levels. One-way ANOVA with Tukey-Kramer tests were used for statistical analysis. RESULTS FA treatment reduced myeloperoxidase activity and prevented the depletion of glutathione in all three tissues. With FA treatments, high malondialdehyde levels in the pancreas and liver were reduced, as were serum TNF- α, amylase, lipase, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, and total bilirubin levels. Additionally, FA ameliorated microscopic damage in the pancreas and liver significantly. CONCLUSION According to the findings, FA protects endogenous antioxidant content, prevents neutrophil infiltration, and decreases lipid peroxidation in PBDL-induced pancreatitis. Furthermore, FA improves tissue damage induced by pancreatitis with its anti-inflammatory effects.
Collapse
Affiliation(s)
- Sumeyye Cilingir
- Institute of Health Sciences, Acibadem Mehmet Ali Aydınlar University, Icerenkoy Mah., Kayisdagi Cad. No: 32, Atasehir, 34752, Istanbul, Turkey
| | - Merve Acikel-Elmas
- Department of Histology and Embryology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Icerenkoy Mah., Kayisdagi Cad. No: 32, Atasehir, 34752, Istanbul, Turkey
| | - Serap Arbak
- Department of Histology and Embryology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Icerenkoy Mah., Kayisdagi Cad. No: 32, Atasehir, 34752, Istanbul, Turkey
| | - Meltem Kolgazi
- Department of Physiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Icerenkoy Mah., Kayisdagi Cad. No: 32, Atasehir, 34752, Istanbul, Turkey.
| |
Collapse
|
6
|
Xia S, Wang J, Kalionis B, Zhang W, Zhao Y. Genistein protects against acute pancreatitis via activation of an apoptotic pathway mediated through endoplasmic reticulum stress in rats. Biochem Biophys Res Commun 2018; 509:421-428. [PMID: 30594397 DOI: 10.1016/j.bbrc.2018.12.108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022]
Abstract
Acute pancreatitis (AP) is a severe and frequently lethal disorder, but the precise mechanisms are not well understood and there is lack of effective drugs. Therefore, our study examined the in vivo intervention effects of genistein and elucidated its mechanism in acute experimental pancreatitis models. We used cerulein or taurocholate to induce acute pancreatitis (AP) in Sprague-Dawley rats with prior genistein treatment. Histological examination of the pancreas was performed and the expression of unfolded protein response (UPR) components and apoptotic mediators like caspase 12 and c-Jun N-terminal protein kinase (JNK) were measured. The amount of apoptosis in pancreatic acinar cells was also determined. Our studies found that the severity of cerulein- or taurocholate-induced AP was rescued by prior genistein treatment. Genistein stimulated the activation of multiple endoplasmic reticulum (ER) stress-related regulators like GRP78, PERK, eIF2α, and upregulated the expression of the apoptotic genes, caspase 12 and CHOP. Moreover, TUNEL assays showed that genistein treatment promoted acinar cell apoptosis. Taken together, we speculated that ER stress-associated apoptotic pathways in AP are induced by genistein, which showed cytoprotective capacity in the exocrine pancreas. These data suggest novel therapeutic strategies that employ genistein in the prevention of AP.
Collapse
Affiliation(s)
- Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| | - Jian Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200030, PR China.
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, 3052, Australia.
| | - Wei Zhang
- Department of Gastroenterology, Huadong Hospital, Fudan University, 200040, Shanghai, PR China.
| | - Yun Zhao
- Department of Emergency Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
7
|
Management of Acute Pancreatitis in the Pediatric Population: A Clinical Report From the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition Pancreas Committee. J Pediatr Gastroenterol Nutr 2018; 66:159-176. [PMID: 29280782 PMCID: PMC5755713 DOI: 10.1097/mpg.0000000000001715] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although the incidence of acute pancreatitis (AP) in children is increasing, management recommendations rely on adult published guidelines. Pediatric-specific recommendations are needed. METHODS The North American Society for Pediatric Gastroenterology, Hepatology and Nutrition Pancreas committee performed a MEDLINE review using several preselected key terms relating to management considerations in adult and pediatric AP. The literature was summarized, quality of evidence reviewed, and statements of recommendations developed. The authorship met to discuss the evidence, statements, and voted on recommendations. A consensus of at least 75% was required to approve a recommendation. RESULTS The diagnosis of pediatric AP should follow the published INternational Study Group of Pediatric Pancreatitis: In Search for a CuRE definitions (by meeting at least 2 out of 3 criteria: (1) abdominal pain compatible with AP, (2) serum amylase and/or lipase values ≥3 times upper limits of normal, (3) imaging findings consistent with AP). Adequate fluid resuscitation with crystalloid appears key especially within the first 24 hours. Analgesia may include opioid medications when opioid-sparing measures are inadequate. Pulmonary, cardiovascular, and renal status should be closely monitored particularly within the first 48 hours. Enteral nutrition should be started as early as tolerated, whether through oral, gastric, or jejunal route. Little evidence supports the use of prophylactic antibiotics, antioxidants, probiotics, and protease inhibitors. Esophago-gastro-duodenoscopy, endoscopic retrograde cholangiopancreatography, and endoscopic ultrasonography have limited roles in diagnosis and management. Children should be carefully followed for development of early or late complications and recurrent attacks of AP. CONCLUSIONS This clinical report represents the first English-language recommendations for the management of pediatric AP. Future aims should include prospective multicenter pediatric studies to further validate these recommendations and optimize care for children with AP.
Collapse
|
8
|
Xiao J, Feng X, Huang XY, Huang Z, Huang Y, Li C, Li G, Nong S, Wu R, Huang Y, Long XD. Spautin-1 Ameliorates Acute Pancreatitis via Inhibiting Impaired Autophagy and Alleviating Calcium Overload. Mol Med 2016; 22:643-652. [PMID: 27579473 PMCID: PMC5082290 DOI: 10.2119/molmed.2016.00034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/11/2016] [Indexed: 01/04/2023] Open
Abstract
Acute pancreatitis is characterized by zymogen pre-activation. Severe inflammation caused by zymogen activation can eventually lead to multiple organ dysfunctions, which contributes to the high mortality rate of severe acute pancreatitis. However, there is no specific treatment available for acute pancreatitis therapy. Here, we show that spautin-1, which effectively inhibits autophagy flux, ameliorated the pathogenesis of acute pancreatitis induced by cerulein or L-Arginine. CaMKII phosphorylation due to cytosolic calcium oeverload was revealed in this paper. It was also demonstrated that autophagic protein aggregates degradation blockade accompanying with impaired autophagy correlated positively to intra acinar cells digestive aymogen activation sitimulated by cerulein or L-Arginine. The role of spautin-1 in ameliorating acute pancreatitis was shown here to be associated with impaired autophagy inhibition and Ca2+ overload alleviation. We provided a promising therapy for acute pancreatitis here through targeting both impaired autophagy and increased cytosolic calcium.
Collapse
Affiliation(s)
- Juan Xiao
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
- Western Guangxi Key Laboratory Cultivation Base of Microbial Infection Research, Baise, People’s Republic of China
| | - Xueping Feng
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Xiao-Ying Huang
- Western Guangxi Key Laboratory Cultivation Base of Microbial Infection Research, Baise, People’s Republic of China
| | - Zhongshi Huang
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
- Western Guangxi Key Laboratory Cultivation Base of Microbial Infection Research, Baise, People’s Republic of China
| | - Yanqiang Huang
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
- Western Guangxi Key Laboratory Cultivation Base of Microbial Infection Research, Baise, People’s Republic of China
| | - Chaogan Li
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Genliang Li
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Song Nong
- Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Ruoshi Wu
- Department of Pathology, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Yongzhi Huang
- Department of Pathology, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, People’s Republic of China
| | - Xi-Dai Long
- Department of Pathology, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, People’s Republic of China
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
9
|
Abstract
BACKGROUND Acute pancreatitis is a serious medical disorder with no current therapies directed to the molecular pathogenesis of the disorder. Inflammation, inappropriate intracellular activation of digestive enzymes, and parenchymal acinar cell death by necrosis are the critical pathophysiologic processes of acute pancreatitis. Thus, it is necessary to elucidate the key molecular signals that mediate these pathobiologic processes and develop new therapeutic strategies to attenuate the appropriate signaling pathways in order to improve outcomes for this disease. A novel serine/threonine protein kinase D (PKD) family has emerged as key participants in signal transduction, and this family is increasingly being implicated in the regulation of multiple cellular functions and diseases. METHODS This review summarizes recent findings of our group and others regarding the signaling pathway and the biological roles of the PKD family in pancreatic acinar cells. In particular, we highlight our studies of the functions of PKD in several key pathobiologic processes associated with acute pancreatitis in experimental models. RESULTS Our findings reveal that PKD signaling is required for NF-κB activation/inflammation, intracellular zymogen activation, and acinar cell necrosis in rodent experimental pancreatitis. Novel small-molecule PKD inhibitors attenuate the severity of pancreatitis in both in vitro and in vivo experimental models. Further, this review emphasizes our latest advances in the therapeutic application of PKD inhibitors to experimental pancreatitis after the initiation of pancreatitis. CONCLUSIONS These novel findings suggest that PKD signaling is a necessary modulator in key initiating pathobiologic processes of pancreatitis, and that it constitutes a novel therapeutic target for treatments of this disorder.
Collapse
Affiliation(s)
- Jingzhen Yuan
- West Los Angeles VA Healthcare Center, UCLA/VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Bldg 258, Rm 340, Los Angeles, CA, 90073, USA.
| | - Stephen J Pandol
- West Los Angeles VA Healthcare Center, UCLA/VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Bldg 258, Rm 340, Los Angeles, CA, 90073, USA
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Abstract
Acute pancreatitis is an inflammatory process of the pancreatic gland that eventually may lead to a severe systemic inflammatory response. A key event in pancreatic damage is the intracellular activation of NF-κB and zymogens, involving also calcium, cathepsins, pH disorders, autophagy, and cell death, particularly necrosis. This review focuses on the new role of redox signaling in acute pancreatitis. Oxidative stress and redox status are involved in the onset of acute pancreatitis and also in the development of the systemic inflammatory response, being glutathione depletion, xanthine oxidase activation, and thiol oxidation in proteins critical features of the disease in the pancreas. On the other hand, the release of extracellular hemoglobin into the circulation from the ascitic fluid in severe necrotizing pancreatitis enhances lipid peroxidation in plasma and the inflammatory infiltrate into the lung and up-regulates the HIF-VEGF pathway, contributing to the systemic inflammatory response. Therefore, redox signaling and oxidative stress contribute to the local and systemic inflammatory response during acute pancreatitis.
Collapse
|
11
|
Schick V, Scheiber JA, Mooren FC, Turi S, Ceyhan GO, Schnekenburger J, Sendler M, Schwaiger T, Omercevic A, Brandt CVD, Fluhr G, Domschke W, Krüger B, Mayerle J, Lerch MM. Effect of magnesium supplementation and depletion on the onset and course of acute experimental pancreatitis. Gut 2014; 63:1469-80. [PMID: 24277728 DOI: 10.1136/gutjnl-2012-304274] [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] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE High calcium concentrations are an established risk factor for pancreatitis. We have investigated whether increasing magnesium concentrations affect pathological calcium signals and premature protease activation in pancreatic acini, and whether dietary or intraperitoneal magnesium administration affects the onset and course of experimental pancreatitis. METHODS Pancreatic acini were incubated with up to 10 mM magnesium; [Ca(2+)](i) (fura-2AM) and intracellular protease activation (fluorogenic substrates) were determined over 60 min. Wistar rats received chow either supplemented or depleted for magnesium (<300 ppm to 30 000 ppm) over two weeks before pancreatitis induction (intravenous caerulein 10 µg/kg/h/4 h); controls received 1 µg/kg/h caerulein or saline. C57BL6/J mice received four intraperitoneal doses of magnesium (NaCl, Mg(2+) 55 192 or 384 mg/kg bodyweight) over 72 h, then pancreatitis was induced by up to eight hourly supramaximal caerulein applications. Pancreatic enzyme activities, protease activation, morphological changes and the immune response were investigated. RESULTS Increasing extracellular Mg(2+) concentration significantly reduced [Ca(2+)](i) peaks and frequency of [Ca(2+)](i) oscillations as well as intracellular trypsin and elastase activity. Magnesium administration reduced pancreatic enzyme activities, oedema, tissue necrosis and inflammation and somewhat increased Foxp3-positiv T-cells during experimental pancreatitis. Protease activation was found in animals fed magnesium-deficient chow-even with low caerulein concentrations that normally cause no damage. CONCLUSIONS Magnesium supplementation significantly reduces premature protease activation and the severity of pancreatitis, and antagonises pathological [Ca(2+)](i) signals. Nutritional magnesium deficiency increases the susceptibility of the pancreas towards pathological stimuli. These data have prompted two clinical trials on the use of magnesium in patients at risk for pancreatitis.
Collapse
Affiliation(s)
- Verena Schick
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany
| | - Jonas A Scheiber
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frank C Mooren
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany Department of Sports Medicine, Justus-Liebig-Universität, Giessen, Germany
| | - Stefan Turi
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Güralp O Ceyhan
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany Department of Surgery, Klinikum Rechts der Isar, Technische Universität, München, Germany
| | | | - Matthias Sendler
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Theresa Schwaiger
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Armin Omercevic
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | | | - Gabriele Fluhr
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Wolfram Domschke
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany
| | - Burkhard Krüger
- Division of Medical Biology, Universität Rostock, Rostock, Germany
| | - Julia Mayerle
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine B, Westfälische Wilhelms-Universität, Münster, Germany Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Athwal T, Huang W, Mukherjee R, Latawiec D, Chvanov M, Clarke R, Smith K, Campbell F, Merriman C, Criddle D, Sutton R, Neoptolemos J, Vlatković N. Expression of human cationic trypsinogen (PRSS1) in murine acinar cells promotes pancreatitis and apoptotic cell death. Cell Death Dis 2014; 5:e1165. [PMID: 24722290 PMCID: PMC5424103 DOI: 10.1038/cddis.2014.120] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 12/17/2013] [Accepted: 12/19/2013] [Indexed: 02/05/2023]
Abstract
Hereditary pancreatitis (HP) is an autosomal dominant disease that displays the features of both acute and chronic pancreatitis. Mutations in human cationic trypsinogen (PRSS1) are associated with HP and have provided some insight into the pathogenesis of pancreatitis, but mechanisms responsible for the initiation of pancreatitis have not been elucidated and the role of apoptosis and necrosis has been much debated. However, it has been generally accepted that trypsinogen, prematurely activated within the pancreatic acinar cell, has a major role in the initiation process. Functional studies of HP have been limited by the absence of an experimental system that authentically mimics disease development. We therefore developed a novel transgenic murine model system using wild-type (WT) human PRSS1 or two HP-associated mutants (R122H and N29I) to determine whether expression of human cationic trypsinogen in murine acinar cells promotes pancreatitis. The rat elastase promoter was used to target transgene expression to pancreatic acinar cells in three transgenic strains that were generated: Tg(Ela-PRSS1)NV, Tg(Ela-PRSS1*R122H)NV and Tg(Ela-PRSS1*N29I)NV. Mice were analysed histologically, immunohistochemically and biochemically. We found that transgene expression is restricted to pancreatic acinar cells and transgenic PRSS1 proteins are targeted to the pancreatic secretory pathway. Animals from all transgenic strains developed pancreatitis characterised by acinar cell vacuolisation, inflammatory infiltrates and fibrosis. Transgenic animals also developed more severe pancreatitis upon treatment with low-dose cerulein than controls, displaying significantly higher scores for oedema, inflammation and overall histopathology. Expression of PRSS1, WT or mutant, in acinar cells increased apoptosis in pancreatic tissues and isolated acinar cells. Moreover, studies of isolated acinar cells demonstrated that transgene expression promotes apoptosis rather than necrosis. We therefore conclude that expression of WT or mutant human PRSS1 in murine acinar cells induces apoptosis and is sufficient to promote spontaneous pancreatitis, which is enhanced in response to cellular insult.
Collapse
Affiliation(s)
- T Athwal
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
| | - W Huang
- Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
- Sichuan Provincial Pancreatitis Centre, West China Hospital, Sichuan University, Chengdu, China
| | - R Mukherjee
- Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
| | - D Latawiec
- Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
| | - M Chvanov
- Department of Cellular and Molecular Physiology, Institute for Translational Medicine, University of Liverpool, Liverpool, UK
| | - R Clarke
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
| | - K Smith
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
| | - F Campbell
- Department of Pathology, Royal Liverpool University Hospital, Liverpool, UK
| | - C Merriman
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
| | - D Criddle
- Department of Cellular and Molecular Physiology, Institute for Translational Medicine, University of Liverpool, Liverpool, UK
| | - R Sutton
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
- Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
| | - J Neoptolemos
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
- Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
| | - N Vlatković
- Department of Molecular and Clinical Cancer Medicine, Institute for Translational Medicine, University of Liverpool, Cancer Research Centre, Liverpool, UK
| |
Collapse
|
13
|
Chaiqinchengqi decoction regulates necrosis-apoptosis via regulating the release of mitochondrial cytochrome c and caspase-3 in rats with acute necrotizing pancreatitis. J TRADIT CHIN MED 2014; 34:178-83. [DOI: 10.1016/s0254-6272(14)60075-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
14
|
Yeh FY, Tseng IH, Chuang SH, Lin CS. Spacer-enhanced chymotrypsin-activated peptide-functionalized gold nanoparticle probes: a rapid assay for the diagnosis of pancreatitis. RSC Adv 2014; 4:22266-22276. [DOI: 10.1039/c4ra00258j] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A spacer-enhanced FITC-labeled peptide self-assembled onto AuNPs was fabricated as a chymotrypsin activated fluorescent AuNP probe and was used for the diagnosis of pancreatitis with fecal specimens.
Collapse
Affiliation(s)
- Fang-Yuan Yeh
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu 300, Taiwan
| | - I-Hua Tseng
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu 300, Taiwan
| | - Shu-Hung Chuang
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu 300, Taiwan
- Department of Surgery
- Mackay Memorial Hospital
| | - Chih-Sheng Lin
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu 300, Taiwan
| |
Collapse
|
15
|
Ma B, Wu L, Lu M, Gao B, Qiao X, Sun B, Xue D, Zhang W. Differentially expressed kinase genes associated with trypsinogen activation in rat pancreatic acinar cells treated with taurolithocholic acid 3-sulfate. Mol Med Rep 2013; 7:1591-6. [PMID: 23467886 DOI: 10.3892/mmr.2013.1355] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 02/27/2013] [Indexed: 11/05/2022] Open
Abstract
Trypsinogen activation is the initial factor involved in the development of all types of acute pancreatitis (AP) and has been suggested to be regulated by protein kinases. In the present study, AR42J rat pancreatic acinar cells were treated with taurolithocholic acid 3-sulfate (TLC-S), and trypsinogen activation was detected with bis-(CBZ-L-isoleucyl-L-prolyl-L-arginine amide) dihydrochloride (BZiPAR) staining and flow cytometry. Differentially expressed protein kinase genes were screened by Gene Chip analysis, and the functions of these kinases were analyzed. A significantly increased activation of trypsinogen in AR42J cells following treatment with TLC-S was observed. A total of 22 differentially expressed protein kinase genes were found in the TLC-S group, among which 19 genes were upregulated and 3 were downregulated. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, kinase genes of the same KEGG pathways were connected to create a network through signaling pathways, and 10 nodes of kinases were identified, which were mitogen-activated protein kinase (Mapk)8, Mapk14, Map2k4, interleukin-1 receptor-associated kinase 3 (Irak3), ribosomal protein S6 kinase, 90 kDa, polypeptide 2 (Rps6ka2), protein kinase C, alpha (Prkca), v-yes-1 Yamaguchi sarcoma viral related oncogene homolog (Lyn), protein tyrosine kinase 2 beta (Ptk2b), p21 protein (Cdc42/Rac)-activated kinase 4 (Pak4) and FYN oncogene related to SRC, FGR, YES (Fyn). The interactions between signaling pathways were further analyzed and a network was created. MAPK and calcium signaling pathways were found to be located at the center of the network. Thus, protein kinases constitute potential drug targets for AP treatment.
Collapse
Affiliation(s)
- Biao Ma
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Acute pancreatitis is an inflammatory disease of the exocrine pancreas that carries considerable morbidity and mortality; its pathophysiology remains poorly understood. Recent findings from experimental models and genetically altered mice summarized in this review reveal that autophagy, the principal cellular degradative pathway, is impaired in pancreatitis and that one cause of autophagy impairment is defective function of lysosomes. We propose that the lysosomal/autophagic dysfunction is a key initiating event in pancreatitis and a converging point of multiple deranged pathways. There is strong evidence supporting this hypothesis. Investigation of autophagy in pancreatitis has just started, and many questions about the "upstream" mechanisms mediating the lysosomal/autophagic dysfunction and the "downstream" links to pancreatitis pathologies need to be explored. Answers to these questions should provide insight into novel molecular targets and therapeutic strategies for treatment of pancreatitis.
Collapse
Affiliation(s)
- Anna S. Gukovskaya
- Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California
| | - Ilya Gukovsky
- Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
17
|
Lee MG, Ohana E, Park HW, Yang D, Muallem S. Molecular mechanism of pancreatic and salivary gland fluid and HCO3 secretion. Physiol Rev 2012; 92:39-74. [PMID: 22298651 DOI: 10.1152/physrev.00011.2011] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fluid and HCO(3)(-) secretion is a vital function of all epithelia and is required for the survival of the tissue. Aberrant fluid and HCO(3)(-) secretion is associated with many epithelial diseases, such as cystic fibrosis, pancreatitis, Sjögren's syndrome, and other epithelial inflammatory and autoimmune diseases. Significant progress has been made over the last 20 years in our understanding of epithelial fluid and HCO(3)(-) secretion, in particular by secretory glands. Fluid and HCO(3)(-) secretion by secretory glands is a two-step process. Acinar cells secrete isotonic fluid in which the major salt is NaCl. Subsequently, the duct modifies the volume and electrolyte composition of the fluid to absorb the Cl(-) and secrete HCO(3)(-). The relative volume secreted by acinar and duct cells and modification of electrolyte composition of the secreted fluids varies among secretory glands to meet their physiological functions. In the pancreas, acinar cells secrete a small amount of NaCl-rich fluid, while the duct absorbs the Cl(-) and secretes HCO(3)(-) and the bulk of the fluid in the pancreatic juice. Fluid secretion appears to be driven by active HCO(3)(-) secretion. In the salivary glands, acinar cells secrete the bulk of the fluid in the saliva that is driven by active Cl(-) secretion and contains high concentrations of Na(+) and Cl(-). The salivary glands duct absorbs both the Na(+) and Cl(-) and secretes K(+) and HCO(3)(-). In this review, we focus on the molecular mechanism of fluid and HCO(3)(-) secretion by the pancreas and salivary glands, to highlight the similarities of the fundamental mechanisms of acinar and duct cell functions, and to point out the differences to meet gland-specific secretions.
Collapse
Affiliation(s)
- Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Despite being a subject of much scientific scrutiny, the pathogenesis of acute pancreatitis is still not well understood. This article reviews recent advances in our understanding of acute pancreatitis. RECENT FINDINGS Zymogen activation, observed within acini early during acute pancreatitis for a long time, was shown to be sufficient to induce acute pancreatitis. Another key early event, NFκB activation, has previously been shown to induce acute pancreatitis. The relationship between these two key early steps is beginning to be clarified. Mechanisms of zymogen activation - pathologic calcium signaling, pH changes, colocalization and autophagy, and of NFκB activation have been investigated intensively along with potential therapeutic targets both upstream and downstream of these key events. Additional key findings have been elucidation of the role of bioenergetics and the dual role of oxidative stress in acute pancreatitis, recognition of endoplasmic reticulum stress as an early step and the status of duct cells as important entities in pancreatic injury. SUMMARY Current findings have provided further insight into the roles and mechanisms of zymogen activation and inflammatory pathways in pancreatic injury. Future studies, which will be of great importance in identifying therapeutic targets, are being undertaken to establish the relative contributions of these pathways during acute pancreatitis.
Collapse
|
19
|
Abstract
OBJECTIVES The objective of this study was to assess the reporting of studies on new prognostic markers of outcome in acute pancreatitis. METHODS We used MEDLINE searches complemented with perusal of review articles' references to identify eligible English-language studies. We included studies evaluating nonroutine markers for acute pancreatitis. Eligible outcomes included Atlanta criteria, Japanese criteria for severity, multiple/single organ failure, complications, interventional treatment, hospitalization length, and death. We generated a 47-item checklist on Acute Pancreatitis Prognosis by adapting a previously constructed reporting guidance instrument for prognostic tumor markers (REMARK [Reporting Recommendations for Tumor Marker Prognostic Studies]). The checklist addresses the reporting of essential information in prognostic studies. RESULTS The 184 identified eligible studies reported on 196 different prognostic markers. One hundred forty-four studies (78.3%) found at least 1 prognostic marker to be nominally statistically significant. Significant improvements over time were seen in the reporting for 17 items, but major deficiencies were noted even in 2004-2009 studies. Particularly, 12 items were reported in less than 10% of studies overall and even within the most recent studies. CONCLUSIONS Despite some improvements over time, the reporting of important aspects of prognostic studies in acute pancreatitis remains suboptimal. The proposed REMARK-based checklist may help improve the quality and reporting of research in this field.
Collapse
|
20
|
Sarmiento N, Sánchez-Yagüe J, Juanes PP, Pérez N, Ferreira L, García-Hernández V, Mangas A, Calvo JJ, Sánchez-Bernal C. Changes in the morphology and lability of lysosomal subpopulations in caerulein-induced acute pancreatitis. Dig Liver Dis 2011; 43:132-8. [PMID: 20934396 DOI: 10.1016/j.dld.2010.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 08/04/2010] [Accepted: 08/21/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Lysosomes play an important role in acute pancreatitis (AP). Here we developed a method for the isolation of lysosome subpopulations from rat pancreas and assessed the stability of lysosomal membranes. METHODS AP was induced by four subcutaneous injections of 20 μg caerulein/kg body weight at hourly intervals. The animals were killed 9h after the first injection. Marker enzymes [N-acetyl-β-D-glucosaminidase (NAG), cathepsin B and succinate dehydrogenase (SDH)] were assayed in subcellular fractions from control pancreas and in pancreatitis. Lysosomal subpopulations were separated by Percoll density gradient centrifugation and observed by electron microscopy. NAG molecular forms were determined by DEAE-cellulose chromatography. RESULTS AP was associated with: (i) increases in the specific activity of lysosomal enzymes in the soluble fraction, (ii) changes in the size and alterations in the morphology of the organelles from the lysosomal subpopulations, (iii) the appearance of large vacuoles in the primary and secondary lysosome subpopulations, (iv) the increase in the amount of the NAG form associated with the pancreatic lysosomal membrane as well as its release towards the soluble fraction. CONCLUSIONS Lysosome subpopulations are separated by a combination of differential and Percoll density gradient centrifugations. Primary lysosome membrane stability decreases in AP.
Collapse
Affiliation(s)
- Nancy Sarmiento
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Voronina S, Barrow S, Simpson A, Gerasimenko O, da Silva Xavier G, Rutter G, Petersen O, Tepikin AV. Dynamic changes in cytosolic and mitochondrial ATP levels in pancreatic acinar cells. Gastroenterology 2010; 138:1976-87. [PMID: 20102715 PMCID: PMC6101211 DOI: 10.1053/j.gastro.2010.01.037] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2009] [Revised: 12/17/2009] [Accepted: 01/21/2010] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Previous studies of pancreatic acinar cells characterized the effects of Ca(2+)-releasing secretagogues and substances, inducing acute pancreatitis on mitochondrial Ca(2+), transmembrane potential, and NAD(P)H, but dynamic measurements of the crucial intracellular adenosine triphosphate (ATP) levels have not been reported. Here we characterized the effects of these agents on ATP levels in the cytosol and mitochondria. METHODS ATP levels were monitored using cytosolic- or mitochondrial-targeted luciferases. RESULTS Inhibition of oxidative phosphorylation produced a substantial decrease in cytosolic ATP comparable to that induced by inhibition of glycolysis. Cholecystokinin-8 (CCK) increased cytosolic ATP in spite of accelerating ATP consumption. Acetylcholine, caerulein, and bombesin had similar effect. A bile acid, taurolithocholic acid 3-sulfate (TLC-S); a fatty acid, palmitoleic acid (POA); and palmitoleic acid ethyl ester (POAEE) reduced cytosolic ATP. The ATP decrease in response to these substances was observed in cells with intact or inhibited oxidative phosphorylation. TLC-S, POA, and POAEE reduced mitochondrial ATP, whereas physiological CCK increased mitochondrial ATP. Supramaximal CCK produced a biphasic response composed of a small initial decline followed by a stronger increase. CONCLUSIONS Both glycolysis and oxidative phosphorylation make substantial contributions to ATP production in acinar cells. Ca(2+)-releasing secretagogues increased ATP level in the cytosol and mitochondria of intact isolated cells. TLC-S, POA, and POAEE reduced cytosolic and mitochondrial ATP. When cells rely on nonoxidative ATP production, secretagogues as well as TLC-S, POA, and POAEE all diminish cytosolic ATP levels.
Collapse
Affiliation(s)
- Svetlana Voronina
- The Physiological Laboratory, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Stephanie Barrow
- The Physiological Laboratory, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Alec Simpson
- Department of Human Anatomy and Cell Biology, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Oleg Gerasimenko
- The Physiological Laboratory, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Gabriela da Silva Xavier
- Section of Cell Biology, Division of Medicine, Imperial College, London, Sir Alexander Fleming Building, Exhibition Road, London SW7 2AZ, UK
| | - Guy Rutter
- Section of Cell Biology, Division of Medicine, Imperial College, London, Sir Alexander Fleming Building, Exhibition Road, London SW7 2AZ, UK
| | - Ole Petersen
- The Physiological Laboratory, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Alexei V. Tepikin
- The Physiological Laboratory, School of Biomedical Sciences, The University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| |
Collapse
|
22
|
Mareninova OA, Hermann K, French SW, O'Konski MS, Pandol SJ, Webster P, Erickson AH, Katunuma N, Gorelick FS, Gukovsky I, Gukovskaya AS. Impaired autophagic flux mediates acinar cell vacuole formation and trypsinogen activation in rodent models of acute pancreatitis. J Clin Invest 2009; 119:3340-55. [PMID: 19805911 DOI: 10.1172/jci38674] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 07/01/2009] [Indexed: 12/11/2022] Open
Abstract
The pathogenic mechanisms underlying acute pancreatitis are not clear. Two key pathologic acinar cell responses of this disease are vacuole accumulation and trypsinogen activation. We show here that both result from defective autophagy, by comparing the autophagic responses in rodent models of acute pancreatitis to physiologic autophagy triggered by fasting. Pancreatitis-induced vacuoles in acinar cells were greater in number and much larger than those induced with fasting. Degradation of long-lived proteins, a measure of autophagic efficiency, was markedly inhibited in in vitro pancreatitis, while it was stimulated by acinar cell starvation. Further, processing of the lysosomal proteases cathepsin L (CatL) and CatB into their fully active, mature forms was reduced in pancreatitis, as were their activities in the lysosome-enriched subcellular fraction. These findings indicate that autophagy is retarded in pancreatitis due to deficient lysosomal degradation caused by impaired cathepsin processing. Trypsinogen activation occurred in pancreatitis but not with fasting and was prevented by inhibiting autophagy. A marker of trypsinogen activation partially localized to autophagic vacuoles, and pharmacologic inhibition of CatL increased the amount of active trypsin in acinar cells. The results suggest that retarded autophagy is associated with an imbalance between CatL, which degrades trypsinogen and trypsin, and CatB, which converts trypsinogen into trypsin, resulting in intra-acinar accumulation of active trypsin in pancreatitis. Thus, deficient lysosomal degradation may be a dominant mechanism for increased intra-acinar trypsin in pancreatitis.
Collapse
Affiliation(s)
- Olga A Mareninova
- Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California 90073, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Change of zinc, copper, and metallothionein concentrations and the copper-zinc superoxide dismutase activity in patients with pancreatitis. Pancreas 2009; 38:681-8. [PMID: 19629005 DOI: 10.1097/mpa.0b013e3181a53d1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The aims of the present studies were to measure the concentrations of zinc (Zn), copper (Cu), and metallothionein and the Cu/Zn superoxide dismutase activity as elements engaged in an essential manner in the prooxidative and antioxidative balance of organism and to demonstrate the degree to which metallothionein and Cu/Zn superoxide dismutase are involved in the inflammatory processes occurring in the pancreas. METHODS The concentration of metallothionein was measured by immunoenzymatic method. Serum Cu/Zn superoxide dismutase activity was determined using a commercial test. The measurements of Zn and Cu concentrations in serum were assessed with the use of flame atomic absorption spectrometry. RESULTS Lowered serum Zn concentration and higher Cu level were observed in the serum of patients with chronic exacerbated pancreatitis and chronic pancreatitis. The significant increase of metallothionein concentration and Cu/Zn superoxide dismutase activity was observed in the blood of patients with chronic exacerbated pancreatitis and chronic pancreatitis. In slices of the pancreas during pancreatitis, we observed in immunohistochemical reaction the variable involvement of Cu/Zn superoxide dismutase and metallothionein. CONCLUSIONS The results presented in these studies indicate an essential and variable involvement of antioxidants such Cu/Zn superoxide dismutase and metallothionein and disordered Cu and Zn homeostasis depending on the progression of inflammatory processes in patients with pancreatitis.
Collapse
|
24
|
Odinokova IV, Sung KF, Mareninova OA, Hermann K, Gukovsky I, Gukovskaya AS. Mitochondrial mechanisms of death responses in pancreatitis. J Gastroenterol Hepatol 2008; 23 Suppl 1:S25-30. [PMID: 18336659 DOI: 10.1111/j.1440-1746.2007.05271.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pancreatitis is a severe and frequently lethal disorder, a major cause of which is alcohol abuse. Parenchymal cell death is a major complication of pancreatitis. In experimental models of acute pancreatitis, acinar cells have been shown to die through both necrosis and apoptosis, the two principal pathways of cell death. The severity of experimental acute pancreatitis correlates directly with the extent of necrosis and inversely with apoptosis. Thus, understanding the regulation of apoptosis and necrosis is becoming exceedingly important in investigations of the pathogenesis and treatment of pancreatitis. Over the past decade, the mitochondria have emerged as a master regulator of cell death in various physiological and pathological processes. Release of mitochondrial cytochrome c into the cytosol is a central event in apoptosis, whereas mitochondrial depolarization resulting in ATP depletion leads to necrosis. The present review focuses on the mitochondrial mechanisms of death responses in pancreatitis, with emphasis on mitochondrial membrane permeabilization and its role in the balance between apoptosis and necrosis in acute pancreatitis, and alcohol's effects on death responses of pancreatitis.
Collapse
Affiliation(s)
- Irina V Odinokova
- USC-UCLA Research Center for Alcoholic Liver and Pancreatic Diseases, VA Greater Los Angeles Healthcare System and University of California, Los Angeles, California 90073, USA
| | | | | | | | | | | |
Collapse
|
25
|
Marshall JC. Acute Pancreatitis. Crit Care Med 2008. [DOI: 10.1016/b978-032304841-5.50080-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
26
|
Eckerwall GE, Tingstedt BBA, Bergenzaun PE, Andersson RG. Immediate oral feeding in patients with mild acute pancreatitis is safe and may accelerate recovery--a randomized clinical study. Clin Nutr 2007; 26:758-763. [PMID: 17719703 DOI: 10.1016/j.clnu.2007.04.007] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 03/12/2007] [Accepted: 04/18/2007] [Indexed: 01/08/2023]
Abstract
BACKGROUND & AIMS In acute pancreatitis, traditional treatment has been initial fasting on purpose to avoid activation of proteolytic enzymes and pancreatic enzyme secretion. The aim of the present study was to evaluate the efficacy and feasibility of immediate oral feeding as compared to traditional fasting in patients with mild acute pancreatitis. METHODS Sixty patients were randomized to the two treatment groups, fasting or immediate oral feeding. The inclusion criteria were pancreas amylase > or =3 times above normal, onset of abdominal pain within 48 h, acute physiological and chronic health evaluation (APACHE) score<8 and C-reactive protein (CRP) <150 mg/L. Outcome measures were pancreas-specific amylase, systemic inflammatory response, feasibility and length of hospital stay (LOHS). RESULTS The groups were comparable with respect to age, sex, etiology, APACHE, time from onset of pain and amylase at admission. No significant differences were seen between the groups concerning levels of amylase, CRP, leukocytes, abdominal pain or number of gastrointestinal symptoms. The LOHS was significantly shorter in the oral feeding group (4 vs. 6 days; p<0.05). CONCLUSIONS No signs of exacerbation of the disease process were seen in terms of significant differences between treatment groups for amylase or systemic inflammatory response. In mild acute pancreatitis, immediate oral feeding was feasible and safe and may accelerate recovery without adverse gastrointestinal events.
Collapse
Affiliation(s)
- Gunilla E Eckerwall
- Department of Surgery, Clinical Sciences Lund, Lund University Hospital, S-221 85 Lund, Sweden
| | | | | | | |
Collapse
|
27
|
Letoha T, Fehér LZ, Pecze L, Somlai C, Varga I, Kaszaki J, Tóth G, Vizler C, Tiszlavicz L, Takács T. Therapeutic proteasome inhibition in experimental acute pancreatitis. World J Gastroenterol 2007; 13:4452-7. [PMID: 17724800 PMCID: PMC4611577 DOI: 10.3748/wjg.v13.i33.4452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish the therapeutic potential of proteasome inhibition, we examined the therapeutic effects of MG132 (Z-Leu-Leu-Leu-aldehyde) in an experimental model of acute pancreatitis.
METHODS: Pancreatitis was induced in rats by two hourly intraperitoneal (ip) injections of cholecystokinin octapeptide (CCK; 2 × 100 μg/kg) and the proteasome inhibitor MG132 (10 mg/kg ip) was administered 30 min after the second CCK injection. Animals were sacrificed 4 h after the first injection of CCK.
RESULTS: Administering the proteasome inhibitor MG132 (at a dose of 10 mg/kg, ip) 90 min after the onset of pancreatic inflammation induced the expression of cell-protective 72 kDa heat shock protein (HSP72) and decreased DNA-binding of nuclear factor-κB (NF-κB).
Furthermore MG132 treatment resulted in milder inflammatory response and cellular damage, as revealed by improved laboratory and histological parameters of pancreatitis and associated oxidative stress.
CONCLUSION: Our findings suggest that proteasome inhibition might be beneficial not only for the prevention, but also for the therapy of acute pancreatitis.
Collapse
Affiliation(s)
- Tamás Letoha
- Department of Medical Chemistry, University of Szeged, Dom ter 8, H-6720, Szeged, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Baumann B, Wagner M, Aleksic T, von Wichert G, Weber CK, Adler G, Wirth T. Constitutive IKK2 activation in acinar cells is sufficient to induce pancreatitis in vivo. J Clin Invest 2007; 117:1502-13. [PMID: 17525799 PMCID: PMC1868787 DOI: 10.1172/jci30876] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Accepted: 03/20/2007] [Indexed: 01/01/2023] Open
Abstract
Activation of the inhibitor of NF-kappaB kinase/NF-kappaB (IKK/NF-kappaB) system and expression of proinflammatory mediators are major events in acute pancreatitis. However, the in vivo consequences of IKK activation on the onset and progression of acute pancreatitis remain unclear. Therefore, we modulated IKK activity conditionally in pancreatic acinar cells. Transgenic mice expressing the reverse tetracycline-responsive transactivator (rtTA) gene under the control of the rat elastase promoter were generated to mediate acinar cell-specific expression of IKK2 alleles. Expression of dominant-negative IKK2 ameliorated cerulein-induced pancreatitis but did not affect activation of trypsin, an initial event in experimental pancreatitis. Notably, expression of constitutively active IKK2 was sufficient to induce acute pancreatitis. This acinar cell-specific phenotype included edema, cellular infiltrates, necrosis, and elevation of serum lipase levels as well as pancreatic fibrosis. IKK2 activation caused increased expression of known NF-kappaB target genes, including mediators of the inflammatory response such as TNF-alpha and ICAM-1. Indeed, inhibition of TNF-alpha activity identified this cytokine as an important effector of IKK2-induced pancreatitis. Our data identify the IKK/NF-kappaB pathway in acinar cells as being key to the development of experimental pancreatitis and the major factor in the inflammatory response typical of this disease.
Collapse
Affiliation(s)
- Bernd Baumann
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Martin Wagner
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Tamara Aleksic
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Götz von Wichert
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Christoph K. Weber
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Guido Adler
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry and
Department of Internal Medicine I, University of Ulm, Ulm, Germany
| |
Collapse
|
29
|
Archer H, Jura N, Keller J, Jacobson M, Bar-Sagi D. A mouse model of hereditary pancreatitis generated by transgenic expression of R122H trypsinogen. Gastroenterology 2006; 131:1844-55. [PMID: 17087933 DOI: 10.1053/j.gastro.2006.09.049] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Accepted: 09/05/2006] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Missense mutations in human cationic trypsinogen PRSS1 are frequently detected in patients with hereditary pancreatitis, a rare genetic disease of the pancreas characterized by autodigestive necrosis, chronic inflammation, and fibrosis. To examine the link between PRSS1 mutations and the initiation and progression of hereditary pancreatitis, we have sought to generate a transgenic mouse that carries a missense mutation in the PRSS1 that is most frequently observed in patients. METHODS A transgenic mouse was generated in which the expression of the mouse PRSS1 mutant R122H (R122H_mPRSS1) is targeted to pancreatic acinar cells by fusion to the elastase promoter. The expression of the mutant trypsinogen was assessed by immunohistochemical staining and real-time reverse transcription polymerase chain reaction analysis. The relationship between transgene expression and inflammation was analyzed by morphologic assessment of H&E-stained tissue sections, responsiveness to cerulein-induced pancreatitis, and immunohistochemical identification of cellular and biochemical components of the inflammatory response. RESULTS Pancreata from transgenic mice display early-onset acinar cell injury and inflammatory cell infiltration. With progressing age, the transgenic mice develop pancreatic fibrosis and display acinar cell dedifferentiation. Moreover, the expression of R122H_mPRSS1 transgene is associated with enhanced response to cerulein-induced pancreatitis. Finally, cell-specific activation of the inflammation-associated signaling pathways, c-jun-N-terminal kinase and extracellular signal-regulated kinase, was observed in response to expression of R122H_mPRSS1. CONCLUSIONS These results underscore the importance of PRSS1 mutations as pathogenic mediators of hereditary pancreatitis and indicate that persistent pancreatic injury might be causally linked to chronic pancreatitis.
Collapse
Affiliation(s)
- Herbert Archer
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York, USA
| | | | | | | | | |
Collapse
|
30
|
Letoha T, Somlai C, Takács T, Szabolcs A, Rakonczay Z, Jármay K, Szalontai T, Varga I, Kaszaki J, Boros I, Duda E, Hackler L, Kurucz I, Penke B. The proteasome inhibitor MG132 protects against acute pancreatitis. Free Radic Biol Med 2005; 39:1142-51. [PMID: 16214030 DOI: 10.1016/j.freeradbiomed.2005.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 06/07/2005] [Accepted: 06/09/2005] [Indexed: 02/06/2023]
Abstract
The cell-permeant MG132 tripeptide (Z-Leu-Leu-Leu-aldehyde) is a peptide aldehyde proteasome inhibitor that also inhibits other proteases, including calpains and cathepsins. By blocking the proteasome, this tripeptide has been shown to induce the expression of cell-protective heat shock proteins (HSPs) in vitro. Effects of MG132 were studied in an in vivo model of acute pancreatitis. Pancreatitis was induced in male Wistar rats by injecting 2 x 100 microug/kg cholecystokinin octapeptide intraperitoneally (ip) at an interval of 1 h. Pretreating the animals with 10 mg/kg MG132 ip before the induction of pancreatitis significantly inhibited IkappaB degradation and subsequent activation of nuclear factor-kappaB (NF-kappaB). MG132 also increased HSP72 expression. Induction of HSP72 and inhibition of NF-kappaB improved parameters of acute pancreatitis. Thus MG132 significantly decreased serum amylase, pancreatic weight/body weight ratio, pancreatic myeloperoxidase activity, proinflammatory cytokine concentrations, and the expression of pancreatitis-associated protein. Parameters of oxidative stress (GSH, MDA, SOD, etc.) were improved in both the serum and the pancreas. Histopathological examinations revealed that pancreatic specimens of animals pretreated with the peptide demonstrated milder edema, cellular damage, and inflammatory activity. Our findings show that simultaneous inhibition of calpains, cathepsins, and the proteasome with MG132 prevents the onset of acute pancreatitis.
Collapse
Affiliation(s)
- Tamás Letoha
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|