1
|
Gagnani R, Srivastava M, Suri M, Singh H, Shanker Navik U, Bali A. A focus on c-Jun-N-terminal kinase signaling in sepsis-associated multiple organ dysfunction: Mechanisms and therapeutic strategies. Int Immunopharmacol 2024; 143:113552. [PMID: 39536486 DOI: 10.1016/j.intimp.2024.113552] [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] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Sepsis is a life-threatening condition characterized by a widespread inflammatory response to infection, inevitably leading to multiple organ dysfunctions. Extensive research, both in vivo and in vitro, has revealed key factors contributing to sepsis, such as apoptosis, inflammation, cytokine release, oxidative stress, and systemic stress. The changes observed during sepsis-induced conditions are mainly attributed to altered signal transduction pathways, which play a critical role in cell proliferation, migration, and apoptosis. C-Jun N-terminal kinases, JNKs, and serine/threonine protein kinases in the mitogen-activated super family have gained considerable interest for their contribution to cellular events under sepsis conditions. JNK1 and JNK2 are present in various tissues like the lungs, liver, and intestine, while JNK3 is found in neurons. The JNK pathway plays a crucial role in the signal transduction of cytokines related to sepsis development, notably TNF-α and IL-1β. Activated JNK leads to apoptosis, causing tissue damage and organ dysfunction. Further, JNK activation is significant in several inflammatory conditions. Pharmacologically inhibiting JNK has been shown to prevent sepsis-associated damage across multiple organs, including the lungs, liver, intestines, heart, and kidneys. Multiple signaling pathways have been implicated in sepsis, including JNK/c-Myc, Mst1-JNK, MKK4-JNK, JNK-dependent autophagy, and Sirt1/FoxO3a. The review examines the role of JNK signaling in the development of sepsis-induced multiple-organ dysfunction through specific mechanisms. It also discusses different therapeutic approaches to target JNK. This review emphasizes the potential of JNKs as targets for the development of therapeutic agents for sepsis and the associated specific organ damage.
Collapse
Affiliation(s)
- Riya Gagnani
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| | - Mukul Srivastava
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Manisha Suri
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Harshita Singh
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Uma Shanker Navik
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
2
|
Dey S, Udari LM, RiveraHernandez P, Kwon JJ, Willis B, Easler JJ, Fogel EL, Pandol S, Kota J. Loss of miR-29a/b1 promotes inflammation and fibrosis in acute pancreatitis. JCI Insight 2021; 6:e149539. [PMID: 34464354 PMCID: PMC8525644 DOI: 10.1172/jci.insight.149539] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
MicroRNA-29 (miR-29) is a critical regulator of fibroinflammatory processes in human diseases. In this study, we found a decrease in miR-29a in experimental and human chronic pancreatitis, leading us to investigate the regulatory role of the miR-29a/b1 cluster in acute pancreatitis (AP) utilizing a conditional miR-29a/b1-KO mouse model. miR-29a/b1-sufficient (WT) and -deficient (KO) mice were administered supramaximal caerulein to induce AP and characterized at different time points, utilizing an array of IHC and biochemical analyses for AP parameters. In caerulein-induced WT mice, miR-29a remained dramatically downregulated at injury. Despite high-inflammatory milieu, fibrosis, and parenchymal disarray in the WT mice during early AP, the pancreata fully restored during recovery. miR-29a/b1-KO mice showed significantly greater inflammation, lymphocyte infiltration, macrophage polarization, and ECM deposition, continuing until late recovery with persistent parenchymal disorganization. The increased pancreatic fibrosis was accompanied by enhanced TGFβ1 coupled with persistent αSMA+ PSC activation. Additionally, these mice exhibited higher circulating IL-6 and inflammation in lung parenchyma. Together, this collection of studies indicates that depletion of miR-29a/b1 cluster impacts the fibroinflammatory mechanisms of AP, resulting in (a) aggravated pathogenesis and (b) delayed recovery from the disease, suggesting a protective role of the molecule against AP.
Collapse
Affiliation(s)
- Shatovisha Dey
- Department of Medical and Molecular Genetics, Indiana University (IU) School of Medicine, Indianapolis, Indiana, USA
| | - Lata M Udari
- Department of Medical and Molecular Genetics, Indiana University (IU) School of Medicine, Indianapolis, Indiana, USA
| | - Primavera RiveraHernandez
- Department of Medical and Molecular Genetics, Indiana University (IU) School of Medicine, Indianapolis, Indiana, USA
| | - Jason J Kwon
- Department of Medical and Molecular Genetics, Indiana University (IU) School of Medicine, Indianapolis, Indiana, USA
| | | | - Jeffrey J Easler
- Department of Medicine, Division of Gastroenterology/Hepatology, IU Health, IU School of Medicine, Indianapolis, Indiana, USA.,The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, Indiana, USA
| | - Evan L Fogel
- Department of Medicine, Division of Gastroenterology/Hepatology, IU Health, IU School of Medicine, Indianapolis, Indiana, USA.,The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, Indiana, USA
| | - Stephen Pandol
- Department of Medicine, Cedar-Sinai Medical Center, Los Angeles, California, USA
| | - Janaiah Kota
- Department of Medical and Molecular Genetics, Indiana University (IU) School of Medicine, Indianapolis, Indiana, USA.,The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Gallo KA, Ellsworth E, Stoub H, Conrad SE. Therapeutic potential of targeting mixed lineage kinases in cancer and inflammation. Pharmacol Ther 2019; 207:107457. [PMID: 31863814 DOI: 10.1016/j.pharmthera.2019.107457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Dysregulation of intracellular signaling pathways is a key attribute of diseases associated with chronic inflammation, including cancer. Mitogen activated protein kinases have emerged as critical conduits of intracellular signal transmission, yet due to their ubiquitous roles in cellular processes, their direct inhibition may lead to undesired effects, thus limiting their usefulness as therapeutic targets. Mixed lineage kinases (MLKs) are mitogen-activated protein kinase kinase kinases (MAP3Ks) that interact with scaffolding proteins and function upstream of p38, JNK, ERK, and NF-kappaB to mediate diverse cellular signals. Studies involving gene silencing, genetically engineered mouse models, and small molecule inhibitors suggest that MLKs are critical in tumor progression as well as in inflammatory processes. Recent advances indicate that they may be useful targets in some types of cancer and in diseases driven by chronic inflammation including neurodegenerative diseases and metabolic diseases such as nonalcoholic steatohepatitis. This review describes existing MLK inhibitors, the roles of MLKs in various aspects of tumor progression and in the control of inflammatory processes, and the potential for therapeutic targeting of MLKs.
Collapse
Affiliation(s)
- Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Edmund Ellsworth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Hayden Stoub
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Susan E Conrad
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
4
|
Zhang X, Gao T, Wang Y. Geniposide alleviates lipopolysaccharide (LPS)-induced inflammation by downregulation of miR-27a in rat pancreatic acinar cell AR42J. Biol Chem 2019; 400:1059-1068. [PMID: 30897061 DOI: 10.1515/hsz-2018-0422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/15/2019] [Indexed: 01/17/2023]
Abstract
Pancreatitis is a disease caused by inflammation of pancreatic acinar cells. Geniposide (GEN) possesses anti-inflammation activities. Hence, we investigated the effects of GEN on lipopolysaccharide (LPS)-stimulated AR42J cells. AR42J cells were stimulated by LPS and then treated with GEN and/or transfected with miR-27a mimic or negative control. Cell viability and cell apoptosis were detected using the Cell Counting Kit-8 and flow cytometry, respectively. All related proteins were measured by Western blot. The expression of miR-27a was detected by quantitative real time-polymerase chain reaction (qRT-PCR). Moreover, the expression of inflammatory cytokines interleukin-6 (IL-6) and monocyte chemoattractant protein (MCP)-1 was analyzed by qRT-PCR and Western blot. LPS significantly decreased cell viability, and enhanced cell apoptosis and IL-6, MCP-1 expression. Then GEN administration alleviated inflammatory injury by increasing cell viability, while reducing apoptosis, and IL-6 and MCP-1 expression. GEN downregulated miR-27a expression which was induced by LPS. Transfection with miR-27a mimic partially eliminated the protective effects of GEN. The phosphorylation of JNK and c-Jun was downregulated by GEN while upregulated by miR-27a overexpression. GEN alleviates LPS-induced AR42J cell injury as evidenced by promoting cell growth, and upregulation of IL-6 and MCP-1. This process might be modulated by down-regulating miR-27a and inactivation of JNK pathway.
Collapse
Affiliation(s)
- Xiaofen Zhang
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
- Affiliated Jining No. 1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Taishan Gao
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
| | - Yanhua Wang
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
| |
Collapse
|
5
|
Gámez-Belmonte R, Hernández-Chirlaque C, Sánchez de Medina F, Martínez-Augustin O. Experimental acute pancreatitis is enhanced in mice with tissue nonspecific alkaline phoshatase haplodeficiency due to modulation of neutrophils and acinar cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3769-3779. [DOI: 10.1016/j.bbadis.2018.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/31/2018] [Accepted: 09/09/2018] [Indexed: 01/13/2023]
|
6
|
Yang Z, Yang W, Lu M, Li Z, Qiao X, Sun B, Zhang W, Xue D. Role of the c-Jun N-terminal kinase signaling pathway in the activation of trypsinogen in rat pancreatic acinar cells. Int J Mol Med 2017; 41:1119-1126. [PMID: 29207022 DOI: 10.3892/ijmm.2017.3266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Bile acid causes trypsinogen activation in pancreatic acinar cells through a complex process. Additional research is required to further elucidate which signaling pathways affect trypsinogen activation when activated. the changes in the whole‑genome expression profile of AR42J cells under the effect of taurolithocholic acid 3‑sulfate (TLC‑S) were investigated. Furthermore, gene groups that may play a regulatory role were analyzed using the modular approach of biological networks. The aim of the present study was to improve our understanding of the changes in TLC‑S‑stimulated AR42J cells through a genetic functional modular analysis. whole‑genome expression profile chip arrays were applied to detect genes that were differentially expressed in pancreatic acinar AR42J cells treated with TLC‑S for 20 min. Based on the human protein reference database, a protein‑protein interaction network was obtained, which was then processed by CFinder software to derive 14 modules. Among these 14 modules, the gene ontology biological processes enrichment analysis identified two as modules of interest. Kyoto encyclopedia of genes and genomes map analysis revealed that MAP2K4, MAPK8 and FLNA are part of the c-Jun N-terminal kinase (JNK) pathway. The JNK signaling pathway is involved in regulating trypsinogen activation in rat pancreatic AR42J cells. Next, a regulatory network of seven kinase inhibitors was constructed. SP600125 is an ATP‑competitive, efficient, selective and reversible inhibitor of JNK. the results were verified by four sets of experiments and demonstrated that trypsinogen activation is mediated by the JNK signaling pathway in the pathogenesis of acute pancreatitis (AP). The present study provided a useful reference for better understanding the pathogenesis of AP and identifying new targets to regulate trypsinogen activation, in addition to providing valuable information for the treatment of AP.
Collapse
Affiliation(s)
- Zhengpeng Yang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weiguang Yang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ming Lu
- Department of Surgery, David Geffen School of Medicine, University of Califonia at Los Angeles, Los Angeles, CA 90095, USA
| | - Zhituo Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xin Qiao
- Department of Surgery, David Geffen School of Medicine, University of Califonia at Los Angeles, Los Angeles, CA 90095, USA
| | - Bei Sun
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weihui Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
7
|
Han B, Zhou H, Jia G, Wang Y, Song Z, Wang G, Pan S, Bai X, Lv J, Sun B. MAPKs and Hsc70 are critical to the protective effect of molecular hydrogen during the early phase of acute pancreatitis. FEBS J 2016; 283:738-56. [PMID: 26683671 DOI: 10.1111/febs.13629] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/15/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022]
Abstract
Molecular hydrogen (H2 ) has been proven to be an effective agent that can cure multiple organ diseases by reducing oxidative stress. Although the protective effect of hydrogen on acute pancreatitis (AP) has been confirmed, its molecular mechanism is still unclear. In this article, we aimed to investigate the changes in pancreatic cell protein expression associated with the protective effect of H2 against AP and attempted to uncover the molecular mechanism underlying this process. A proteomic analysis identified 73 differentially expressed proteins and generated the protein-protein interaction networks of these proteins. The results triggered our interest in mitogen-activated protein kinase (MAPK) and heat shock cognate 71 kDa protein (Hsc70). The subsequent in vitro experiments showed that H2 treatment inhibited the phosphorylation of extracellular signal-regulated kinase (ERK), c-jun N-terminal kinase (JNK), and p38 MAPK, and activated NF-κB and the expression of tumor necrosis factor α and interleukin-1β, while simultaneously preventing the translocation of phospho-ERK, phospho-JNK, and phospho-p38 from the cytoplasm to the nucleus. Furthermore, Hsc70 expression was upregulated by H2 administration. The animal experimental results were consistent with those of the in vitro experiments. In conclusion, H2 treatment can ameliorate the inflammatory response and reduce the expression of inflammatory mediators during the early phase of AP by inhibiting the MAPK pathways and increasing Hsc70 expression.
Collapse
Affiliation(s)
- Bing Han
- Department of Hepatobiliary Surgery, Yantai Yuhuangding Hospital of Qingdao University Medical College, China.,Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Haoxin Zhou
- Department of Emergency Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Guang Jia
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Zengfu Song
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Shangha Pan
- Central Laboratory, First Affiliated Hospital of Harbin Medical University, China
| | - Xuewei Bai
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Jiachen Lv
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, China
| |
Collapse
|
8
|
Cerulein-induced acute pancreatitis is associated with c-Jun NH(2)-terminal kinase 1-dependent ferritin degradation and iron-dependent free radicals formation. Pancreas 2013; 42:1070-7. [PMID: 23921964 DOI: 10.1097/mpa.0b013e318287d097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVES The main goal of this work was to get insight into the mechanism of cerulein-induced reactive oxygen species (ROS) formation and impact of c-Jun NH(2)-terminal kinase (JNK) on this process. METHODS The study was performed on Wistar rats and on a cellular model of acute pancreatitis (AP) using AR42J cell line. RESULTS First of all, we observed that during AP, the iron storage protein ferritin in the rat pancreas undergoes degradation accompanied by an increased formation of protein carbonyls. Pancreatic acinar AR42J cells stimulated by cerulein showed increased labile iron pool that was accompanied by a decrease in the cellular ferritin-L level and an increase in the ROS formation. The changes in the ferritin-L level were inversely correlated with the ROS formation. The cells expressing inactive JNK1 mutant were completely resistant to cerulein-induced ferritin degradation. CONCLUSIONS Our data showed that cerulein-induced AP in rats and on cellular model is accompanied by JNK1-dependent ferritin degradation, increases labile iron pool and ROS formation.
Collapse
|
9
|
Feng Z, Fei J, Wenjian X, Jiachen J, Beina J, Zhonghua C, Xiangyi Y, Shaoying W. Rhubarb attenuates the severity of acute necrotizing pancreatitis by inhibiting MAPKs in rats. Immunotherapy 2013; 4:1817-21. [PMID: 23240749 DOI: 10.2217/imt.12.131] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM To investigate the effect of rhubarb on MAPK activation in sodium taurocholate-induced pancreatitis in rats. METHODS All rats were randomly divided into three groups: control, acute pancreatitis (AP) and rhubarb treatment groups. The MAPK activation was detected by western blotting. The levels of TNFα and IL6 in serum were estimated by ELISA. The pathological changes of the pancreas were examined microscopically. RESULTS It was found that hemorrhage, exudates and infiltration of inflammatory cells in the pancreas were significantly less in the rhubarb group than in the AP group. In the AP group, the activation of MAPKs in pancreatic tissues was enhanced significantly at every time point compared with the control group and the levels of TNFα and IL6 were also increased. Rhubarb treatment markedly inhibited activation of MAPKs concomitantly with inhibition of TNFα and IL6 expression. CONCLUSION We suggest that rhubarb probably attenuates AP by inhibiting activation of MAPKs and expression of inflammatory mediators.
Collapse
Affiliation(s)
- Zhen Feng
- Department of Gastroenterology, Central Hospital of Xuhui District, 966# HuaiHai Middle Road, Shanghai 200031, China
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Moens U, Kostenko S, Sveinbjørnsson B. The Role of Mitogen-Activated Protein Kinase-Activated Protein Kinases (MAPKAPKs) in Inflammation. Genes (Basel) 2013; 4:101-33. [PMID: 24705157 PMCID: PMC3899974 DOI: 10.3390/genes4020101] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 01/18/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways are implicated in several cellular processes including proliferation, differentiation, apoptosis, cell survival, cell motility, metabolism, stress response and inflammation. MAPK pathways transmit and convert a plethora of extracellular signals by three consecutive phosphorylation events involving a MAPK kinase kinase, a MAPK kinase, and a MAPK. In turn MAPKs phosphorylate substrates, including other protein kinases referred to as MAPK-activated protein kinases (MAPKAPKs). Eleven mammalian MAPKAPKs have been identified: ribosomal-S6-kinases (RSK1-4), mitogen- and stress-activated kinases (MSK1-2), MAPK-interacting kinases (MNK1-2), MAPKAPK-2 (MK2), MAPKAPK-3 (MK3), and MAPKAPK-5 (MK5). The role of these MAPKAPKs in inflammation will be reviewed.
Collapse
Affiliation(s)
- Ugo Moens
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, NO-9037 Tromsø, Norway.
| | - Sergiy Kostenko
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, NO-9037 Tromsø, Norway.
| | - Baldur Sveinbjørnsson
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, NO-9037 Tromsø, Norway.
| |
Collapse
|
11
|
Kostadinova R, Montagner A, Gouranton E, Fleury S, Guillou H, Dombrowicz D, Desreumaux P, Wahli W. GW501516-activated PPARβ/δ promotes liver fibrosis via p38-JNK MAPK-induced hepatic stellate cell proliferation. Cell Biosci 2012; 2:34. [PMID: 23046570 PMCID: PMC3519722 DOI: 10.1186/2045-3701-2-34] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/04/2012] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED BACKGROUND After liver injury, the repair process comprises activation and proliferation of hepatic stellate cells (HSCs), which produce extracellular matrix (ECM) proteins. Peroxisome proliferator-activated receptor beta/delta (PPARβ/δ) is highly expressed in these cells, but its function in liver repair remains incompletely understood. This study investigated whether activation of PPARβ/δ with the ligand GW501516 influenced the fibrotic response to injury from chronic carbon tetrachloride (CCl4) treatment in mice. Wild type and PPARβ/δ-null mice were treated with CCl4 alone or CCl4 co-administered with GW501516. To unveil mechanisms underlying the PPARβ/δ-dependent effects, we analyzed the proliferative response of human LX-2 HSCs to GW501516 in the presence or absence of PPARβ/δ. RESULTS We found that GW501516 treatment enhanced the fibrotic response. Compared to the other experimental groups, CCl4/GW501516-treated wild type mice exhibited increased expression of various profibrotic and pro-inflammatory genes, such as those involved in extracellular matrix deposition and macrophage recruitment. Importantly, compared to healthy liver, hepatic fibrotic tissues from alcoholic patients showed increased expression of several PPAR target genes, including phosphoinositide-dependent kinase-1, transforming growth factor beta-1, and monocyte chemoattractant protein-1. GW501516 stimulated HSC proliferation that caused enhanced fibrotic and inflammatory responses, by increasing the phosphorylation of p38 and c-Jun N-terminal kinases through the phosphoinositide-3 kinase/protein kinase-C alpha/beta mixed lineage kinase-3 pathway. CONCLUSIONS This study clarified the mechanism underlying GW501516-dependent promotion of hepatic repair by stimulating proliferation of HSCs via the p38 and JNK MAPK pathways.
Collapse
Affiliation(s)
- Radina Kostadinova
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of Lausanne, Genopode Building, 1015, Lausanne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Li YY, Ochs S, Gao ZR, Malo A, Chen CJ, Lv S, Gallmeier E, Göke B, Schäfer C. Regulation of HSP60 and the role of MK2 in a new model of severe experimental pancreatitis. Am J Physiol Gastrointest Liver Physiol 2009; 297:G981-9. [PMID: 20501446 DOI: 10.1152/ajpgi.00225.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The objective of this study was to investigate the role of MAPKAP kinase 2 (MK2) and heat shock protein (HSP) HSP60 in the pathogenesis of a new model of severe acute pancreatitis (AP). MK2 plays a significant role in the regulation of cytokines. It has been shown that induction and expression of several HSPs can protect against experimental pancreatitis. Interplay between both systems seems of high interest. Mice with a homozygous deletion of the MK2 gene were used. Severe AP was induced by combined intraperitoneal injections of cerulein with lipopolysaccharide (LPS). Severity of AP was assessed by biochemical markers and histology. The serum IL-6 and lung myeloperoxidase (MPO) levels were determined for assessing the extent of systemic inflammatory response. Expression of HSP25, HSP60, HSP70, and HSP90 was analyzed by Western blotting. Repeated injections of cerulein alone or cerulein plus LPS (Cer+LPS) resulted in local inflammatory responses in the pancreas and corresponding systemic inflammatory changes with pronounced severity in the Cer+LPS group. Compared with the C57Bl wild-type mice, the MK2-/- mice presented with significant milder pancreatitis and attenuated responses of serum amylase and trypsinogen activity. Furthermore, serum IL-6 was decreased as well as lung MPO activity. Injection of LPS alone displayed neither pancreatic inflammatory responses nor alterations of pancreatic enzyme activities but evidently elevated serum IL-6 levels and increased lung MPO activity. In contrast hereto, in the MK2-/- mice, these changes were much milder. Increased expression of HSP25 and HSP60 occurred after induction of AP. Especially, HSP60 was robustly elevated after Cer+LPS treatment, in both MK2-/- and wild-type mice. Thus the homozygous deletion of the MK2 gene ameliorates the severity of acute pancreatitis and accompanying systemic inflammatory reactions in a new model of severe acute pancreatitis. Our data support the hypothesis that MK2 participates in the multifactorial regulation of early inflammatory responses in AP, independently of the regulation of stress proteins like HSP25 and HSP60 and most likely due to its effect on cytokine regulation.
Collapse
Affiliation(s)
- Yong-Yu Li
- Department of Pathophysiology, School of Medicine, Tongji University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Namkung W, Yoon JS, Kim KH, Lee MG. PAR2 exerts local protection against acute pancreatitis via modulation of MAP kinase and MAP kinase phosphatase signaling. Am J Physiol Gastrointest Liver Physiol 2008; 295:G886-94. [PMID: 18755806 DOI: 10.1152/ajpgi.00053.2008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
During acute pancreatitis, protease-activated receptor 2 (PAR2) can be activated by interstitially released trypsin. In the mild form of pancreatitis, PAR2 activation exerts local protection against intrapancreatic damage, whereas, in the severe form of pancreatitis, PAR2 activation mediates some systemic complications. This study aimed to identify the molecular mechanisms of PAR2-mediated protective effects against intrapancreatic damage. A mild form of acute pancreatitis was induced by an intraperitoneal injection of caerulein (40 microg/kg) in rats. Effects of PAR2 activation on intrapancreatic damage and on mitogen-activated protein (MAP) kinase signaling were assessed. Caerulein treatment activated extracellular signal-regulated kinase (ERK) and c-Jun NH(2)-terminal kinase (JNK) within 15 min and maintained phosphorylation of ERK and JNK for 2 h in the rat pancreas. Although PAR2 activation by the pretreatment with PAR2-activating peptide (AP) itself increased ERK phosphorylation in rat pancreas, the same treatment remarkably decreased caerulein-induced activation of ERK and JNK principally by accelerating their dephosphorylation. Inhibition of ERK and JNK phosphorylation by the pretreatment with MAP/ERK kinase (MEK) or JNK inhibitors decreased caerulein-induced pancreatic damage that was similar to the effect induced by PAR2-AP. Notably, in caerulein-treated rats, PAR2-AP cotreatment highly increased the expression of a group of MAP kinase phosphatases (MKPs) that deactivate ERK and JNK. The above results imply that downregulation of MAP kinase signaling by MKP induction is a key mechanism involved in the protective effects of PAR2 activation on caerulein-induced intrapancreatic damage.
Collapse
Affiliation(s)
- Wan Namkung
- Dept. of Pharmacology, Yonsei Univ. College of Medicine, Seoul 120-752, Korea
| | | | | | | |
Collapse
|
14
|
Meyerholz DK, Williard DE, Grittmann AM, Samuel I. Murine pancreatic duct ligation induces stress kinase activation, acute pancreatitis, and acute lung injury. Am J Surg 2008; 196:675-82. [PMID: 18789417 DOI: 10.1016/j.amjsurg.2008.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 07/21/2008] [Accepted: 07/21/2008] [Indexed: 01/25/2023]
Abstract
BACKGROUND Acute lung injury is a major determinant of outcomes in acute pancreatitis. We evaluated acute lung injury and stress kinase activation in ligation-induced acute pancreatitis in mice. METHODS Mice with duct ligation or sham operation were killed after 24 or 48 hours. RESULTS In addition to acute pancreatitis, duct ligation was associated with pulmonary morphologic changes indicative of acute lung injury (alveolar septal thickening, congestion, and neutrophil infiltration). Furthermore, immunoblotting showed stress kinase activation in the pancreas and lung after ligation. Although mortality was observed in the ligated group, that is consistent with severe lung injury, it requires further evaluation. CONCLUSIONS Bile and pancreatic duct ligation in the mouse is associated with pancreatic and pulmonary stress kinase activation and acute inflammatory changes consistent with early acute pancreatitis and acute lung injury. Our findings are important as acute lung injury increases mortality in clinical acute pancreatitis and stress kinases are established proinflammatory signal transducers.
Collapse
Affiliation(s)
- David K Meyerholz
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
| | | | | | | |
Collapse
|
15
|
Abstract
OBJECTIVES Protease inhibitors showed protective effects on animal models of acute pancreatitis when administered before induction of pancreatitis, and results when administered after induction are uncertain. We assessed the effects of nafamostat mesilate in a mouse model of cerulein-induced pancreatitis comparing results of before and after induction. METHODS Cerulein was injected to mice intraperitoneally to induce pancreatitis, and they received intravenous nafamostat mesilate before and after induction. Serum concentrations of amylase and lipase, histological changes, and tissue expression of myeloperoxidase were measured. In addition, tissue activation of p38 mitogen-activated protein kinase (MAPK) and interleukin-6 was evaluated. RESULTS Development of pancreatitis was prevented by pretreatment with nafamostat mesilate. However, such effect was not shown when given after induction, although it partially suppressed myeloperoxidase expression and infiltration of inflammatory cells. Tissue expression of phospho-p38 MAPK was prominent in mice with pancreatitis and suppressed by pretreatment with nafamostat mesilate. Interleukin-6 expression was not influenced by either cerulein or nafamostat mesilate. CONCLUSIONS The development of pancreatitis was prevented by treating mice with nafamostat mesilate before induction, however, this finding was not observed if administered after injection of cerulein. Pretreatment with nafamostat mesilate suppressed activation of p38 MAPK.
Collapse
|
16
|
Mercer BA, D'Armiento JM. Emerging role of MAP kinase pathways as therapeutic targets in COPD. Int J Chron Obstruct Pulmon Dis 2008; 1:137-50. [PMID: 18046891 PMCID: PMC2706609 DOI: 10.2147/copd.2006.1.2.137] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Studies examining the cellular mechanisms of inflammation and protease production in the lung tissue and airways of COPD patients have shed light on the important role of kinase-based signaling cascades. These pathways can be activated by environmental stimuli such as tobacco smoke, and by endogenous signals such as cytokines, growth factors, and inflammation-derived oxidants. The three most widely characterized cascades are those directed by the classical mitogen activated protein (MAP) kinase (ERK1/2), stress activated protein kinase/c-Jun N-terminal protein kinase, and p38 enzymes. These phosphorylation cascades transmit and amplify extracellular, receptor-mediated signals through the cytoplasm of the cell to activate nuclear transcription factors which bind and induce expression of target genes. The result is tight control of diverse cellular events, and rapid responses to external stimuli. However, recent research suggests that constitutive or aberrant activation of MAP kinases contributes to several COPD-associated phenotypes, including mucus overproduction and secretion, inflammation, cytokine expression, apoptosis, T cell activation, matrix metalloproteinase production, and fibrosis. This review explores the biological functions of the MAP kinase pathways in the pathogenesis of COPD, their activation by cigarette smoke, and discusses the potential role of MAP kinase inhibitors in COPD therapy.
Collapse
Affiliation(s)
- Becky A Mercer
- Institute of Human Nutrition, Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York 10032, USA.
| | | |
Collapse
|
17
|
Malleo G, Mazzon E, Siriwardena AK, Cuzzocrea S. Role of tumor necrosis factor-alpha in acute pancreatitis: from biological basis to clinical evidence. Shock 2007; 28:130-40. [PMID: 17529903 DOI: 10.1097/shk.0b013e3180487ba1] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor necrosis factor (TNF)-alpha is a pleiotropic cytokine that exerts host-damaging effects in different autoimmune and inflammatory diseases. It is a key regulator of other proinflammatory cytokines and of leukocyte adhesion molecules, and it is a priming activator of immune cells. In recent years, several research lines-mostly derived from animal models and in vitro studies-suggested that TNF-alpha plays a pivotal role in the pathogenesis of acute pancreatitis. In particular, it contributes to the systemic progression of the inflammatory response and to the end-organ dysfunction often observed in severe disease. Current clinical applications of TNF-alpha in acute pancreatitis include the assessment of blood concentrations to predict disease severity and to identify individuals prone to develop complications such as multiple organ failure and septic shock. However, TNF-alpha is rapidly cleared from the bloodstream, and sensitivity and overall accuracy of its measurement seem strictly time dependent, thereby being of potential prognostic value only in the first days after the onset of the disease. In parallel, TNF-alpha has been evaluated as a novel pharmacologic target for treating pancreatitis. Although promising results have been observed in the laboratory, transition to clinical practice seems problematic, in particular, in the light of divergent results obtained in sepsis trials. Therefore, in future clinical trials pertaining to TNF-alpha neutralization in acute pancreatitis, timing of intervention should be related to changes in TNF-alpha serum levels, and inclusion and exclusion criteria should be accurately selected to better define the population most likely to benefit.
Collapse
Affiliation(s)
- Giuseppe Malleo
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Via C. Valeria-Gazzi, 98100 Messina, Italy
| | | | | | | |
Collapse
|
18
|
|
19
|
Jaeschke A, Davis RJ. Metabolic stress signaling mediated by mixed-lineage kinases. Mol Cell 2007; 27:498-508. [PMID: 17679097 PMCID: PMC1986670 DOI: 10.1016/j.molcel.2007.07.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 05/31/2007] [Accepted: 07/06/2007] [Indexed: 01/22/2023]
Abstract
Saturated free fatty acid (FFA) is a major source of metabolic stress that activates the c-Jun NH(2)-terminal kinase (JNK). This FFA-stimulated JNK pathway is relevant to hallmarks of metabolic syndrome, including insulin resistance. Here we used gene ablation studies in mice to demonstrate a central role for mixed-lineage protein kinases (MLK) in this signaling pathway. Saturated FFA causes protein kinase C (PKC)-dependent activation of MLK3 that subsequently causes increased JNK activity by a mechanism that requires the MAP kinase kinases MKK4 and MKK7. Loss of PKC, MLK3, MKK4, or MKK7 expression prevents FFA-stimulated JNK activation. Together, these data establish a signaling pathway that mediates effects of metabolic stress on insulin resistance.
Collapse
Affiliation(s)
- Anja Jaeschke
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School and Worcester, Massachusetts 01605, USA
| | - Roger J. Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School and Worcester, Massachusetts 01605, USA
| |
Collapse
|
20
|
Algül H, Treiber M, Lesina M, Nakhai H, Saur D, Geisler F, Pfeifer A, Paxian S, Schmid RM. Pancreas-specific RelA/p65 truncation increases susceptibility of acini to inflammation-associated cell death following cerulein pancreatitis. J Clin Invest 2007; 117:1490-501. [PMID: 17525802 PMCID: PMC1868784 DOI: 10.1172/jci29882] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 03/30/2007] [Indexed: 12/21/2022] Open
Abstract
Activation of the transcription factor NF-kappaB/Rel has been shown to be involved in inflammatory disease. Here we studied the role of RelA/p65, the main transactivating subunit, during acute pancreatitis using a Cre-loxP strategy. Selective truncation of the rela gene in pancreatic exocrine cells led to both severe injury of the acinar cells and systemic complications including lung and liver damage. Our data demonstrated that expression and induction of the protective pancreas-specific acute phase protein pancreatitis-associated protein 1 (PAP1) depended on RelA/p65. Lentiviral gene transfer of PAP1 cDNA reduced the extent of necrosis and infiltration in the pancreata of mice with selective truncation of RelA/p65. These results provide in vivo evidence for RelA/p65 protection of acinar cell death via upregulation of PAP1. Moreover, our data underscore the pancreas-specific role of NF-kappaB/Rel and suggest multidimensional roles of NF-kappaB/Rel in different cells and contexts during inflammation.
Collapse
Affiliation(s)
- Hana Algül
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Matthias Treiber
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Marina Lesina
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Hassan Nakhai
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Dieter Saur
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Fabian Geisler
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Alexander Pfeifer
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Stephan Paxian
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| | - Roland M. Schmid
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
Department of Neurology, Molecular Neurology Unit, University of Muenster, Muenster, Germany
| |
Collapse
|
21
|
Abstract
c-Jun N-terminal protein kinases (JNK), also known as stress-activated protein kinases, were originally identified by their ability to phosphorylate the N-terminal of the transcription factor c-Jun and by their activation in response to a variety of stresses. JNK are multifunctional kinases involved in many physiological processes. The JNK pathway has been shown to play a major role in apoptosis in many cell death paradigms and its association with a variety of pathological processes is gradually been recognized. This review will concentrate on describing the involvement of the JNK pathway in the context of different diseases and the potential to adopt the JNK pathway components as therapeutic targets.
Collapse
Affiliation(s)
- Jie Cui
- Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|
22
|
Affiliation(s)
- Stephen J Pandol
- Department of Medicine, Department of Veterans Affairs and University of California, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
23
|
Samuel I, Zaheer A, Fisher RA. In vitro evidence for role of ERK, p38, and JNK in exocrine pancreatic cytokine production. J Gastrointest Surg 2006; 10:1376-83. [PMID: 17175457 DOI: 10.1016/j.gassur.2006.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 09/13/2006] [Indexed: 02/07/2023]
Abstract
Elucidation of mechanisms of acinar cell cytokine production is essential for a better understanding of acute pancreatitis pathogenesis. We hypothesize that the stress kinases ERK, p38, and JNK play an important role in acinar cell cytokine production. Rat pancreatic fragments were incubated with 100 nM concentration of the cholecystokinin analog caerulein or 100 nM caerulein and specific ERK inhibitor (100 microM PD98059), specific p38 inhibitor (10 microM SB203580), or specific JNK inhibitor (20 microM SP600125). After 3 hours of caerulein treatment, pancreatic fragments were homogenized and assayed for total and phosphorylated ERK, p38, and JNK, and for tumor necrosis factor-alpha or interleukin-1beta concentrations (ELISA). Pancreatic fragments stimulated with caerulein showed activation of ERK, p38, and JNK and increased cytokine concentrations (ANOVA, P<0.05). Specific stress kinase inhibitors significantly attenuated caerulein-induced activation of the corresponding stress kinase and cytokine production; however, the effect of the JNK inhibitor was comparatively less convincing. Increased activation of ERK, p38, and JNK in pancreatic fragments was not associated with significant increases in total ERK, total p38, or total JNK concentrations. The stress kinases ERK and p38 play an important role in caerulein-stimulated exocrine pancreatic overproduction of cytokines. The role of JNK needs further evaluation in this experimental model.
Collapse
Affiliation(s)
- Isaac Samuel
- Department of Surgery, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, and Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
24
|
Abstract
Cholecystokinin and gastrin receptors (CCK1R and CCK2R) are G protein-coupled receptors that have been the subject of intensive research in the last 10 years with corresponding advances in the understanding of their functioning and physiology. In this review, we first describe general properties of the receptors, such as the different signaling pathways used to exert short- and long-term effects and the structural data that explain their binding properties, activation, and regulation. We then focus on peripheral cholecystokinin receptors by describing their tissue distribution and physiological actions. Finally, pathophysiological peripheral actions of cholecystokinin receptors and their relevance in clinical disorders are reviewed.
Collapse
Affiliation(s)
- Marlène Dufresne
- Institut National de la Santé et de la Recherche Médicale U. 531, Institut Louis Bugnard, Centre Hospitalier Universitaire Rangueil, France
| | | | | |
Collapse
|
25
|
Tietz AB, Malo A, Diebold J, Kotlyarov A, Herbst A, Kolligs FT, Brandt-Nedelev B, Halangk W, Gaestel M, Göke B, Schäfer C. Gene deletion of MK2 inhibits TNF-alpha and IL-6 and protects against cerulein-induced pancreatitis. Am J Physiol Gastrointest Liver Physiol 2006; 290:G1298-306. [PMID: 16423921 DOI: 10.1152/ajpgi.00530.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory effects contribute to the pathogenesis of pancreatitis. Clearly, proinflammatory cytokines like TNF-alpha and IL-6 are involved in this process and the associated systemic complications. The MAPKAPK-2 (MK2) signaling pathway is involved in cytokine gene expression. Therefore, we hypothesized that blockade of this pathway inhibits the expression of proinflammatory cytokines and thereby protects against pancreatitis. To investigate this, we used an in vivo mouse model with a homozygous deletion of the MK2 gene. Pancreatitis was induced by injection of cerulein. The severity was determined by measuring serum lipase, pancreatic trypsin activation, pancreatic edema, and morphological changes by quantitative scoring of histological sections. Systemic inflammation was evaluated by measuring myeloperoxidase activity in lung tissue. Serum levels of TNF-alpha and IL-6 were measured using an ELISA, and mRNA levels were identified using RT-PCR and subsequent quantitative PCR analysis. Pancreatitis in animals with deletion of the MK2 gene is less severe and accompanied with reduced serum levels of TNF-alpha and IL-6. Pancreatic mRNA levels revealed a fourfold reduction of IL-6 mRNA expression in MK2 -/- mice. Effects were associated with suppression of pancreatic trypsin activity and reduced acinar cell injury. In summary, these data show that gene deletion of MK2 ameliorates cerulein-induced pancreatitis. TNF-alpha and IL-6 signaling is mediated by the MK2 pathway and therefore crucial for the regulatory inflammatory processes. TNF-alpha expression is supposably regulated by a posttranscriptional mechanism, whereas IL-6 expression is most likely regulated by transcriptional effects.
Collapse
Affiliation(s)
- Anne Barbara Tietz
- Department of Internal Medicine II, Klinikum Grosshadern, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shi C, Wang X, Zhao X, Andersson R. Cellular and molecular events in acute pancreatitis. JOURNAL OF ORGAN DYSFUNCTION 2006; 2:142-150. [DOI: 10.1080/17471060600776858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Gallmeier E, Schäfer C, Moubarak P, Tietz A, Plössl I, Huss R, Göke B, Wagner ACC. JAK and STAT proteins are expressed and activated by IFN-gamma in rat pancreatic acinar cells. J Cell Physiol 2005; 203:209-16. [PMID: 15493010 DOI: 10.1002/jcp.20216] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of acute pancreatitis (AP) is triggered by acinar events, but the subsequent extra-acinar events, particularly a distinct immune response, appear to determine its severity. Cytokines modulate this immune response and are derived not only from immunocytes but also from pancreatic acinar cells. We studied whether pancreatic acinar cells were also capable of responding to cytokines. The JAK/STAT-pathway represents the main effector for many cytokines. Therefore, expression and regulation of JAK and STAT proteins were investigated in rat pancreatic acinar cells. Western blotting showed expression of JAK1, JAK2, Tyk2, and STAT1, STAT2, STAT3, STAT5, STAT6. In addition, STAT1 was reversibly tyrosine-phosphorylated upon the procedure of acinar cell isolation. In contrast, STAT3-phosphorylation occurred spontaneously after pancreas removal and was not reversible within 8 h. STAT1 phosphorylation was also observed upon treatment with IFN-gamma but not upon EGF, TNF-alpha or IL-6, and inhibited by the JAK2-inhibitor AG-490. Immunohistochemistry revealed cytoplasmic expression of unphosphorylated STAT1 in untreated acinar cells and nuclear translocation of phosphorylated STAT1 following IFN-gamma-treatment. Interestingly, although CCK leads to the activation of multiple stress pathways in pancreatic acinar cells, we found no influence of CCK on phosphorylation of STAT1, STAT3, or STAT5 in the pancreas. In conclusion, our data provide further evidence that pancreatic acinar cells are able to interact with immune cells. Besides stimulating immune cells via cytokine secretion, acinar cells are in turn capable of responding to IFN-gamma via JAK2 and STAT1 which may have an impact on the development of AP.
Collapse
Affiliation(s)
- E Gallmeier
- Department of Internal Medicine II, Klinikum Grosshadern, Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Minutoli L, Altavilla D, Marini H, Passaniti M, Bitto A, Seminara P, Venuti FS, Famulari C, Macrì A, Versaci A, Squadrito F. Protective effects of SP600125 a new inhibitor of c-jun N-terminal kinase (JNK) and extracellular-regulated kinase (ERK1/2) in an experimental model of cerulein-induced pancreatitis. Life Sci 2004; 75:2853-2866. [PMID: 15454338 DOI: 10.1016/j.lfs.2004.03.040] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2003] [Accepted: 03/18/2004] [Indexed: 12/29/2022]
Abstract
Extracellular regulated kinases (ERK1/2) and c-Jun N-terminal Kinases (JNK), are generally considered to play a key role in signal transduction pathways activated by a wide range of stimuli. We studied the effects of SP600125, a novel inhibitor of both JNK and ERK1/2, in male C57/BL6 mice given with an hyper-stimulating dose of cerulein (50 microg/kg for each of four injections at hourly intervals) to elicit secretagogue-induced pancreatitis. A control group received four intra-peritoneal injections of 0.9% saline at hourly intervals. Animals were randomized to receive either SP600125 (15 mg/kg i.p. administered 2 h before and 30 min after the first injection of cerulein) or its vehicle (1 ml/kg of a 10% DMSO/NaCl solution). A group of animals was killed 30 minutes after the last cerulein injection to evaluate pancreatic JNK and ERK1/2 activation by Western Blot analysis. Another group was sacrificed 2 hours after the last cerulein injection to evaluate serum lipase and amylase levels, pancreas oedema, pancreatic content of Tumor Necrosis Factor-alpha (TNF-alpha) and Intercellular adhesion molecule-1 (ICAM-1) and the histological alterations. SP600125 inhibited almost totally JNK activation (90%) and partially ERK1/2 activation (45%), reduced the serum lipase and amylase levels and the degree of oedema, blunted the increased pancreatic content of TNF-alpha and ICAM-1 and protected against the histological damage. Our data confirm that both JNK and ERK1/2 activation plays a key role in acute pancreatitis and that SP600125 may represent a potential therapeutic approach to the treatment of patients at high risk of developing this life-threatening condition.
Collapse
Affiliation(s)
- Letteria Minutoli
- Department of Clinical and Experimental Medicine and Pharmacology, Section of Pharmacology, University of Messina AOU G. Martino Torre Biologica 5th floor, Via Consolare Valeria, Gazzi 98100, Messina, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ploessl I, Gallmeier E, Schaefer C, Bilzer M, Bittmann I, Göke B, Wagner ACC. ANP preconditioning does not increase protection against experimental pancreatitis, observed after general anesthesia and jugular vein catheterization. Pancreas 2004; 28:166-73. [PMID: 15028949 DOI: 10.1097/00006676-200403000-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
It has been widely shown that preconditioning, inducing heat shock proteins, can protect against experimentally induced pancreatitis. Solid evidence indicates that HSP70 plays a central role in this context, possibly by inhibition of premature intracellular trypsinogen activation. Current preconditioning protocols such as whole body hyperthermia are, however, quite strenuous and clinically not applicable. There is little data on other means to induce pancreatic HSPs such as pharmacologic pretreatment.However, in models of ischemic liver reperfusion injury, it has been demonstrated that atrial natriuretic peptide (ANP) can be used for such pharmacologic preconditioning. Evidence indicates that ANP exerts its protective effects via increased cGMP levels, activation of heat shock transcription factor (HSF) and, increased protein levels of HSP70. Pancreatic acinar cells express ANP receptors and respond to ANP treatment with increased cGMP levels. We have, therefore, investigated whether intravenous ANP pretreatment could be used to protect the pancreas against experimental pancreatitis. When given 20 minutes prior to pancreatitis induction, ANP pretreatment had no effect on cerulein-induced pancreatitis. In contrast, 24 hours after preconditioning, induction of HSP70 protein expression and protection against experimental pancreatitis were found. However, controls treated with NaCl without ANP showed a similar response. This indicates that stress caused by general anesthesia and jugular vein catheterization can be sufficient for preconditioning while ANP, in contrast to models of ischemic liver reperfusion injury, does not confer additional protection.
Collapse
Affiliation(s)
- I Ploessl
- Department of Medicine II, Grosshadern Hospital, University of Munich, Institute of Pathology, University of Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Affiliation(s)
- M Löwenberg
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
31
|
Samuel I, Zaheer S, Fisher RA, Zaheer A. Cholinergic receptor induction and JNK activation in acute pancreatitis. Am J Surg 2003; 186:569-74. [PMID: 14599627 DOI: 10.1016/j.amjsurg.2003.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cholecystokinin-A (CCK-A) and cholinergic receptor pathways, capable of activating stress kinases p38 mitogen-activated protein kinase (p38(MAPK)) and cJUN N-terminal kinase (JNK), are implicated in the pathogenesis of ligation-induced acute pancreatitis in rats. As ligation-induced acute pancreatitis in rats is associated with CCK-A receptor induction and p38(MAPK) activation, and as receptor induction could amplify acinar hyperstimulation and exacerbate cell stress, we tested the hypothesis that the cholinergic M3 receptor is induced and JNK is activated in this model. METHODS Cholinergic M3 receptor expression and JNK activation was compared in rats 1, 3, or 24 hours after sham operation or duct ligation. RESULTS Immunoblot analysis of pancreatic homogenates showed a time-dependent increase in cholinergic M3 receptor protein, total JNK, and phospho-JNK after duct ligation. CONCLUSIONS There is a rapid and progressive cholinergic M3 receptor induction and JNK activation in ligation-induced acute pancreatitis in rats. These findings may have significance in the mechanism of disease pathogenesis.
Collapse
Affiliation(s)
- Isaac Samuel
- Department of Surgery, Veterans Affairs Medical Center, University of Iowa Roy J. and Lucille A. Carver College of Medicine, 200 Hawkins Drive, 4625 JCP, Iowa City, IA 52242, USA.
| | | | | | | |
Collapse
|
32
|
Ji B, Chen XQ, Misek DE, Kuick R, Hanash S, Ernst S, Najarian R, Logsdon CD. Pancreatic gene expression during the initiation of acute pancreatitis: identification of EGR-1 as a key regulator. Physiol Genomics 2003; 14:59-72. [PMID: 12709512 DOI: 10.1152/physiolgenomics.00174.2002] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We hypothesized that genes expressed in pancreatic acinar cells during the initiation of acute pancreatitis determine the severity of the disease. Therefore, we utilized microarrays to identify those genes commonly induced in rat pancreatic acinar cells within 1-4 h in two in vivo models, caerulein and taurocholate administration. This strategy yielded 51 known genes representing a complex array of molecules, including those that are likely to either reduce or increase the severity of the disease. Novel genes identified in the current study included ATF3, BRF1, C/EBPbeta, CGRP, EGR-1, ephrinA1, villin2, ferredoxin, latexin, lipocalin, MKP-1, NGFI-B, RhoA, tissue factor (TF), and syndecan. To validate these microarray results, the role of EGR-1 was further investigated using quantitative RT-PCR, Western blotting, and immunocytochemistry. EGR-1 expression occurred within acinar cells and correlated with the development of caerulein-induced acute pancreatitis in rats. Furthermore, the levels of the inflammation-related genes MCP-1, PAI, TF, IL-6, and ICAM-1 and the extent of lung inflammation were reduced during the initiation of caerulein-induced acute pancreatitis in EGR-1-deficient mice. Thus this study identified EGR-1 and several other novel genes likely to be important in the development and severity of acute pancreatitis.
Collapse
Affiliation(s)
- Baoan Ji
- Department of Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ding SP, Li JC, Jin C. A mouse model of severe acute pancreatitis induced with caerulein and lipopolysaccharide. World J Gastroenterol 2003; 9:584-9. [PMID: 12632523 PMCID: PMC4621587 DOI: 10.3748/wjg.v9.i3.584] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a non-traumatic, easy to induce and reproducible mouse model of severe acute pancreatitis (SAP) induced with caerulein and lipopolyasccharide (LPS).
METHODS: Thirty-two healthy mature NIH female mice were selected and divided at random into four groups (each of 8 mice), i.e., the control group (NS group), the caerulein group (Cn group), the lipopolysaccharide group (LPS group), and the caerulein+LPS group (Cn + LPS group). Mice were injected intraperitoneally with caerulein only, or LPS only, and caerulein and LPS in combination. All the animals were then killed by neck dislocation three hours after the last intraperitoneal injection. The pancreas and exo-pancreatic organs were then carefully removed for microscopic examination. And the pancreatic acinus was further observed under transmission electron microscope (TEM). Pancreatic weight, serum amylase, serum nitric oxide (NO) concentration, superoxide dismutase (SOD) and malondialdehyde (MDA) concentration of the pancreas were assayed respectively.
RESULTS: (1) NS animals displayed normal pancreatic structure both in the exocrine and endocrine. In the LPS group, the pancreas was slightly edematous, with the infiltration of a few inflammatory cells and the necrosis of the adjacent fat tissues. All the animals of the Cn group showed distinct signs of a mild edematous pancreatitis characterized by interstitial edema, infiltration of neutrophil and mononuclear cells, but without obvious parenchyma necrosis and hemorrhage. In contrast, the Cn + LPS group showed more diffuse focal areas of nonviable pancreatic and hemorrhage as well as systemic organ dysfunction. According to Schmidt’s criteria, the pancreatic histologic score showed that there existed significant difference in the Cn + LPS group in the interstitial edema, inflammatory infiltration, parenchyma necrosis and parenchyma homorrhage in comparison with those of the Cn group, LPS group and NS group (P < 0.01 or P < 0.05). (2) The ultrasturcture of acinar cells was seriously damaged in the Cn + LPS group. Chromatin margination of nuclei was present, the number and volume of vacuoles greatly increased. Zymogen granules (ZGs) were greatly decreased in number and endoplasmic reticulum exhibited whorls. The swollen mitochondria appeared, the crista of which was decreased in number or disappeared. (3) Pancreatic weight and serum amylase levels in the Cn +LPS was significantly higher than those of the NS group and the LPS group respectively (P < 0.01 or P < 0.05). However, the pancreatic wet weight and serum amylase concentration showed no significant difference between the Cn + LPS group and the Cn group. (4) NO concentration in the Cn + LPS group was significantly higher than that of NS group, LPS group and Cn group(P < 0.05 or P < 0.01). 5) The SOD and MDA concentration of the pancreas in the Cn + LPS group were significantly higher than those of NS, LPS and Cn groups (P < 0.05 or P < 0.01).
CONCLUSION: The mouse model of severe acute pancreatitis could be induced with caerulein and LPS, which could be non-traumatic and easy to induce, reproducible with the same pathological characteristics as those of SAP in human, and could be used in the research on the mechanism of human SAP.
Collapse
Affiliation(s)
- Shi-Ping Ding
- Department of Lymphology, Department of Histology and Embryology, Medical College of Zhejiang University, Hangzhou 310031, Zhejiang Province, China.
| | | | | |
Collapse
|
34
|
Clemons AP, Holstein DM, Galli A, Saunders C. Cerulein-induced acute pancreatitis in the rat is significantly ameliorated by treatment with MEK1/2 inhibitors U0126 and PD98059. Pancreas 2002; 25:251-9. [PMID: 12370536 DOI: 10.1097/00006676-200210000-00007] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Both cerulein and cholecystokinin activate mitogen-activated protein (MAP) kinase (ERK1/2) in vivo and in isolated pancreatic acini. AIMS AND METHODOLOGY ERK1/2 in pancreas homogenates was activated in rats rendered pancreatitic by subcutaneous injections of cerulein (5 microg/kg per hour). To determine if blocking ERK1/2 activity might rescue cerulein-induced acute pancreatitis, the "MAP kinase kinase" (also known as MEK1/2) inhibitors PD98059 and U0126 were administered in vivo. RESULTS In rats pretreated with PD98059 (10 mg/kg per i.v. injection) or U0126 (5 mg/kg per i.v. injection) 30 minutes before and then together with hourly cerulein injections for 3 hours, pancreatitis was significantly attenuated on the basis of pancreatic wet weight and histology. Serum amylase concentration was significantly reduced when PD98059 was administered intraperitoneally (10 mg/kg per intraperitoneal injection). PD98059 also ameliorated pancreatitis over a 6-hour cerulein time course. The phosphorylation of pancreatic ERK1/2 was attenuated in PD98059- and U0126-treated animals at both 30 minutes and 3 hours after cerulein injection. Rats rendered neutropenic with vinblastine and pretreated with U0126 still showed attenuated manifestations of cerulein-induced acute pancreatitis, a finding suggesting that pancreatic ERK1/2 is mostly responsible for the effect, rather than infiltrating neutrophils. CONCLUSIONS Inhibition of pancreatic ERK1/2 in vivo affords significant protection against inflammatory sequelae following cerulein-induced acute pancreatitis.
Collapse
Affiliation(s)
- Antoinette P Clemons
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | | | |
Collapse
|
35
|
Williams JA. Intracellular signaling mechanisms activated by cholecystokinin-regulating synthesis and secretion of digestive enzymes in pancreatic acinar cells. Annu Rev Physiol 2001; 63:77-97. [PMID: 11181949 DOI: 10.1146/annurev.physiol.63.1.77] [Citation(s) in RCA: 200] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intracellular signaling mechanisms by which cholecystokinin (CCK) and other secretagogues regulate pancreatic acinar function are more complex than originally realized. CCK couples through heterotrimeric G proteins of the Gq family to lead to an increase in intracellular free Ca2+, which shows spatial and temporal patterns of signaling. The actions of Ca2+ are mediated in part by activation of a number of Ca2+-activated protein kinases and the protein phosphatase calcineurin. By the process of exocytosis the intracellular messengers Ca2+, diacylglycerol, and cAMP activate the release of the zymogen granule content in a manner that is poorly understood. This fusion event most likely involves SNARE and Rab proteins present on zymogen granules and cellular membrane domains. More likely related to nonsecretory aspects of cell function, CCK also activates three MAPK cascades leading to activation of ERKs, JNKs, and p38 MAPK. Although the function of these pathways is not well understood, ERKs are probably related to cell growth, and through phosphorylation of hsp27, p38 can affect the actin cytoskeleton. The PI3K (phosphatidylinositol 3-kinase)-mTOR (mammalian target of rapamycin) pathway is important for regulation of acinar cell protein synthesis because it leads to both activation of p70S6K and regulation of the availability of eIF4E in response to CCK. CCK also activates a number of tyrosyl phosphorylation events including that of p125FAK and other proteins associated with focal adhesions.
Collapse
Affiliation(s)
- J A Williams
- Department of Physiology, University of Michigan, Ann Arbor, Michigan 48109-0622, USA.
| |
Collapse
|
36
|
Abstract
AIM: To examine the role of p38 during acute experimental cerulein pancreatitis.
METHODS: Rats were treated with cerulein with or without a specific JNK inhibitor (CEP1347) and/or a specific p38 inhbitor (SB203580) and pancreatic stress kinase activity was determined. Parameters to assess pancreatitis included trypsin, amylase, lipase, pancreatic weight and histology.
RESULTS: JNK inhibition with CEP1347 ameliorated pancreatitis, reducing pancreatic edema. In contrast, p38 inhibition with SB203580 aggravated pancreatitis with higher trypsin levels and, with induction of acinar necrosis not normally found after cerulein hyperstimulation. Simultaneous treatment with both CEP1347 and SB203580 mutually abolished the effects of either compound on cerulein pancreatitis.
CONCLUSION: Stress kinases modulate pancreatitis differentially. JNK seems to promote pancreatitis development, possibly by supporting inflammatory reactions such as edema formation while its inhibition ameliorates pancreatitis. In contrast, p38 may help reduce organ destruction while inhibition of p38 during induction of cerulein pancreatitis leads to the occurrence of acinar necrosis.
Collapse
Affiliation(s)
- F Fleischer
- Department of Gastroenterology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | | | | | | |
Collapse
|