51
|
Liu Y, Yang L, Chen KL, Zhou B, Yan H, Zhou ZG, Li Y. Knockdown of GRP78 promotes apoptosis in pancreatic acinar cells and attenuates the severity of cerulein and LPS induced pancreatic inflammation. PLoS One 2014; 9:e92389. [PMID: 24643222 PMCID: PMC3958537 DOI: 10.1371/journal.pone.0092389] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/22/2014] [Indexed: 02/05/2023] Open
Abstract
Acute pancreatitis (AP) is a potentially lethal disease characterized by inflammation and parenchymal cell death; also, the severity of AP correlates directly with necrosis and inversely with apoptosis. However, mechanisms of regulating cell death in AP remain unclear. The endoplasmic reticulum (ER) chaperone protein GRP78 has anti-apoptotic properties, in addition to modulating ER stress responses. This study used RNA interference (RNAi) approach to investigate the potential role of GRP78 in regulating apoptosis during AP. In vitro models of AP were successfully developed by treating AR42J cells with cerulein or cerulein plus lipoplysaccharide (LPS). There was more pancreatic inflammation and less apoptosis with the cerulein plus LPS treatment. Furthermore, knockdown of GRP78 expression markedly promoted apoptosis and reduced necrosis in pancreatic acinar cells. This was accomplished by enhancing the activation of caspases and inhibiting the activity of X-linked inhibitor of apoptosis protein (XIAP), as well as a receptor interacting protein kinase-1(RIPK1), which is a key mediator of necrosis. This attenuated the severity of pancreatic inflammation, especially after cerulein plus LPS treatment. In conclusion, these findings indicate that GRP78 plays an anti-apoptotic role in regulating the cell death response during AP. Therefore, GRP78 is a potential therapeutic target for AP.
Collapse
Affiliation(s)
- Yong Liu
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lie Yang
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke-Ling Chen
- Institute of Digestive Surgery and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Zhou
- Institute of Digestive Surgery and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Yan
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zong-Guang Zhou
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Digestive Surgery and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Institute of Digestive Surgery and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
52
|
Abstract
OBJECTIVE The objective of this study was to investigate the potential protective effects of fucoidan, an L- and P-selectin modulator, in 2 murine models of acute pancreatitis. METHODS Acute pancreatitis was induced in mice either by the retrograde infusion of taurolithocholic acid sulfate into the pancreatic duct or by intraperitoneal injections of cerulein (50 μg/kg per hour). The experimental groups received fucoidan (25 mg/kg, intravenously) before pancreatitis induction, whereas control groups received only saline. After 24 hours, serum amylase, lipase, interleukin 1β (IL-1β), tumor necrosis factor α (TNF-α), and nitrite were measured. In addition, myeloperoxidase (MPO) activity (lung and pancreas) and histological assessment (pancreas) were determined. RESULTS Serum amylase, lipase, nitrite, TNF-α, and IL-1β, and pancreatic and lung MPO were increased in both taurolithocholic acid sulfate and cerulein acute pancreatitis compared with the respective control groups. Fucoidan significantly decreased the augmented levels of amylase, lipase, pancreatic and lung MPO, TNF-α, IL-1β, and nitrite in both models. Pancreas histological changes observed in both acute pancreatitis models were significantly attenuated by fucoidan. CONCLUSIONS Fucoidan reduced the severity of acute pancreatitis in mice by decreasing neutrophil infiltration and systemic inflammation, suggesting that modulation of selectins may constitute a promising therapeutic approach.
Collapse
|
53
|
Inhibition of hydrogen sulfide synthesis provides protection for severe acute pancreatitis rats via apoptosis pathway. Apoptosis 2013; 18:28-42. [PMID: 23054084 DOI: 10.1007/s10495-012-0770-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We aimed to investigate the relationship between the synthesis of hydrogen sulfide (H(2)S) and the pancreatic acinar cell apoptosis in severe acute pancreatitis (SAP) rats, as well as analyse the potential apoptotic pathway involved in this process. Sixty rats had been equally divided into four groups: sham, SAP, SAP + sodium hydrosulfide (NaHS) and SAP + DL-propargylglycine (PAG). 24 h after SAP induction, all surviving animals of each group were sacrificed to collect blood and tissue samples for the following measurements: the level of serum H(2)S as well as the levels of tumor necrosis factor-alpha (TNF-α), interleukin-10 (IL-10), H(2)S synthesizing activity, CSE mRNA and protein expression, maleic dialdehyde (MDA) and myeloperoxidase (MPO) activity, the expression of Bax, Bcl-2, caspase-3, -8 and -9, the release of cytochrome c and the activation of nuclear factor-kappa B (NF-κB), ERK1/2, JNK1/2 and p38 in pancreas. Furthermore, in situ detection of cell apoptosis was examined and the severity of pancreatic damage was analyzed by pathological grading and scoring. Results Significant differences in every index except IL-10 had been found between the SAP, NaHS and PAG groups (P < 0.05). Treatment with PAG obviously induced the pancreatic acinar cell apoptosis as well as improved all the pathological changes and inflammatory parameters. In contrast, administration of NaHS significantly attenuated apoptosis in the pancreas and aggravated the severity of pancreatic damage. Moreover, the expressions of caspase-3, -8, -9 and the release of cytochrome c were all increased in the apoptotic cells, and the activity of NF-κB as well as the phosphorylation of ERK1/2, JNK1/2 and p38 decreased accompanying with the reduction of the serum H(2)S level. H(2)S plays a pivotal role in the regulation of pancreatic acinar cell apoptosis in SAP rats. The present results showed that inhibition of H(2)S synthesis provided protection for SAP rats via inducing acinar cell apoptosis. This process acted through both extrinsic and intrinsic apoptotic pathways, and may be regulated by reducing the activity of NF-κB.
Collapse
|
54
|
Hage C, Rausch V, Giese N, Giese T, Schönsiegel F, Labsch S, Nwaeburu C, Mattern J, Gladkich J, Herr I. The novel c-Met inhibitor cabozantinib overcomes gemcitabine resistance and stem cell signaling in pancreatic cancer. Cell Death Dis 2013; 4:e627. [PMID: 23661005 PMCID: PMC3674365 DOI: 10.1038/cddis.2013.158] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignancies. Cancer stem cells (CSCs), which are not targeted by current therapies, may be the reason for pronounced therapy resistance. A new treatment option in phase II trials is cabozantinib that inhibits the pancreatic CSC surface marker and tyrosine kinase receptor c-Met. The purpose of this study was to evaluate the effect of cabozantinib to stem-like features and therapy resistance. Established PDA cell lines, a gemcitabine-resistant subclone, non-malignant pancreatic ductal cells and primary spheroidal cultures from patient tumors were analyzed by MTT-assay, flow cytometry, colony and spheroid formation assays, western blotting, qRT-PCR, antibody protein array, immunohistochemistry and morphological features. Cabozantinib inhibited viability and spheroid formation and induced apoptosis in malignant cells with minor effects in non-malignant cells. After long-term cabozantinib treatment, PDA cells had altered anti- and pro-apoptotic signaling, but still responded to cabozantinib, as apoptosis only slightly decreased and viability only slightly increased suggesting a low resistance-inducing potential of cabozantinib. In parallel, c-Met expression and the pluripotency transcription factor SOX2 were downregulated, which might counteract development of full therapy resistance in long-term treated subclones. In single-treatment studies, cabozantinib increased efficacy of gemcitabine. Most importantly, cabozantinib strongly induced apoptosis and reduced viability in PDA cell lines, which are completely resistant toward gemcitabine. In primary, CSC-enriched spheroidal cultures cabozantinib downregulated CSC markers SOX2, c-Met and CD133 and induced apoptosis. These findings suggest that the clinical use of cabozantinib may be more effective than current chemotherapeutics.
Collapse
Affiliation(s)
- C Hage
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - V Rausch
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - N Giese
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - T Giese
- Department of Molecular Immunodiagnostics, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - F Schönsiegel
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - S Labsch
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - C Nwaeburu
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - J Mattern
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - J Gladkich
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - I Herr
- Department of Experimental Medicine, Molecular OncoSurgery Group, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General Surgery, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
55
|
1,4-dihydroxy-2-naphthoic Acid Induces Apoptosis in Human Keratinocyte: Potential Application for Psoriasis Treatment. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:792840. [PMID: 23690852 PMCID: PMC3638593 DOI: 10.1155/2013/792840] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 01/31/2023]
Abstract
Psoriasis, which affects approximately 1–3% of the population worldwide, is a chronic inflammatory skin disorder characterized by epidermal keratinocytes hyperproliferation, abnormal differentiation, and inflammatory infiltration. Decrease in keratinocyte apoptosis is a specific pathogenic phenomenon in psoriasis. Chinese herbs have been used for the treatment of psoriasis in China showing promising effect in clinical trials. A traditional Chinese medicine has relatively fewer side effects with longer remission time and lower recurrence rate. The extract of Rubia cordifolia L. (EA) was previously found by us to induce HaCaT keratinocytes apoptosis. In this study we identified one of the components in Rubia cordifolia L., the anthraquinone precursor 1,4-dihydroxy-2-naphthoic acid (DHNA), induces HaCaT keratinocytes apoptosis through G0/G1 cell cycle arrest. We have also demonstrated that DHNA acts through both caspase-dependent and caspase-independent pathways. Besides, cytotoxicity and IL-1α release assays indicate that DHNA causes less irritation problems than dithranol, which is commonly employed to treat psoriasis in many countries. Since DHNA possesses similar apoptotic effects on keratinocytes as dithranol but causes less irritation, DHNA therefore constitutes a promising alternative agent for treating psoriasis. Our studies also provide an insight on the potential of using EA and DHNA, alternatively, as a safe and effective treatment modality for psoriasis.
Collapse
|
56
|
Maléth J, Rakonczay Z, Venglovecz V, Dolman NJ, Hegyi P. Central role of mitochondrial injury in the pathogenesis of acute pancreatitis. Acta Physiol (Oxf) 2013; 207:226-35. [PMID: 23167280 DOI: 10.1111/apha.12037] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 10/26/2012] [Accepted: 10/30/2012] [Indexed: 12/11/2022]
Abstract
Acute pancreatitis is an inflammatory disease with no specific treatment. One of the main reasons behind the lack of specific therapy is that the pathogenesis of acute pancreatitis is poorly understood. During the development of acute pancreatitis, the disease-inducing factors can damage both cell types of the exocrine pancreas, namely the acinar and ductal cells. Because damage of either of the cell types can contribute to the inflammation, it is crucial to find common intracellular mechanisms that can be targeted by pharmacological therapies. Despite the many differences, recent studies revealed that the most common factors that induce pancreatitis cause mitochondrial damage with the consequent breakdown of bioenergetics, that is, ATP depletion in both cell types. In this review, we summarize our knowledge of mitochondrial function and damage within both pancreatic acinar and ductal cells. We also suggest that colloidal ATP delivery systems for pancreatic energy supply may be able to protect acinar and ductal cells from cellular damage in the early phase of the disease. An effective energy delivery system combined with the prevention of further mitochondrial damage may, for the first time, open up the possibility of pharmacological therapy for acute pancreatitis, leading to reduced disease severity and mortality.
Collapse
Affiliation(s)
- J. Maléth
- First Department of Medicine; University of Szeged; Szeged; Hungary
| | - Z. Rakonczay
- First Department of Medicine; University of Szeged; Szeged; Hungary
| | - V. Venglovecz
- Department of Pharmacology and Pharmacotherapy; University of Szeged; Szeged; Hungary
| | - N. J. Dolman
- Molecular Probes Labelling and Detection Technologies; Life Technologies Corporation; Eugene; OR; USA
| | - P. Hegyi
- First Department of Medicine; University of Szeged; Szeged; Hungary
| |
Collapse
|
57
|
Wang LW, Huang Z, Lin H, Li ZS, Hetzel F, Liu Md B. Effect of Photofrin-mediated photocytotoxicity on a panel of human pancreatic cancer cells. Photodiagnosis Photodyn Ther 2013; 10:244-251. [PMID: 23993850 DOI: 10.1016/j.pdpdt.2012.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 10/24/2012] [Accepted: 11/01/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Pancreatic cancer is a leading cause of cancer-related deaths in men and women. Early clinical studies suggest that photodynamic therapy (PDT) might be a useful modality in the management of this deadly disease. In this study, the photocytotoxicity of Photofrin-mediated PDT on different human pancreatic cancer cells (BxPc-3, HPAF-II, Mia PaCa-2, MPanc-96, PANC-1 and PL-45) was examined. MATERIALS AND METHODS After co-incubating cancer cells with Photofrin (0-10 μg/ml) for 4h, the cells were irradiated with 0-6J/cm(2) of 630 nm light. The effect of Photofrin PDT on the survival of cells were examined using tetrazolium-based colorimetric assay and clonogenic assay. PDT-induced apoptosis was analyzed by flow cytometry. Expressions of apoptosis-related proteins were determined by western blot analysis. RESULTS Photofrin PDT strongly inhibited the survival of pancreatic cancer cells. A small portion of cells (<15%) underwent apoptosis 24h after PDT at LD50. Cleavage of caspase-3, caspase-8, caspase-9 and PARP after PDT were also confirmed. BxPc-3, Mia PaCa-2, MPanc-96, and PANC-1 cells were more sensitive and HPAF-II and PL-45 cells less sensitive. CONCLUSION Photofrin PDT can induce apoptosis and inhibit survival of human pancreatic cancer cells.
Collapse
Affiliation(s)
- Luo-Wei Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
- Division of Gastroenterology & Hepatology, University of Colorado Denver, Aurora, CO, USA
- Department of Pathology, University of Colorado Denver, Aurora, CO, USA
| | - Zheng Huang
- Department of Radiation Oncology, University of Colorado Denver, Aurora, CO, USA
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Fred Hetzel
- Department of Radiation Oncology, University of Colorado Denver, Aurora, CO, USA
| | - Bolin Liu Md
- Department of Pathology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
58
|
Li S, Sun J, Yang J, Zhang L, Wang LE, Wang X, Guo Z. XIAP expression is associated with pancreatic carcinoma outcome. Mol Clin Oncol 2013; 1:305-308. [PMID: 24649165 DOI: 10.3892/mco.2013.58] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 12/19/2012] [Indexed: 11/05/2022] Open
Abstract
Pancreatic carcinoma is an aggressive tumor that is resistant to treatment regimens. Although the X-linked inhibitor of apoptosis protein (XIAP) overexpression in pancreatic carcinoma cells has been previously reported, the aim of this study was to investigate the association of XIAP expression with pancreatic carcinoma outcome. Specimens were obtained from 54 patients who underwent resection for pancreatic cancer. Kaplan-Meier survival analysis was used to estimate post-operative survival. The results of this analysis revealed a significantly shorter overall survival time in patients with a high compared to those with a low XIAP expression (P=0.041). Results of the multivariate analysis revealed that XIAP expression was identified as an independent predictor for pancreatic carcinoma outcome [relative risk, 1.771; 95% confidence interval (CI), 1.099-2.852; P=0.019]. Moreover, XIAP levels were associated with pancreatic carcinoma characteristics, including tumor invasion status and histological grade. XIAP overexpression shortens the survival of pancreatic cancer patients probably by modifying their resistance to apoptosis and the proliferation capacity of pancreatic carcinoma cells. Thus, XIAP expression analysis can help in the identification of patient subgroups at high risk of a poor disease outcome.
Collapse
Affiliation(s)
- Shengmian Li
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| | - Jianjian Sun
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| | - Jian Yang
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| | - Lan Zhang
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| | - LE Wang
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| | - Xiaoling Wang
- Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhanjun Guo
- Departments of Gastroenterology and Hepatology, Hebei 050011, P.R. China
| |
Collapse
|
59
|
Huang H, Liu Y, Daniluk J, Gaiser S, Chu J, Wang H, Logsdon C, Ji B. Activation of nuclear factor-κB in acinar cells increases the severity of pancreatitis in mice. Gastroenterology 2013; 144:202-10. [PMID: 23041324 PMCID: PMC3769090 DOI: 10.1053/j.gastro.2012.09.059] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Nuclear factor-κB (NF-κB) is activated during early stages of pancreatitis. This transcription factor regulates genes that control many cell activities, including inflammation and survival. There is evidence that activation of NF-κB protects against pancreatitis, and, in other cases, that it promotes this disease. We compared the effects of NF-κB in different mouse models of pancreatitis to understand these complications. METHODS To model constitutive activation of NF-κB, we expressed a transgene that encodes its p65 subunit or the inhibitor of κB kinase (IKK)2 in pancreatic acinar cells of mice. We analyzed effects on pancreatic tissues and levels of NF-κB target genes in these mice and compared them with mice that did not express transgenic p65 or IKK2 (controls). RESULTS Transgenic expression of p65 led to compensatory expression of the inhibitory subunit IKB-α and, therefore, no clear phenotype. However, p65 transgenic mice given injections of cerulein, to induce acute pancreatitis, had higher levels of NF-κB activity in acinar cells, greater levels of inflammation, and more severe outcomes than control mice. In contrast, constitutive expression of IKK2 directly increased the activity of NF-κB in acinar cells and induced pancreatitis. Prolonged activity of IKK2 (3 months) resulted in activation of stellate cells, loss of acinar cells, and fibrosis, which are characteristics of chronic pancreatitis. Co-expression of IKK2 and p65 greatly increased the expression of inflammatory mediators and the severity of pancreatitis, compared with control mice. CONCLUSIONS The level of NF-κB activation correlates with the severity of acute pancreatitis in mice. Longer periods of activation (3 months) lead to chronic pancreatitis. These findings indicate that strategies to inactivate NF-κB might be used to treat patients with acute or chronic pancreatitis.
Collapse
Affiliation(s)
- Haojie Huang
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yan Liu
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jaroslaw Daniluk
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
- Department of Gastroenterology, Medical University of Bialystok, Poland
| | - Sebastian Gaiser
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jun Chu
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Huamin Wang
- Department of pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Craig Logsdon
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
- Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Baoan Ji
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| |
Collapse
|
60
|
Salnikov AV, Liu L, Platen M, Gladkich J, Salnikova O, Ryschich E, Mattern J, Moldenhauer G, Werner J, Schemmer P, Büchler MW, Herr I. Hypoxia induces EMT in low and highly aggressive pancreatic tumor cells but only cells with cancer stem cell characteristics acquire pronounced migratory potential. PLoS One 2012; 7:e46391. [PMID: 23050024 PMCID: PMC3458836 DOI: 10.1371/journal.pone.0046391] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 08/29/2012] [Indexed: 12/27/2022] Open
Abstract
Tumor hypoxia induces epithelial-mesenchymal transition (EMT), which induces invasion and metastasis, and is linked to cancer stem cells (CSCs). Whether EMT generates CSCs de novo, enhances migration of existing CSCs or both is unclear. We examined patient tissue of pancreatic ductal adenocarcinoma (PDA) along with carcinomas of breast, lung, kidney, prostate and ovary. For in vitro studies, five established PDA cell lines classified as less (CSC(low)) and highly aggressive CSC-like cells (CSC(high)) were examined by single and double immunofluorescence microscopy, wound-, transwell-, and time-lapse microscopy. HIF-1α and Slug, as well as HIF-2α and CD133 were co-expressed pointing to a putative co-existence of hypoxia, EMT and CSCs in vivo. CSC(high) cells exhibited high basal expression of the mesenchymal Vimentin protein but low or absent expression of the epithelial marker E-cadherin, with the opposite result in CSC(low) cells. Hypoxia triggered altering of cell morphology from an epithelial to a mesenchymal phenotype, which was more pronounced in CSC(high) cells. Concomitantly, E-cadherin expression was reduced and expression of Vimentin, Slug, Twist2 and Zeb1 enhanced. While hypoxia caused migration in all cell lines, velocity along with the percentage of migrating, polarized and pseudopodia-forming cells was significantly higher in CSC(high) cells. These data indicate that hypoxia-induced EMT occurs in PDA and several other tumor entities. However although hypoxia-induced EMT signaling occurs in all tumor cell populations, only the stem-like cells acquire high migratory potential and thus may be responsible for invasion and metastasis.
Collapse
Affiliation(s)
- Alexei V. Salnikov
- Molecular OncoSurgery Group, Department of General Surgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of Translational Immunology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery Group, Department of General Surgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Mitja Platen
- Molecular OncoSurgery Group, Department of General Surgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Jury Gladkich
- Molecular OncoSurgery Group, Department of General Surgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Olga Salnikova
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Eduard Ryschich
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jürgen Mattern
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Gerhard Moldenhauer
- Department of Translational Immunology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Jens Werner
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Schemmer
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Markus W. Büchler
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery Group, Department of General Surgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
61
|
Paterniti I, Mazzon E, Riccardi L, Galuppo M, Impellizzeri D, Esposito E, Bramanti P, Cappellani A, Cuzzocrea S. Peroxisome proliferator-activated receptor β/δ agonist GW0742 ameliorates cerulein- and taurocholate-induced acute pancreatitis in mice. Surgery 2012; 152:90-106. [DOI: 10.1016/j.surg.2012.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 02/09/2012] [Indexed: 01/01/2023]
|
62
|
Rausch V, Liu L, Apel A, Rettig T, Gladkich J, Labsch S, Kallifatidis G, Kaczorowski A, Groth A, Gross W, Gebhard MM, Schemmer P, Werner J, Salnikov AV, Zentgraf H, Büchler MW, Herr I. Autophagy mediates survival of pancreatic tumour-initiating cells in a hypoxic microenvironment. J Pathol 2012; 227:325-35. [PMID: 22262369 DOI: 10.1002/path.3994] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 01/07/2012] [Accepted: 01/12/2012] [Indexed: 02/07/2023]
Abstract
Involvement of dysregulated autophagy in cancer growth and progression has been shown in different tumour entities, including pancreatic ductal adenocarcinoma (PDA). PDA is an extremely aggressive tumour characterized by a small population of highly therapy-resistant cancer stem cells (CSCs) capable of self-renewal and migration. We examined whether autophagy might be involved in the survival of CSCs despite nutrition and oxygen deprivation typical for the hypoxic tumour microenvironment of PDA. Immunohistochemistry revealed that markers for hypoxia, CSCs and autophagy are co-expressed in patient-derived tissue of PDA. Hypoxia starvation (H/S) enhanced clonogenic survival and migration of established pancreatic cancer cells with stem-like properties (CSC(high)), while pancreatic tumour cells with fewer stem cell markers (CSC(low)) did not survive these conditions. Electron microscopy revealed more advanced autophagic vesicles in CSC(high) cells, which exhibited higher expression of autophagy-related genes under normoxic conditions and relative to CSC(low) cells, as found by RT-PCR and western blot analysis. LC3 was already fully converted to the active LC3-II form in both cell lines, as evaluated by western blot and detection of accumulated GFP-LC3 protein by fluorescence microscopy. H/S increased formation of autophagic and acid vesicles, as well as expression of autophagy-related genes, to a higher extent in CSC(high) cells. Modulation of autophagy by inhibitors and activators resensitized CSC(high) to apoptosis and diminished clonogenicity, spheroid formation, expression of CSC-related genes, migratory activity and tumourigenicity in mice. Our data suggest that enhanced autophagy levels may enable survival of CSC(high) cells under H/S. Interference with autophagy-activating or -inhibiting drugs disturbs the fine-tuned physiological balance of enhanced autophagy in CSC and switches survival signalling to suicide.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Autophagy/drug effects
- Autophagy/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/ultrastructure
- Cell Hypoxia
- Cell Line, Tumor
- Cell Movement
- Cell Survival
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Mice
- Mice, Nude
- Microscopy, Electron
- Microscopy, Fluorescence
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/ultrastructure
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/ultrastructure
- Polymerase Chain Reaction
- Time Factors
- Tumor Burden
- Tumor Microenvironment
Collapse
Affiliation(s)
- Vanessa Rausch
- Molecular OncoSurgery, University of Heidelberg and German Cancer Research Centre, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Long-lasting effect of infant rats endotoxemia on heat shock protein 60 in the pancreatic acinar cells: involvement of toll-like receptor 4. Int J Inflam 2012; 2012:354904. [PMID: 22685683 PMCID: PMC3364569 DOI: 10.1155/2012/354904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 03/10/2012] [Accepted: 03/14/2012] [Indexed: 01/22/2023] Open
Abstract
Introduction. Lipopolysaccharide endotoxin (LPS) is responsible for septic shock and multiorgan failure, but pretreatment of rats with low doses of LPS reduced pancreatic acute damage. Aim. We investigated the effects of the endotoxemia induced in the early period of life on Toll-like receptor 4 (TLR4), heat shock protein 60 (HSP60) and proapoptotic Bax, caspase-9 and -3 or antiapoptotic Bcl-2 protein expression in the pancreatic acinar cells of adult animals. Material and Methods. Newborn rats (25 g) were injected with endotoxin (Escherichia coli) for 5 consecutive days. Two months later, pancreatic acinar cells were isolated from all groups of animals and subjected to caerulein stimulation (10−8 M). Protein expression was assessed employing Western blot. For detection of apoptosis we have employed DNA fragmentation ladder assay. Results. Preconditioning of newborn rats with LPS increased TLR4, Caspase-9 and -3 levels, but failed to affect basal expression of HSP60, Bax, and Bcl-2. Subsequent caerulein stimulation increased TLR4, Bcl-2, and caspases, but diminished HSP60 and Bax proteins in pancreatic acinar cells. Endotoxemia dose-dependently increased TLR4, Bax, HSP60, and both caspases protein signals in the pancreatic acini, further inhibiting antiapoptotic Bcl-2. Conclusions. Endotoxemia promoted the induction of HSP60 via TLR4 in the infant rats and participated in the LPS-dependent pancreatic tissue protection against acute damage.
Collapse
|
64
|
Effects of mitogen-activated protein kinase signaling pathway inhibition on the development of cerulein-induced acute pancreatitis in mice. Pancreas 2012; 41:560-70. [PMID: 22228051 DOI: 10.1097/mpa.0b013e31823acd56] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Extracellular signal-regulated kinase (ERK) influences a number of pathways in all cells. The ERK cascade has long been known to be central to the activation of cellular processes such as proliferation, differentiation, and oncogenic transformation. The mitogen-activated protein (MAP) serine/threonine family of protein kinases, of which ERK is a member, is evolutionarily conserved and is activated by a mechanism that includes protein kinase cascades. The aim of this study was to investigate the effects of PD98059 [2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one], a highly selective inhibitor of MAP/ERK kinase 1 (MEK1) activation, on the development of acute pancreatitis. METHODS Pancreatitis was induced by intraperitoneal injection of cerulein (hourly × 5, 50 μg/kg) and PD98059 (10 mg/kg, 10% dimethylsulfoxide, intraperitoneally) was administrated 1 and 3 hours after cerulein administration. RESULTS Cerulein injection resulted in acute necrotizing pancreatitis. On the contrary, pancreatitis histological features, amylase, lipase, pancreas edema, and immunohistochemical staining for leukocyte adhesion molecules, transforming growth factor β, and apoptosis-related proteins were found reduced in PD98059-treated mice. CONCLUSIONS We propose that this study could help to clarify the role of MAPK in the regulation of the inflammatory process as acute pancreatitis.
Collapse
|
65
|
Yuan J, Liu Y, Tan T, Guha S, Gukovsky I, Gukovskaya A, Pandol SJ. Protein kinase d regulates cell death pathways in experimental pancreatitis. Front Physiol 2012; 3:60. [PMID: 22470346 PMCID: PMC3313474 DOI: 10.3389/fphys.2012.00060] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/29/2012] [Indexed: 11/13/2022] Open
Abstract
Inflammation and acinar cell necrosis are two major pathological responses of acute pancreatitis, a serious disorder with no current therapies directed to its molecular pathogenesis. Serine/threonine protein kinase D family, which includes PKD/PKD1, PKD2, and PKD3, has been increasingly implicated in the regulation of multiple physiological and pathophysiological effects. We recently reported that PKD/PKD1, the predominant PKD isoform expressed in rat pancreatic acinar cells, mediates early events of pancreatitis including NF-κB activation and inappropriate intracellular digestive enzyme activation. In current studies, we investigated the role and mechanisms of PKD/PKD1 in the regulation of necrosis in pancreatic acinar cells by using two novel small molecule PKD inhibitors CID755673 and CRT0066101 and molecular approaches in in vitro and in vivo experimental models of acute pancreatitis. Our results demonstrated that both CID755673 and CRT0066101 are PKD-specific inhibitors and that PKD/PKD1 inhibition by either the chemical inhibitors or specific PKD/PKD1 siRNAs attenuated necrosis while promoting apoptosis induced by pathological doses of cholecystokinin-octapeptide (CCK) in pancreatic acinar cells. Conversely, up-regulation of PKD expression in pancreatic acinar cells increased necrosis and decreased apoptosis. We further showed that PKD/PKD1 regulated several key cell death signals including inhibitors of apoptotic proteins, caspases, receptor-interacting protein kinase 1 to promote necrosis. PKD/PKD1 inhibition by CID755673 significantly ameliorated necrosis and severity of pancreatitis in an in vivo experimental model of acute pancreatitis. Thus, our studies indicate that PKD/PKD1 is a key mediator of necrosis in acute pancreatitis and that PKD/PKD1 may represent a potential therapeutic target in acute pancreatitis.
Collapse
Affiliation(s)
- Jingzhen Yuan
- South California Research Center for Alcoholic Liver and Pancreatic Diseases, Veterans Affairs Greater Los Angeles Healthcare System, University of California at Los Angeles Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
66
|
Gukovsky I, Pandol SJ, Mareninova OA, Shalbueva N, Jia W, Gukovskaya AS. Impaired autophagy and organellar dysfunction in pancreatitis. J Gastroenterol Hepatol 2012; 27 Suppl 2:27-32. [PMID: 22320913 PMCID: PMC3281514 DOI: 10.1111/j.1440-1746.2011.07004.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent findings from our group, obtained on experimental in vivo and ex vivo models of pancreatitis, reveal that this disease causes a profound dysfunction of key cellular organelles, lysosomes and mitochondria. We found that autophagy, the main cellular degradative, lysosome-driven process, is activated but also impaired in acute pancreatitis because of its' inefficient progression/resolution (flux) resulting from defective function of lysosomes. One mechanism underlying the lysosomal dysfunction in pancreatitis is abnormal processing (maturation) and activation of cathepsins, major lysosomal hydrolases; another is a decrease in pancreatic levels of key lysosomal membrane proteins LAMP-1 and LAMP-2. Our data indicate that lysosomal dysfunction plays an important initiating role in pancreatitis pathobiology. The impaired autophagy mediates vacuole accumulation in acinar cells; furthermore, the abnormal maturation and activation of cathepsins leads to increase in intra-acinar trypsin, the hallmark of pancreatitis; and LAMP-2 deficiency causes inflammation and acinar cell necrosis. Thus, the autophagic and lysosomal dysfunctions mediate key pathologic responses of pancreatitis. On the other hand, we showed that pancreatitis causes acinar cell mitochondria depolarization, mediated by the permeability transition pore (PTP). Genetic (via deletion of cyclophilin D) inactivation of PTP prevents mitochondrial depolarization and greatly ameliorates the pathologic responses of pancreatitis. Further, our data suggest that mitochondrial damage, by stimulating autophagy, increases the demand for efficient lysosomal degradation and therefore aggravates the pathologic consequences of lysosomal dysfunction. Thus, the combined autophagic, lysosomal and mitochondrial dysfunctions are key to the pathogenesis of pancreatitis.
Collapse
Affiliation(s)
- Ilya Gukovsky
- Southern California Research Centers for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, VA Greater Los Angeles Healthcare System, and University of California at Los Angeles, California 90073, USA
| | | | | | | | | | | |
Collapse
|
67
|
Gukovsky I, Pandol SJ, Gukovskaya AS. Organellar dysfunction in the pathogenesis of pancreatitis. Antioxid Redox Signal 2011; 15:2699-710. [PMID: 21834686 PMCID: PMC3183656 DOI: 10.1089/ars.2011.4068] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Acute pancreatitis is an inflammatory disease of exocrine pancreas that carries considerable morbidity and mortality; its pathophysiology remains poorly understood. During the past decade, new insights have been gained into signaling pathways and molecules that mediate the inflammatory response of pancreatitis and death of acinar cells (the main exocrine pancreas cell type). By contrast, much less is known about the acinar cell organellar damage in pancreatitis and how it contributes to the disease pathogenesis. RECENT ADVANCES This review summarizes recent findings from our group, obtained on experimental in vivo and ex vivo models, which reveal disordering of key cellular organelles, namely, mitochondria, autophagosomes, and lysosomes, in pancreatitis. Our results indicate a critical role for mitochondrial permeabilization in determining the balance between apoptosis and necrosis in pancreatitis, and thus the disease severity. We further investigate how the mitochondrial dysfunction (and hence acinar cell death) is regulated by Ca(2+), reactive oxygen species, and Bcl-xL, in relation to specific properties of pancreatic mitochondria. Our results also reveal that autophagy, the principal cellular degradative, lysosome-driven pathway, is impaired in pancreatitis due to inefficient lysosomal function, and that impaired autophagy mediates two key pathological responses of pancreatitis-accumulation of vacuoles in acinar cells and the abnormal, intra-acinar activation of digestive enzymes such as trypsinogen. CRITICAL ISSUES AND FUTURE DIRECTIONS The findings discussed in this review indicate critical roles for mitochondrial and autophagic/lysosomal dysfunctions in the pathogenesis of pancreatitis and delineate directions for detailed investigations into the molecular events that underlie acinar cell organellar damage.
Collapse
Affiliation(s)
- Ilya Gukovsky
- VA Greater Los Angeles Healthcare System, University of California, Los Angeles, California 90073, USA.
| | | | | |
Collapse
|
68
|
Morak MJ, van Koetsveld PM, Kanaar R, Hofland LJ, van Eijck CH. Type I interferons as radiosensitisers for pancreatic cancer. Eur J Cancer 2011; 47:1938-45. [DOI: 10.1016/j.ejca.2011.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 03/08/2011] [Indexed: 01/02/2023]
|
69
|
Galuppo M, Nocentini G, Mazzon E, Ronchetti S, Esposito E, Riccardi L, Sportoletti P, Di Paola R, Bruscoli S, Riccardi C, Cuzzocrea S. The glucocorticoid-induced TNF receptor family-related protein (GITR) is critical to the development of acute pancreatitis in mice. Br J Pharmacol 2011; 162:1186-201. [PMID: 21091650 DOI: 10.1111/j.1476-5381.2010.01123.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Pancreatitis represents a life-threatening inflammatory condition where leucocytes, cytokines and vascular endothelium contribute to the development of the inflammatory disease. The glucocorticoid-induced tumour necrosis factor (TNF) receptor family-related protein (GITR) is a costimulatory molecule for T lymphocytes, modulates innate and adaptive immune system and has been found to participate in a variety of immune responses and inflammatory processes. Our purpose was to verify whether inhibition of GITR triggering results in a better outcome in experimental pancreatitis. EXPERIMENTAL APPROACH In male GITR knock-out (GITR(-/-)) and GITR(+/+) mice on Sv129 background, acute pancreatitis was induced after i.p. administration of cerulein. Other experimental groups of GITR(+/+) mice were also treated with different doses of Fc-GITR fusion protein (up to 6.25 µg·mouse⁻¹), given by implanted mini-osmotic pump. Clinical score and pro-inflammatory parameters were evaluated. KEY RESULTS A less acute pancreatitis was found in GITR(-/-) mice than in GITR(+/+) mice, with marked differences in oedema, neutrophil infiltration, pancreatic dysfunction and injury. Co-treatment of GITR(+/+) mice with cerulein and Fc-GITR fusion protein (6.25 µg·mouse⁻¹) decreased the inflammatory response and tissue injury, compared with treatment with cerulein alone. Inhibition of GITR triggering was found to modulate activation of nuclear factor κB as well as the production of TNF-α, interleukin-1β, inducible nitric oxide synthase, nitrotyrosine, poly-ADP-ribose, intercellular adhesion molecule-1 and P-selectin. CONCLUSIONS AND IMPLICATIONS The GITR-GITR ligand system is crucial to the development of acute pancreatitis in mice. Our results also suggest that the Fc-GITR fusion protein could be useful in the treatment of acute pancreatitis.
Collapse
Affiliation(s)
- M Galuppo
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Pandol SJ, Lugea A, Mareninova OA, Smoot D, Gorelick FS, Gukovskaya AS, Gukovsky I. Investigating the pathobiology of alcoholic pancreatitis. Alcohol Clin Exp Res 2011; 35:830-7. [PMID: 21284675 DOI: 10.1111/j.1530-0277.2010.01408.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alcohol abuse is one of the most common causes of pancreatitis. The risk of developing alcohol-induced pancreatitis is related to the amount and duration of drinking. However, only a small portion of heavy drinkers develop disease, indicating that other factors (genetic, environmental, or dietary) contribute to disease initiation. Epidemiologic studies suggest roles for cigarette smoking and dietary factors in the development of alcoholic pancreatitis. The mechanisms underlying alcoholic pancreatitis are starting to be understood. Studies from animal models reveal that alcohol sensitizes the pancreas to key pathobiologic processes that are involved in pancreatitis. Current studies are focussed on the mechanisms responsible for the sensitizing effect of alcohol; recent findings reveal disordering of key cellular organelles including endoplasmic reticulum, mitochondria, and lysosomes. As our understanding of alcohol's effects continue to advance to the level of molecular mechanisms, insights into potential therapeutic strategies will emerge providing opportunities for clinical benefit.
Collapse
Affiliation(s)
- Stephen J Pandol
- Pancreatic Research Group, Department of Veterans Affairs Greater Los Angeles, University of California Los Angeles, 90073, USA.
| | | | | | | | | | | | | |
Collapse
|
71
|
Stadel D, Mohr A, Ref C, MacFarlane M, Zhou S, Humphreys R, Bachem M, Cohen G, Möller P, Zwacka RM, Debatin KM, Fulda S. TRAIL-induced apoptosis is preferentially mediated via TRAIL receptor 1 in pancreatic carcinoma cells and profoundly enhanced by XIAP inhibitors. Clin Cancer Res 2010; 16:5734-49. [PMID: 20940278 DOI: 10.1158/1078-0432.ccr-10-0985] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We previously reported that small molecule X-linked inhibitor of apoptosis (XIAP) inhibitors synergize with soluble TRAIL to trigger apoptosis in pancreatic carcinoma cells. Because cancers may preferentially signal via 1 of the 2 agonistic TRAIL receptors, we investigated these receptors as a therapeutic target in pancreatic cancer in the present study. EXPERIMENTAL DESIGN We examined TRAIL receptor expression and cytotoxicity of specific monoclonal antibodies to TRAIL-R1 (HGS-ETR1, mapatumumab) or TRAIL-R2 (HGS-ETR2, lexatumumab) and of TRAIL receptor selective mutants alone and in combination with small molecule XIAP inhibitors in pancreatic cancer cell lines, in primary specimens, and in a xenotransplant model in vivo. RESULTS The majority of primary pancreatic carcinoma samples and all cell lines express one or both agonistic TRAIL receptors. Nine of 13 cell lines are more sensitive to mapatumumab-induced apoptosis, whereas lexatumumab requires cross-linking for maximal activity. Similarly, TRAIL-R1 selective mutants display higher cytotoxicity than TRAIL-R2 selective mutants. Small molecule XIAP inhibitors preferentially act in concert with mapatumumab to trigger caspase activation, caspase-dependent apoptosis, and suppress clonogenic survival. Also, primary cultured pancreatic carcinoma cells are more susceptible to mapatumumab than lexatumumab, which is significantly enhanced by a XIAP inhibitor. Importantly, combined treatment with mapatumumab and a XIAP inhibitor cooperates to suppress tumor growth in vivo. CONCLUSIONS Mapatumumab exerts antitumor activity, especially in combination with XIAP inhibitors against most pancreatic carcinoma cell lines, whereas lexatumumab requires cross-linking for optimal cytotoxicity. These findings have important implications for the design of TRAIL-based protocols for pancreatic cancer.
Collapse
Affiliation(s)
- Dominic Stadel
- University Children's Hospital, Ulm University, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Chvanov M, Petersen OH, Tepikin AV. Pharmacologically directed cell disposal: labeling damaged cells for phagocytosis as a strategy against acute pancreatitis. Mol Interv 2010; 10:80-5. [PMID: 20368368 DOI: 10.1124/mi.10.2.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Michael Chvanov
- The Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK.
| | | | | |
Collapse
|
73
|
Rausch V, Liu L, Kallifatidis G, Baumann B, Mattern J, Gladkich J, Wirth T, Schemmer P, Büchler MW, Zöller M, Salnikov AV, Herr I. Synergistic activity of sorafenib and sulforaphane abolishes pancreatic cancer stem cell characteristics. Cancer Res 2010; 70:5004-13. [PMID: 20530687 DOI: 10.1158/0008-5472.can-10-0066] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent evidence suggests that pancreatic cancer and other solid tumors contain a subset of tumorigenic cells capable of extensive self-renewal that contribute to metastasis and treatment resistance. Sorafenib (SO) is a promising new multikinase inhibitor for treatment of advanced kidney and liver cancers. We report here targeting of pancreatic cancer stem cells (CSC) by SO and the development of a strategy to enhance this effect. Although SO administration diminished clonogenicity, spheroid formation, aldehyde dehydrogenase 1 (ALDH1) activity, growth on immunodeficient mice, proliferation, and angiogenesis and induced apoptosis, we observed SO-induced activation of NF-kappaB associated with survival and regrowth of spheroids. For enhanced elimination of CSC characteristics by SO, we cotreated cells with sulforaphane (SF). This broccoli isothiocyanate was recently described to eliminate pancreatic CSCs by downregulation of NF-kappaB activity without inducing toxic side effects. On combination treatment, SF completely eradicated SO-induced NF-kappaB binding, which was associated with abrogated clonogenicity, spheroid formation, ALDH1 activity, migratory capacity, and induction of apoptosis. In vivo, combination therapy reduced the tumor size in a synergistic manner. This was due to induction of apoptosis, inhibition of proliferation and angiogenesis, and downregulation of SO-induced expression of proteins involved in epithelial-mesenchymal transition. Our data suggest that SF may be suited to increase targeting of CSCs by SO.
Collapse
Affiliation(s)
- Vanessa Rausch
- Molecular OncoSurgery, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Hyvönen MT, Sinervirta R, Keinänen TA, Fashe T, Grigorenko N, Khomutov AR, Vepsäläinen J, Alhonen L. Acute pancreatitis induced by activated polyamine catabolism is associated with coagulopathy: effects of alpha-methylated polyamine analogs on hemostasis. Pancreatology 2010; 10:208-21. [PMID: 20453551 DOI: 10.1159/000243730] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 09/21/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS Polyamines are ubiquitous organic cations essential for cellular proliferation and tissue integrity. We have previously shown that pancreatic polyamine depletion in rats overexpressing the catabolic enzyme, spermidine/spermine N(1)-acetyltransferase (SSAT), results in the development of severe acute pancreatitis, and that therapeutic administration of metabolically stable alpha-methylated polyamine analogs protects the animals from pancreatitis-associated mortality. Our aim was to elucidate the therapeutic mechanism(s) of alpha-methylspermidine (MeSpd). METHODS The effect of MeSpd on hemostasis and the extent of organ failure were studied in SSAT transgenic rats with either induced pancreatitis or lipopolysaccharide (LPS)-induced coagulopathy. The effect of polyamines on fibrinolysis and coagulation was also studied in vitro. RESULTS Pancreatitis caused a rapid development of intravascular coagulopathy, as assessed by prolonged coagulation times, decreased plasma fibrinogen level and antithrombin activity, enhanced fibrinolysis, reduced platelet count and presence of schistocytes. Therapeutic administration of MeSpd restored these parameters to almost control levels within 24 h. In vitro, polyamines dose-dependently inhibited fibrinolysis and intrinsic coagulation pathway. In LPS-induced coagulopathy, SSAT transgenic rats were more sensitive to the drug than their syngeneic littermates, and MeSpd-ameliorated LPS-induced coagulation disorders. CONCLUSION Pancreatitis-associated mortality in SSAT rats is due to coagulopathy that is alleviated by treatment with MeSpd.
Collapse
Affiliation(s)
- M T Hyvönen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Kuopio, Kuopio, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Dahlhoff M, Algül H, Siveke JT, Lesina M, Wanke R, Wartmann T, Halangk W, Schmid RM, Wolf E, Schneider MR. Betacellulin protects from pancreatitis by activating stress-activated protein kinase. Gastroenterology 2010; 138:1585-94, 1594.e1-3. [PMID: 20038432 DOI: 10.1053/j.gastro.2009.12.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 11/25/2009] [Accepted: 12/15/2009] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Acute pancreatitis (AP) is a serious, unpredictable clinical problem, the pathophysiology of which is poorly understood. Here, we evaluate whether betacellulin (BTC), a ligand of the epidermal growth factor receptor also able to activate the proapoptotic ERBB4 receptor, can protect against experimental AP. METHODS AP was induced in transgenic mice overexpressing BTC (BTC-tg), control mice, or control mice after administration of recombinant BTC. The severity of pancreatitis was assessed by measurements of serum amylase and lipase and histologic grading. The involvement of the stress-activated protein kinase (SAPK) was evaluated by treating BTC-tg mice with an SAPK inhibitor before induction of AP. RESULTS BTC-tg mice showed increased apoptosis and proliferation in the exocrine pancreas, indicating an increased cell turnover. There was a marked, epidermal growth factor receptor-independent decrease in pancreas weight. After induction of AP by cerulein injection, BTC-tg mice showed a significantly lower increase in serum amylase and lipase levels as well as less pronounced tissue necrosis, edema, and inflammation, as compared to nontransgenic littermates. This protective effect, also confirmed in the L-arginine AP model, was associated with increased phosphorylation of SAPK and abrogated after treatment of BTC-tg mice with a SAPK inhibitor. Finally, the protective effect of BTC against AP was confirmed by treating nontransgenic mice with recombinant BTC. CONCLUSIONS These findings indicate a potential application of the BTC/ERBB4 pathway for modulating the course of AP.
Collapse
Affiliation(s)
- Maik Dahlhoff
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center of the LMU Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Tapia JA, Salido GM, González A. Ethanol consumption as inductor of pancreatitis. World J Gastrointest Pharmacol Ther 2010; 1:3-8. [PMID: 21577288 PMCID: PMC3091140 DOI: 10.4292/wjgpt.v1.i1.3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 01/11/2010] [Accepted: 01/18/2010] [Indexed: 02/06/2023] Open
Abstract
Alcohol abuse is a major cause of pancreatitis, a condition that can manifest as both acute necroinflammation and chronic damage (acinar atrophy and fibrosis). Pancreatic acinar cells can metabolize ethanol via the oxidative pathway, which generates acetaldehyde and involves the enzymes alcohol dehydrogenase and possibly cytochrome P4502E1. Additionally, ethanol can be metabolized via a nonoxidative pathway involving fatty acid ethyl ester synthases. Metabolism of ethanol by acinar and other pancreatic cells and the consequent generation of toxic metabolites, are postulated to play an important role in the development of alcohol-related acute and chronic pancreatic injury. This current work will review some recent advances in the knowledge about ethanol actions on the exocrine pancreas and its relationship to inflammatory disease and cancer.
Collapse
Affiliation(s)
- José A Tapia
- José A Tapia, Ginés M Salido, Antonio González, PhD, Department of Physiology, Faculty of Veterinary Sciences, University of Extremadura, Avenida Universidad s/n, Cáceres E-10071, Spain
| | | | | |
Collapse
|
77
|
Huang J, Yao WY, Zhu Q, Tu SP, Yuan F, Wang HF, Zhang YP, Yuan YZ. XAF1 as a prognostic biomarker and therapeutic target in pancreatic cancer. Cancer Sci 2010; 101:559-67. [PMID: 19922503 PMCID: PMC11158990 DOI: 10.1111/j.1349-7006.2009.01396.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
XAF1 (X chromosome-linked inhibitor of apoptosis [XIAP]-associated factor 1) is a novel XIAP modulator that negatively regulates the anti-apoptotic effects of XIAP and sensitizes cells to other cell death triggers. It has been reported to be downregulated in a variety of human cancer cell lines. However, the role of XAF1 in pancreatic carcinogenesis remains unclear. In the present study, we investigated the prognostic values of XAF1 expression and its regulation in cancer cell growth and apoptosis both in vitro and in vivo. From the immunohistochemistry staining of tissue microarray, 40 of 89 (44.9%) pancreatic specimens showed low levels of XAF1 expression. Statistical analysis suggested the downregulation of XAF1 was significantly correlated with tumor staging (P = 0.047) and those patients with low XAF1 levels had shorter survival times (P = 0.0162). Multivariate analysis indicated that XAF1 expression was an independent prognostic indicator of the survival of patients with pancreatic cancer (P = 0.007). Furthermore, we found that restoration of XAF1 expression mediated by Ad5/F35 virus suppressed cell proliferation and induced cell cycle arrest and apoptosis, accompanied by the activation of caspases 3, 8, and 9 and poly(ADP-ribose) polymerase as well as increased level of cytochrome c and Bid cleavage. Notably, XAF1 restoration robustly decreased survivin expression rather than XIAP. In addition, in vivo s.c. xenografts from Ad5/F35-XAF1 treatment, which showed less cellular proliferation and enhanced apoptosis, were significantly smaller than those from control groups. Our findings document that XAF1 is a valuable prognostic marker in pancreatic cancer and could be a potential candidate for cancer gene therapy.
Collapse
Affiliation(s)
- Jia Huang
- Department of Gastroenterology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Nakamura Y, Do JH, Yuan J, Odinokova IV, Mareninova O, Gukovskaya AS, Pandol SJ. Inflammatory cells regulate p53 and caspases in acute pancreatitis. Am J Physiol Gastrointest Liver Physiol 2010; 298:G92-100. [PMID: 19850968 PMCID: PMC2806100 DOI: 10.1152/ajpgi.00324.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The inflammatory response during pancreatitis regulates necrotic and apoptotic rates of parenchymal cells. Neutrophil depletion by use of anti-polymorphonuclear serum (anti-PMN) increases apoptosis in experimental pancreatitis but the mechanism has not been determined. Our study was designed to investigate signaling mechanisms in pancreatic parenchymal cells regulating death responses with neutrophil depletion. Rats were neutrophil depleted with anti-PMN treatment. Then cerulein pancreatitis was induced, followed by measurements of apoptosis signaling pathways. There was greater activation of executioner caspases-3 in the pancreas with anti-PMN treatment compared with control. There were no differences between these groups of animals in mitochondrial cytochrome c release or in activities of initiator caspase-8 and -9. However, there was greater activation of caspase-2 with anti-PMN treatment during cerulein pancreatitis. The upstream regulation of caspases-2 includes p53, which was increased; the p53 negative regulator, Mdm2, was decreased by anti-PMN treatment during cerulein pancreatitis. In vitro experiments using isolated pancreatic acinar cells a pharmacological inhibitor of Mdm2 increased caspase-2/-3 activities, and an inhibitor of p53 decreased these activities during cholecystokinin-8 treatment. Furthermore, experiments using the AR42J cell line Mdm2 small interfering RNA (siRNA) increased caspase-2/-3 activities, and p53 siRNA decreased these activities during cholecystokinin-8 treatment. These results suggest that during acute pancreatitis the inflammatory response inhibits apoptosis. The mechanism of this inhibition involves caspase-2 and its upstream regulation by p53 and Mdm2. Because previous findings indicate that promotion of apoptosis decreases necrosis and severity of pancreatitis, these results suggest that strategies to inhibit Mdm2 or activate p53 will have beneficial effects for treatment of pancreatitis.
Collapse
Affiliation(s)
- Yuji Nakamura
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California; ,2Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan;
| | - Jae Hyuk Do
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California; ,3Division of Gastroenterology, Department of Internal Medicine, Chung-Ang University School of Medicine, Seoul, Korea; and
| | - Jingzhen Yuan
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California;
| | - Irina V. Odinokova
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California; ,4Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Olga Mareninova
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California;
| | - Anna S. Gukovskaya
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California;
| | - Stephen J. Pandol
- 1Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, California;
| |
Collapse
|
79
|
Koh SL, Tan JW, Muralidharan V, Christophi C. The effect of hyperbaric oxygen on apoptosis and proliferation in severe acute pancreatitis. HPB (Oxford) 2009; 11:629-37. [PMID: 20495630 PMCID: PMC2799615 DOI: 10.1111/j.1477-2574.2009.00099.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 05/23/2009] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This paper investigates the significance of apoptosis in severe acute pancreatitis (SAP) and the possible modulating effects of hyperbaric oxygen (HBO). METHODS Wistar rats (250-350 g) were induced with SAP by biliopancreatic infusion of 4% sodium taurocholate. Rats were randomized for HBO treatment. Pancreatic tissue was stained for apoptosis with immunohistochemistry (anti-CASPASE-3 antibody and TUNEL), and histopathology haematoxylin and eosin (H&E). Acini were stained for proliferation with an anti-KI67 antibody. ImageProPlus was used to quantify apoptosis and proliferation in acinar cells. Statistical analysis was performed with two-independent-sample t-test or non-parametric Mann-Whitney test. RESULTS In normal acini there is a low rate of apoptosis (0.165 +/- 0.157%, 0.181 +/- 0.168%, 0.130 +/- 0.298% in CASPASE-3, H&E and TUNEL, respectively) and proliferation (0.951 +/- 0.926%) (mean +/- standard deviation [SD]). When compared with normal, apoptosis (CASPASE-3: 1.28 +/- 1.12%, P= 0.008; 2.40 +/- 3.04%, P= 0.101; 1.23 +/- 0.87%, P= 0.091; H&E: 0.47 +/- 0.36%, P= 0.051; 0.69 +/- 0.63%, P= 0.001; 0.68 +/- 0.28%, P= 0; TUNEL: 1.08 +/- 1.42%, P= 0; 1.96 +/- 1.87%, P= 0; 2.36 +/- 2.26%, P= 0) and proliferation (1.96 +/- 1.89%, P= 0.187; 1.73 +/- 1.76%, P= 0.165; 1.36 +/- 1.40%, P= 0.571) were increased on days 1, 2 and 3 post-induction, respectively. In comparison with the untreated controls, HBO increased apoptosis on day 1 (CASPASE-3: 3.11 +/- 1.97%, P= 0.04; H&E: 0.97 +/- 0.76%, P= 0.005) and day 2 (TUNEL: 3.61 +/- 3.05%, P= 0.034). Treatment with HBO increased proliferation (3.04 +/- 3.14%, P= 0.519; 7.33 +/- 7.55%, P= 0.153) on days 2 and 3, respectively, compared with the untreated controls. CONCLUSIONS During SAP, acini apoptosis and proliferation were increased. Hyperbaric oxygen therapy may improve the condition of SAP by promoting apoptosis and proliferation.
Collapse
Affiliation(s)
- Shir Lin Koh
- Department of Surgery, University of Melbourne, Austin Health Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
80
|
Babu BI, Malleo G, Genovese T, Mazzon E, Di Paola R, Crisafulli C, Caminiti R, Siriwardena AK, Cuzzocrea S. Green tea polyphenols ameliorate pancreatic injury in cerulein-induced murine acute pancreatitis. Pancreas 2009; 38:954-67. [PMID: 19672210 DOI: 10.1097/mpa.0b013e3181b28d11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Green tea polyphenols (GTPs) are naturally occurring antioxidants acting through pathways that include reactive oxygen species and nuclear factor kappa B (NF-kappaB). This study investigates the effect of GTPs in a cerulein-induced murine model of acute pancreatitis (AP). METHODS Male CD mice (median weight, 37.7 g) were divided into 4 groups: mice administered with cerulein alone, cerulein and GTP, saline alone (sham), and GTP alone. Acute pancreatitis was induced by serial intraperitoneal administration of cerulein (50 microg/kg, x6). Green tea polyphenol was administered intraperitoneally at 25 mg/kg on the first, third, and sixth hours after pancreatitis induction.We analyzed histologic and biochemical features of AP, NF-kappaB pathway activity, leukocyte-mediated damage, cytokine levels, oxidative stress injury, lipid peroxidation, expression of poly-(adenosine diphosphate-ribose) synthetase, and presence of apoptosis. RESULTS Treatment with GTP reduced the histologic and biochemical features of AP. Western blot revealed significant NF-kappaB inactivation. Immunostaining for P selectin and intercellular adhesion molecule 1, tumor necrosis factor alpha, transforming growth factor beta, vascular endothelial growth factor, nitrotirosine, poly-(adenosine diphosphate ribose) synthetase, and malondialdheide levels were significantly reduced. There was a significant down-regulation of apoptotic markers. CONCLUSIONS Our results demonstrated that GTP significantly ameliorated the effects of cerulein-induced AP in mice. These effects of GTP are mediated by actions at the NF-kappaB/IkB (inhibitor kB) proteins and oxidative stress pathways.
Collapse
Affiliation(s)
- Benoy I Babu
- Manchester Royal Infirmary, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Singh VP, Bren GD, Algeciras-Schimnich A, Schnepple D, Navina S, Rizza SA, Dawra RK, Saluja AK, Chari ST, Vege SS, Badley AD. Nelfinavir/ritonavir reduces acinar injury but not inflammation during mouse caerulein pancreatitis. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1040-6. [PMID: 19282375 PMCID: PMC2696210 DOI: 10.1152/ajpgi.90642.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 03/04/2009] [Indexed: 01/31/2023]
Abstract
There is no clinical treatment that reduces acinar injury during pancreatitis. Human immunodeficiency virus (HIV) protease inhibitors (PI), including nelfinavir (NFV) and ritonavir (RTV), may reduce the rate of pancreatitis in HIV-infected patients. Since permeability transition pore (PTPC)-mediated mitochondrial dysfunction occurs during pancreatitis, and we have shown that PI prevents PTPC opening, we studied its effects in a model of pancreatitis. The effect of NFV plus RTV (NFV/RTV) or vehicle on caerulein-induced pancreatitis in mice was compared by measuring changes in mitochondrial membrane potential in vitro and cytochrome c leakage in vivo. Histological and inflammatory makers were also compared. NFV/RTV improved DiOC6 retention in acini exposed to caerulein in vitro. In vivo NFV prevented cytosolic leakage of cytochrome c and reduced pancreatic acinar injury, active caspase-3 staining, TUNEL-positive acinar cells, and serum amylase (P < 0.05). Conversely, trypsin activity, serum cytokine levels, and pancreatic and lung inflammation were unaffected. NFV/RTV reduces pancreatic injury and acinar cell death in experimental mouse caerulein-induced pancreatitis but does not impact inflammation.
Collapse
Affiliation(s)
- Vijay P Singh
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, Bhanot U, Hasel C, Möller P, Gschwend JE, Simmet T, Debatin KM, Fulda S. Small molecule XIAP inhibitors enhance TRAIL-induced apoptosis and antitumor activity in preclinical models of pancreatic carcinoma. Cancer Res 2009; 69:2425-34. [PMID: 19258513 DOI: 10.1158/0008-5472.can-08-2436] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Evasion of apoptosis is a characteristic feature of pancreatic cancer, a prototypic cancer that is refractory to current treatment approaches. Hence, there is an urgent need to design rational strategies that counter apoptosis resistance. To explore X-linked inhibitor of apoptosis (XIAP) as a therapeutic target in pancreatic cancer, we analyzed the expression of XIAP in pancreatic tumor samples and evaluated the effect of small molecule XIAP inhibitors alone and in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) against pancreatic carcinoma in vitro and in vivo. Here, we report that XIAP is highly expressed in pancreatic adenocarcinoma samples compared with normal pancreatic ducts. Small molecule XIAP inhibitors synergize with TRAIL to induce apoptosis and to inhibit long-term clonogenic survival of pancreatic carcinoma cells. In contrast, they do not reverse the lack of toxicity of TRAIL on nonmalignant cells in vitro or normal tissues in vivo, pointing to a therapeutic index. Most importantly, XIAP inhibitors cooperate with TRAIL to trigger apoptosis and suppress pancreatic carcinoma growth in vivo in two preclinical models, i.e., the chorioallantoic membrane model and a mouse xenograft model. Parallel immunohistochemical analysis of tumor tissue under therapy reveals that the XIAP inhibitor acts in concert with TRAIL to cause caspase-3 activation and apoptosis. In conclusion, our findings provide, for the first time, evidence in vivo that XIAP inhibitors prime pancreatic carcinoma cells for TRAIL-induced apoptosis and potentiate the antitumor activity of TRAIL against established pancreatic carcinoma. These findings build the rationale for further (pre)clinical development of XIAP inhibitors and TRAIL against pancreatic cancer.
Collapse
Affiliation(s)
- Meike Vogler
- University Children's Hospital, Ulm University, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Meriläinen S, Mäkelä J, Anttila V, Koivukangas V, Kaakinen H, Niemelä E, Ohtonen P, Risteli J, Karttunen T, Soini Y, Juvonen T. Acute edematous and necrotic pancreatitis in a porcine model. Scand J Gastroenterol 2009; 43:1259-68. [PMID: 18609130 DOI: 10.1080/00365520802158580] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE It is unclear why pancreatitis progresses either to mild edematous disease or to severe necrotic disease. The aim of the study was to shed some light on this topic by investigating differences during the early stages of necrotic and edematous pancreatitis. MATERIAL AND METHODS Piglets were randomized into two groups. Necrotic pancreatitis was induced with retrograde injection of 20% taurocholic acid (1 ml/kg), and edematous pancreatitis was induced with 0.9% NaCl (1 ml/kg). Central hemodynamics was measured, and pancreatic microcirculation was directly examined by intravital microscopy. Vascular permeability to proteins and albumin was measured by microdialysis. Apoptosis and claudins 2, 3, 4, 5, and 7 were analyzed from pancreatic tissue samples. Blood samples were taken for analysis of blood cell counts, blood gases, lipase, and amylase. RESULTS Hemodynamic changes were similar in both groups, whereas microcirculatory impairment was more pronounced in necrotic pancreatitis. Necrosis was associated only with necrotic pancreatitis. Apoptosis increased only in edematous pancreatitis. The number of blood neutrophils and monocytes increased and lymphocyte and platelet counts decreased in both groups. Necrotic pancreatitis was associated with increased permeability to albumin and proteins. Expression of claudins 3, 4, 5, and 7 was not changed during pacreatitis, but in acinar cells, membranous expression of claudin-2 increased in both groups. CONCLUSIONS The results show that acute edematous pancreatitis is characterized by induction of apoptosis, whereas full-blown pancreatitis is characterized by necrosis. Impaired vascular permeability to albumin and protein is related to the early phase of necrotic pancreatitis. Claudin-2 increases during acute necrotic and edematous pancreatitis and may be related to impaired permeability.
Collapse
Affiliation(s)
- Sanna Meriläinen
- Department of Surgery, Oulu University Hospital, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Sung KF, Odinokova IV, Mareninova OA, Rakonczay Z, Hegyi P, Pandol SJ, Gukovsky I, Gukovskaya AS. Prosurvival Bcl-2 proteins stabilize pancreatic mitochondria and protect against necrosis in experimental pancreatitis. Exp Cell Res 2009; 315:1975-89. [PMID: 19331832 DOI: 10.1016/j.yexcr.2009.01.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 01/14/2009] [Accepted: 01/14/2009] [Indexed: 02/07/2023]
Abstract
Acinar cells in pancreatitis die through apoptosis and necrosis, the roles of which are different. The severity of experimental pancreatitis correlates directly with the extent of necrosis and inversely, with apoptosis. Apoptosis is mediated by the release of cytochrome c into the cytosol followed by caspase activation, whereas necrosis is associated with the mitochondrial membrane potential (DeltaPsim) loss leading to ATP depletion. Here, we investigate the role of Bcl-2 proteins in apoptosis and necrosis in pancreatitis. We found up-regulation of prosurvival Bcl-2 proteins in pancreas in various experimental models of acute pancreatitis, most pronounced for Bcl-xL. This up-regulation translated into increased levels of Bcl-xL and Bcl-2 in pancreatic mitochondria. Bcl-xL/Bcl-2 inhibitors induced DeltaPsim loss and cytochrome c release in isolated mitochondria. Corroborating the results on mitochondria, Bcl-xL/Bcl-2 inhibitors induced DeltaPsim loss, ATP depletion and necrosis in pancreatic acinar cells, both untreated and hyperstimulated with CCK-8 (in vitro pancreatitis model). Together Bcl-xL/Bcl-2 inhibitors and CCK induced more necrosis than either treatment alone. Bcl-xL/Bcl-2 inhibitors also stimulated cytochrome c release in acinar cells leading to caspase-3 activation and apoptosis. However, different from their effect on pronecrotic signals, the stimulation by Bcl-xL/Bcl-2 inhibitors of apoptotic responses was less in CCK-treated than control cells. Therefore, Bcl-xL/Bcl-2 inhibitors potentiated CCK-induced necrosis but not apoptosis. Correspondingly, transfection with Bcl-xL siRNA stimulated necrosis but not apoptosis in the in vitro pancreatitis model. Further, in animal models of pancreatitis Bcl-xL up-regulation inversely correlated with necrosis, but not apoptosis. Results indicate that Bcl-xL and Bcl-2 protect acinar cells from necrosis in pancreatitis by stabilizing mitochondria against death signals. We conclude that Bcl-xL/Bcl-2 inhibition would aggravate acute pancreatitis, whereas Bcl-xL/Bcl-2 up-regulation presents a strategy to prevent or attenuate necrosis in pancreatitis.
Collapse
Affiliation(s)
- Kai-Feng Sung
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
Apoptosis, or programmed cell death, is a physiological process of cellular autodestruction, or cell suicide. This process is strictly controlled in response to integrity of pro-death signaling and plays critical roles in development, maintenance of homeostasis and host defense in multicellular organisms. As pancreatologists, apoptosis plays a central role in the pancreas and its disease states, from diabetes to pancreatitis to pancreatic cancer. In pancreatic beta-cells, apoptotic cell death is involved in the pathogenesis of diabetes, as signals from death receptors and DNA damage have been widely accepted as being triggers of apoptosis in beta-cells. During acute pancreatitis, this common clinical condition is of variable severity in which some patients experience mild, self-limited attacks while others manifest a severe, highly morbid, and frequently lethal attack. However, recent research in this area has demonstrated the importance of acinar cell death in the form of apoptosis and necrosis as a determinant of pancreatitis severity. In pancreatic cancer, various survival mechanisms have been shown to act in the prevention of cell death to result in promotion of tumor growth and metastasis. Thus, resistance of pancreatic cancer to apoptosis is the key factor preventing responses to therapies. Thus, it is for these reasons that in the current 'Primers on Molecular Pathways,' we take a closer look at the pathway cascade that is triggered during apoptosis.
Collapse
Affiliation(s)
- Gwen Lomberk
- Division of Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA.
| | | |
Collapse
|
86
|
Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, Gschwend JE, Simmet T, Debatin KM, Fulda S. Targeting XIAP bypasses Bcl-2-mediated resistance to TRAIL and cooperates with TRAIL to suppress pancreatic cancer growth in vitro and in vivo. Cancer Res 2008; 68:7956-65. [PMID: 18829553 DOI: 10.1158/0008-5472.can-08-1296] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Resistance to apoptosis is a hallmark of pancreatic cancer, a leading cause of cancer deaths. Therefore, novel strategies are required to target apoptosis resistance. Here, we report that the combination of X-linked inhibitor of apoptosis (XIAP) inhibition and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an effective approach to trigger apoptosis despite Bcl-2 overexpression and to suppress pancreatic cancer growth in vitro and in vivo. Knockdown of XIAP by RNA interference cooperates with TRAIL to induce caspase activation, loss of mitochondrial membrane potential, cytochrome c release, and apoptosis in pancreatic carcinoma cells. Loss of mitochondrial membrane potential and cytochrome c release are extensively inhibited by a broad range or caspase-3 selective caspase inhibitor and by RNAi-mediated silencing of caspase-3, indicating that XIAP inhibition enhances TRAIL-induced mitochondrial damage in a caspase-3-dependent manner. XIAP inhibition combined with TRAIL even breaks Bcl-2-imposed resistance by converting type II cells that depend on the mitochondrial contribution to the death receptor pathway to type I cells in which TRAIL-induced activation of caspase-3 and caspase-9 and apoptosis proceeds irrespective of high Bcl-2 levels. Most importantly, XIAP inhibition potentiates TRAIL-induced antitumor activity in two preclinical models of pancreatic cancer in vivo. In the chicken chorioallantoic membrane model, XIAP inhibition significantly enhances TRAIL-mediated apoptosis and suppression of tumor growth. In a tumor regression model in xenograft-bearing mice, XIAP inhibition acts in concert with TRAIL to cause even regression of established pancreatic carcinoma. Thus, this combination of XIAP inhibition plus TRAIL is a promising strategy to overcome apoptosis resistance of pancreatic cancer that warrants further investigation.
Collapse
Affiliation(s)
- Meike Vogler
- Divison of Apoptosis Regulation, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Greenhalf W, Malats N, Nilsson M, Bartsch D, Neoptolemos J. International registries of families at high risk of pancreatic cancer. Pancreatology 2008; 8:558-65. [PMID: 18818508 DOI: 10.1159/000159214] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To describe the need for multinational registries of families at high risk of pancreatic cancer and the issues surrounding identification of such families. RESULTS A consensus position was published describing surveillance of individuals at high risk of pancreatic cancer. Hereditary pancreatitis patients, people with Peutz-Jeghers syndrome, individuals with CDKN2A or BRCA1/2 mutations with a family history of pancreatic cancer and kindred with multiple pancreatic cancers were considered suitable for research-based screening. Mutations responsible for familial predisposition are mostly unknown, although BRCA2 mutations have been identified in some families and a mutation in the palladin gene has been shown to segregate with pancreatic cancer in one kindred. Specific autosomal dominant inheritance of pancreatic cancer risk seems to involve anticipation; this finding aids identification of families and determination of individual risk. Diabetes mellitus is an early symptom of pancreatic cancer, but recent publications suggest that it may not be a significant predisposing factor; this remains controversial. However, in the context of hereditary pancreatitis, diabetes probably does predispose to pancreatic cancer as shown in a recent description of French families. CONCLUSION Appropriate inclusion of patients within registries of high-risk families provides a framework for secondary screening and research on risk stratification and early tumorigenesis. and IAP.
Collapse
Affiliation(s)
- William Greenhalf
- Division of Surgery, University of Liverpool, Royal Liverpool University Hospital, Liverpool, UK.
| | | | | | | | | |
Collapse
|
88
|
Abstract
Autophagy is an evolutionarily preserved degradation process of cytoplasmic cellular constituents, which has been known for its role in protecting cells against stresses such as starvation and in eliminating defective subcellular structures. It is essentially a form of self-cannibalism - hence the name that means 'self-eating' - in which the cell breaks down its own components. By mostly morphological studies, autophagy has been linked to a variety of pathological processes such as neurodegenerative diseases and tumorigenesis, which highlights its biological and medical importance. However, whether autophagy protects from or causes disease is unclear. Autophagic morphology was described in human pancreatitis by Helin et al. in 1980. Actually, acute pancreatitis is one of the earlier pathological processes where autophagy has been described in a human tissue. Autophagy, autodigestion and cell death are early cellular events in acute pancreatitis. The aim of this review is to introduce a description of the autophagic process and to discuss the possible role of autophagy in acute pancreatitis.
Collapse
Affiliation(s)
- María I Vaccaro
- Department of Physiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
89
|
Hamacher R, Schmid RM, Saur D, Schneider G. Apoptotic pathways in pancreatic ductal adenocarcinoma. Mol Cancer 2008; 7:64. [PMID: 18652674 PMCID: PMC2515336 DOI: 10.1186/1476-4598-7-64] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 07/24/2008] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common causes of cancer related death. Despite the advances in understanding of the molecular pathogenesis, pancreatic cancer remains a major unsolved health problem. Overall, the 5-year survival rate is less than 5% demonstrating the insufficiency of current therapies. Most cytotoxic therapies induce apoptosis and PDAC cells have evolved a plethora of molecular mechanisms to assure survival. We will present anti-apoptotic strategies working at the level of the death receptors, the mitochondria or involving the caspase inhibitors of the IAP family. Furthermore, the survival function of the phosphotidylinositol-3' kinase (PI3K)/AKT- and NF-kappaB-pathways are illustrated. A detailed molecular knowledge of the anti-apoptotic mechanisms of PDAC cells will help to improve therapies for this dismal disease and therapeutic strategies targeting the programmed cell death machinery are in early preclinical and clinical development.
Collapse
Affiliation(s)
- Rainer Hamacher
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Roland M Schmid
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Dieter Saur
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Günter Schneider
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| |
Collapse
|
90
|
Edderkaoui M, Odinokova I, Ohno I, Gukovsky I, Go VLW, Pandol SJ, Gukovskaya AS. Ellagic acid induces apoptosis through inhibition of nuclear factor κB in pancreatic cancer cells. World J Gastroenterol 2008; 14:3672-80. [PMID: 18595134 PMCID: PMC2719230 DOI: 10.3748/wjg.14.3672] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the effect of ellagic acid on apop-tosis and proliferation in pancreatic cancer cells and to determine the mechanism of the pro-survival effects of ellagic acid.
METHODS: The effect of ellagic acid on apoptosis was assessed by measuring Phosphatidylserine externalization, caspase activity, mitochondrial membrane potential and DNA fragmentation; and proliferation by measuring DNA thymidine incorporation. Mitochondrial membrane potential was measured in permeabilized cells, and in isolated mitochondria. Nuclear factor κB (NF-κB) activity was measured by electromobility shift assay (EMSA).
RESULTS: We show that ellagic acid, a polyphenolic compound in fruits and berries, at concentrations 10 to 50 mmol/L stimulates apoptosis in human pancreatic adenocarcinoma cells. Further, ellagic acid decreases proliferation by up to 20-fold at 50 mmol/L. Ellagic acid stimulates the mitochondrial pathway of apoptosis associated with mitochondrial depolarization, cytochrome C release, and the downstream caspase activation. Ellagic acid does not directly affect mitochondria. Ellagic acid dose-dependently decreased NF-κB binding activity. Furthermore, inhibition of NF-κB activity using IkB wild type plasmid prevented the effect of ellagic acid on apoptosis.
CONCLUSION: Our data indicate that ellagic acid stimulates apoptosis through inhibition of the prosu-rvival transcription factor NF-κB.
Collapse
|
91
|
Abstract
Pancreatitis and Calcium Signalling was an international research workshop organized by the authors and held at the Liverpool Medical Institution, Liverpool, United Kingdom, from Sunday 12th to Tuesday 14th November 2006. The overall goal of the workshop was to review progress and explore new opportunities for understanding the mechanisms of acute pancreatitis with an emphasis on the role of pathological calcium signaling. The participants included those with significant interest and expertise in pancreatitis research and others who are in fields outside gastroenterology but with significant expertise in areas of cell biology relevant to pancreatitis. The workshop was designed to enhance interchange of ideas and collaborations, to engage and encourage younger researchers in the field, and promote biomedical research through the participating and supporting organizations and societies. The workshop was divided into 8 topic-oriented sessions. The sessions were: (1) Physiology and pathophysiology of calcium signaling; (2) Interacting signaling mechanisms; (3) Premature digestive enzyme activation; (4) Physiology Society Lecture: Aberrant Ca2+ signaling, bicarbonate secretion, and pancreatitis; (5) NFkappaB, cytokines, and immune mechanisms; (6) Mitochondrial injury; (7) Cell death pathways; and (8) Overview of areas for future research. In each session, speakers presented work appropriate to the topic followed by discussion of the material presented by the group. The publication of these proceedings is intended to provide a platform for enhancing research and therapeutic development for acute pancreatitis.
Collapse
|
92
|
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: 21] [Impact Index Per Article: 1.2] [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
|
93
|
Abstract
Current knowledge shows that pathophysiology of acute pancreatitis is characterized by intraacinar enzyme activation and subsequent dysregulation in immune response. Interactions between leukocytes, soluble mediators such as cytokines and vascular endothelium contribute to the systemic progression of the inflammatory response, whose entity may--in the end--determine disease severity and outcome. Recently, it has been shown that TNF-[alpha] may be a novel target for the treatment of acute pancreatitis; but the role of thalidomide, an immunomodulatory agent that inhibits TNF-(alpha) and angiogenesis, has not been investigated so far. The aim of the present study was to assess the effects of thalidomide in a murine model of necrotizing acute pancreatitis. Necrotizing acute pancreatitis was induced in mice by intraperitoneal injection of cerulein (hourly, x5, 50 microg/kg); in another group of animals, thalidomide was administered (200 mg/kg orally) at 1 h after first cerulein injection. After 24 h, biochemical, histological, and immunohistochemical evidences of acute pancreatitis developed in all cerulein-treated mice. On the contrary, pancreatitis histological features, amylase, lipase, TNF-alpha and IL-1beta levels, pancreas edema, and myeloperoxidase activity as well as immunohistochemical staining for inflammatory cytokines, leukocyte adhesion molecules, transforming growth factor [beta], vascular endothelial growth factor, and apoptosis-related proteins were found reduced in thalidomide-treated mice. Therefore, thalidomide treatment attenuates the development of acute pancreatitis caused by cerulein in mice. We propose that this evidence may help to clarify the role of anti-TNF-alpha and immunomodulatory agents in patients with acute pancreatitis.
Collapse
|
94
|
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
|
95
|
Malleo G, Mazzon E, Genovese T, Di Paola R, Muià C, Centorrino T, Siriwardena AK, Cuzzocrea S. Etanercept attenuates the development of cerulein-induced acute pancreatitis in mice: a comparison with TNF-alpha genetic deletion. Shock 2007; 27:542-51. [PMID: 17438460 DOI: 10.1097/01.shk.0000246900.50445.1d] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
TNF-alpha plays a pivotal role in the pathogenesis of acute pancreatitis. Recent studies have shown that TNF-alpha inhibition significantly ameliorates the course of experimental acute pancreatitis, but in this context, the effects of Etanercept, a novel anti-TNF-alpha agent, have not been investigated so far. The aims of the present study are (i) to assess the effects of pharmacological inhibition of TNF-alpha by means of Etanercept on the inflammatory response and apoptosis in a murine model of necrotizing acute pancreatitis and (ii) to compare the results to those observed in TNF-alpha receptor 1 knockout (TNFR1-KO) mice. Necrotizing acute pancreatitis was induced in TNF-alpha wild type for TNFR1 (WT) and TNFR1-KO mice by intraperitoneal injection of cerulein (hourly x5, 50 microg/kg). In another group of WT mice, Etanercept was administered (5 or 10 mg/kg, s.c.) at 1 h after first cerulein injection. Control groups received saline treatment. After 24 h, biochemical, histological, and immunohistochemical evidences of acute pancreatitis developed in all cerulein-treated mice; apoptosis was also present in the pancreas. Contrarily, pancreatitis histological features, amylase and lipase levels, pancreas water content, and myeloperoxidase activity were reduced in a similar degree in Etanercept-treated and TNFR1-KO mice. Likewise, in these two groups, immunohistochemical stainings and terminal deoxynucleotidyltransferase-mediated UTP nick-end labeling assay were found negative. TNF-alpha receptor 1 gene deletion and Etanercept administration ameliorate the course of experimental acute pancreatitis in a similar degree. Future studies on clinical applications of Etanercept in pancreatitis seem promising.
Collapse
Affiliation(s)
- Giuseppe Malleo
- Department of Clinical, Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
96
|
|
97
|
Baumgartner HK, Gerasimenko JV, Thorne C, Ashurst LH, Barrow SL, Chvanov MA, Gillies S, Criddle DN, Tepikin AV, Petersen OH, Sutton R, Watson AJM, Gerasimenko OV. Caspase-8-mediated apoptosis induced by oxidative stress is independent of the intrinsic pathway and dependent on cathepsins. Am J Physiol Gastrointest Liver Physiol 2007; 293:G296-307. [PMID: 17431216 DOI: 10.1152/ajpgi.00103.2007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cell-death programs executed in the pancreas under pathological conditions remain largely undetermined, although the severity of experimental pancreatitis has been found to depend on the ratio of apoptosis to necrosis. We have defined mechanisms by which apoptosis is induced in pancreatic acinar cells by the oxidant stressor menadione. Real-time monitoring of initiator caspase activity showed that caspase-9 (66% of cells) and caspase-8 (15% of cells) were activated within 30 min of menadione administration, but no activation of caspase-2, -10, or -12 was detected. Interestingly, when caspase-9 activation was inhibited, activation of caspase-8 was increased. Half-maximum activation (t(0.5)) of caspase-9 occurred within approximately 2 min and was identified at or in close proximity to mitochondria, whereas t(0.5) for caspase-8 occurred within approximately 26 min of menadione application and was distributed homogeneously throughout cells. Caspase-9 but not caspase-8 activation was blocked completely by the calcium chelator BAPTA or bongkrekic acid, an inhibitor of the mitochondrial permeability transition pore. In contrast, caspase-8 but not caspase-9 activation was blocked by the destruction of lysosomes (preincubation with Gly-Phe beta-naphthylamide, a cathepsin C substrate), loss of lysosomal acidity (bafilomycin A1), or inhibition of cathepsin L or D. Using pepstatin A-BODIPY FL conjugate, we confirmed translocation of cathepsin D out of lysosomes in response to menadione. We conclude that the oxidative stressor menadione induces two independent apoptotic pathways within pancreatic acinar cells: the classical mitochondrial calcium-dependent pathway that is initiated rapidly in the majority of cells, and a slower, caspase-8-mediated pathway that depends on the lysosomal activities of cathepsins and is used when the caspase-9 pathway is disabled.
Collapse
Affiliation(s)
- Heidi K Baumgartner
- The Physiological Laboratory, Biomedical Sciences, Liverpool University, Crown Street, Liverpool, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
This paper provides a summary of the effects of alcohol abuse on the pathobiologic responses that occur during acute and chronic pancreatitis considering both the human disease and animal/tissue models. The effects are multiple and include ones on cell death leading to necrosis; on inflammation resulting in a sensitized response to pancreatic stress; and fibrosis through effects of ethanol on pancreatic stellate cells and the plasminogen system. Although the effects of alcohol are multiple and complex, it is likely that a combination of a few key effects on these pathobiologic responses drive the increased sensitivity of the pancreas to acute pancreatitis with pancreatic stress and the promotion of chronic pancreatitis with pancreatic injury occurring during acute pancreatitis.
Collapse
Affiliation(s)
- Stephen J Pandol
- Department of Medicine, University of California, Department of Veterans Affairs, Los Angeles, California, USA.
| | | |
Collapse
|
99
|
Retzer-Lidl M, Schmid RM, Schneider G. Inhibition of CDK4 impairs proliferation of pancreatic cancer cells and sensitizes towards TRAIL-induced apoptosis via downregulation of survivin. Int J Cancer 2007; 121:66-75. [PMID: 17304504 DOI: 10.1002/ijc.22619] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pancreatic ductal adenocarcinoma is one of the most common causes of cancer death in Western countries with an average survival after diagnosis of 3-6 months and a five-year survival rate under 5%. Because of the lack of effective therapies, there is the need to characterize new molecular treatment strategies. Abnormal regulation of the cell cycle is a hallmark of neoplasia. Cyclin-dependent kinase 4 (CDK4), a key regulator of G1-phase of the cell cycle, has been shown to be overexpressed in pancreatic cancer. Until now, the contribution of CDK4 to tumor maintenance of pancreatic cancer has not been investigated. In this study, we used the chemical CDK4 inhibitor 2-bromo-12,13-dihydro-5H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-5,7(6H)-dione, as well as RNA interference, to investigate the function of CDK4 in pancreatic cancer cells. Both approaches led to a reduction of pancreatic cancer cell proliferation due to G1-phase cell cycle arrest and Rb activation. Furthermore, we observed increased sensitivity of G1-arrested pancreatic cancer cells towards TRAIL-induced apoptosis. Sensitization towards TRAIL was due to the transcriptional downregulation of survivin. These findings show that a combined sensitizer/inducer strategy may be a potential therapeutic strategy for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Michaela Retzer-Lidl
- II Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | | | | |
Collapse
|
100
|
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
|