101
|
Ding L, Liou GY, Schmitt DM, Storz P, Zhang JS, Billadeau DD. Glycogen synthase kinase-3β ablation limits pancreatitis-induced acinar-to-ductal metaplasia. J Pathol 2017. [PMID: 28639695 DOI: 10.1002/path.4928] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acinar-to-ductal metaplasia (ADM) is a reversible epithelial transdifferentiation process that occurs in the pancreas in response to acute inflammation. ADM can rapidly progress towards pre-malignant pancreatic intraepithelial neoplasia (PanIN) lesions in the presence of mutant KRas and ultimately pancreatic adenocarcinoma (PDAC). In the present work, we elucidate the role and related mechanism of glycogen synthase kinase-3beta (GSK-3β) in ADM development using in vitro 3D cultures and genetically engineered mouse models. We show that GSK-3β promotes TGF-α-induced ADM in 3D cultured primary acinar cells, whereas deletion of GSK-3β attenuates caerulein-induced ADM formation and PanIN progression in KrasG12D transgenic mice. Furthermore, we demonstrate that GSK-3β ablation influences ADM formation and PanIN progression by suppressing oncogenic KRas-driven cell proliferation. Mechanistically, we show that GSK-3β regulates proliferation by increasing the activation of S6 kinase. Taken together, these results indicate that GSK-3β participates in early pancreatitis-induced ADM and thus could be a target for the treatment of chronic pancreatitis and the prevention of PDAC progression. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Li Ding
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Jin-San Zhang
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA.,Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, and Center for Precision Medicine, The First Affiliated Hospital, Wenzhou Medical University; Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, PR China
| | - Daniel D Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
102
|
Murakami S, Shahbazian D, Surana R, Zhang W, Chen H, Graham GT, White SM, Weiner LM, Yi C. Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma. Oncogene 2017; 36:1232-1244. [PMID: 27546622 PMCID: PMC5322249 DOI: 10.1038/onc.2016.288] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a high degree of inflammation and profound immune suppression. Here we identify Yes-associated protein (Yap) as a critical regulator of the immunosuppressive microenvironment in both mouse and human PDAC. Within Kras:p53 mutant pancreatic ductal cells, Yap drives the expression and secretion of multiple cytokines/chemokines, which in turn promote the differentiation and accumulation of myeloid-derived suppressor cells (MDSCs) both in vitro and in vivo. Pancreas-specific knockout of Yap or antibody-mediated depletion of MDSCs promoted macrophage reprogramming, reactivation of T cells, apoptosis of Kras mutant neoplastic ductal cells and pancreatic regeneration after acute pancreatitis. In primary human PDAC, YAP expression levels strongly correlate with an MDSC gene signature, and high expression of YAP or MDSC-related genes predicts decreased survival in PDAC patients. These results reveal multifaceted roles of YAP in PDAC pathogenesis and underscore its promise as a therapeutic target for this deadly disease.
Collapse
Affiliation(s)
- Shigekazu Murakami
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - David Shahbazian
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Rishi Surana
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Weiying Zhang
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hengye Chen
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Garrett T. Graham
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shannon M. White
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Louis M. Weiner
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chunling Yi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
103
|
Abstract
Acute pancreatitis (AP) is a rare manifestation of pancreatic cancer (PC). The relationship between AP and PC remains less distinct.From January 2009 to November 2015, 47consecutive patients with PC who presented with AP were reviewed for this study. Clinical features, clinicopathologic variables, postoperative complications, and follow-up evaluations of patients were documented in detail from our database. In order to identify cutoff threshold time for surgery, receiver operating curve (ROC) was built according to patients with or without postoperative complications. Cumulative rate of survival was calculated by using the Kaplan-Meier method. The study was conducted in accordance with the principles of the Declaration of Helsinki and the guidelines of West China Hospital.This study included 35 men (74.5%) and 12 women (25.5%) (mean age: 52 years), with a median follow-up of 40 months. AP was clinically mild in 45 (95.7%) and severe in 2 (4.3%). The diagnosis of PC was delayed by 2 to 660 days (median 101 days). Thirty-nine (83.0%) cases underwent surgery. Eight (17.0%) cases performed biopsies only. Of 39 patients, radical surgery was performed in 32 (82.1%) cases and palliative in 7 (19.9%) cases. Two (8.0%) patients were needed for vascular resection and reconstruction. Postoperative complications occurred in 12 (30.8%) patients. About 24.5 days was the best cutoff point, with an area under curve (AUC) of 0.727 (P = 0.025, 95% confidence interval: 0.555-0.8999). The survival rate of patients at 1 year was 23.4%. The median survival in patients with vascular resection and reconstruction was 18 months, compared with 10 months in patients without vascular resection (P = 0.042). For the primary stage (T), Tix was identified in 3 patients, the survival of whom were 5, 28, 50 months, respectively. And 2 of them were still alive at the follow-up period.The severity of AP was mainly mild. Surgical intervention after 24.5 days may benefit for reducing postoperative complications. Patients with vascular resection and reconstruction, thus achieving tumor-free margins, had a long-time survival.
Collapse
|
104
|
Asati V, Mahapatra DK, Bharti SK. K-Ras and its inhibitors towards personalized cancer treatment: Pharmacological and structural perspectives. Eur J Med Chem 2017; 125:299-314. [DOI: 10.1016/j.ejmech.2016.09.049] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023]
|
105
|
Leal AS, Sporn MB, Pioli PA, Liby KT. The triterpenoid CDDO-imidazolide reduces immune cell infiltration and cytokine secretion in the KrasG12D;Pdx1-Cre (KC) mouse model of pancreatic cancer. Carcinogenesis 2016; 37:1170-1179. [PMID: 27659181 DOI: 10.1093/carcin/bgw099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/11/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
Abstract
Because the 5-year survival rate for pancreatic cancer remains under 10%, new drugs are needed for the prevention and treatment of this devastating disease. Patients with chronic pancreatitis have a 12-fold higher risk of developing pancreatic cancer. LSL-KrasG12D/+;Pdx-1-Cre (KC) mice replicate the genetics, symptoms and histopathology found in human pancreatic cancer. Immune cells infiltrate into the pancreas of these mice and produce inflammatory cytokines that promote tumor growth. KC mice are particularly sensitive to the effects of lipopolysaccharide (LPS), as only 48% of KC mice survived an LPS challenge while 100% of wildtype (WT) mice survived. LPS also increased the percentage of CD45+ immune cells in the pancreas and immunosuppressive Gr1+ myeloid-derived suppressor cell in the spleen of these mice. The triterpenoid CDDO-imidazolide (CDDO-Im) not only reduced the lethal effects of LPS (71% survival) but also decreased the infiltration of CD45+ cells into the pancreas and the percentage of Gr1+ myeloid-derived suppressor cell in the spleen of KC mice 4-8 weeks after the initial LPS challenge. While the levels of inflammatory cytokine levels were markedly higher in KC mice versus WT mice challenged with LPS, CDDO-Im significantly decreased the production of IL-6, CCL-2, vascular endothelial growth factor and G-CSF in the KC mice. All of these cytokines are prognostic markers in pancreatic cancer or play important roles in the progression of this disease. Disrupting the inflammatory process with drugs such as CDDO-Im might be useful for preventing pancreatic cancer, especially in high-risk populations.
Collapse
Affiliation(s)
- Ana S Leal
- Department of Pharmacology, Geisel School of Medicine at Dartmouth, Hanover, NH 03756, USA.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA and
| | - Michael B Sporn
- Department of Pharmacology, Geisel School of Medicine at Dartmouth, Hanover, NH 03756, USA
| | - Patricia A Pioli
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Karen T Liby
- Department of Pharmacology, Geisel School of Medicine at Dartmouth, Hanover, NH 03756, USA, .,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA and
| |
Collapse
|
106
|
Hidalgo-Sastre A, Brodylo RL, Lubeseder-Martellato C, Sipos B, Steiger K, Lee M, von Figura G, Grünwald B, Zhong S, Trajkovic-Arsic M, Neff F, Schmid RM, Siveke JT. Hes1 Controls Exocrine Cell Plasticity and Restricts Development of Pancreatic Ductal Adenocarcinoma in a Mouse Model. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2934-2944. [PMID: 27639167 DOI: 10.1016/j.ajpath.2016.07.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 06/18/2016] [Accepted: 07/15/2016] [Indexed: 02/09/2023]
Abstract
Perturbation of pancreatic acinar cell state can lead to acinar-to-ductal metaplasia (ADM), a precursor lesion to the development of pancreatic ductal adenocarcinoma (PDAC). In the pancreas, Notch signaling is active both during development and in adult cellular differentiation processes. Hes1, a key downstream target of the Notch signaling pathway, is expressed in the centroacinar compartment of the adult pancreas as well as in both preneoplastic and malignant lesions. In this study, we used a murine genetic in vivo approach to ablate Hes1 in pancreatic progenitor cells (Ptf1a+/Cre; Hes1fl/fl). Using this model, we studied the role of Hes1 in both acinar cell plasticity and pancreatic regeneration after caerulein-induced pancreatitis and in KrasG12D-driven PDAC development. We show that, although pancreatic development is not perturbed on the deletion of Hes1, terminal acinar differentiation in the adult pancreas is compromised. Moreover, the loss of Hes1 leads to the impaired regeneration of the exocrine compartment, accelerated fatty metaplasia, and persistent ADM after acute caerulein-induced pancreatitis. In KrasG12D-driven carcinogenesis, Hes1 ablation resulted in increased ADM, decreased formation of high-grade pancreatic intraepithelial neoplasias, and accelerated development of PDAC with shortened survival time. In conclusion, Hes1 plays a key role in acinar cell integrity and plasticity on cellular insults. Furthermore, Hes1 is an essential component of the pancreatic intraepithelial neoplasias-to-PDAC route in KrasG12D-driven mouse pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Ana Hidalgo-Sastre
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Roxanne L Brodylo
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Clara Lubeseder-Martellato
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bence Sipos
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Katja Steiger
- Comparative Experimental Pathology Unit, Institute for General Pathology and Pathological Anatomy, Technical University of Munich, Munich, Germany
| | - Marcel Lee
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Guido von Figura
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Barbara Grünwald
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Suyang Zhong
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marija Trajkovic-Arsic
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Florian Neff
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Roland M Schmid
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Jens T Siveke
- II Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.
| |
Collapse
|
107
|
Gruber R, Panayiotou R, Nye E, Spencer-Dene B, Stamp G, Behrens A. YAP1 and TAZ Control Pancreatic Cancer Initiation in Mice by Direct Up-regulation of JAK-STAT3 Signaling. Gastroenterology 2016; 151:526-39. [PMID: 27215660 PMCID: PMC5007286 DOI: 10.1053/j.gastro.2016.05.006] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/27/2016] [Accepted: 05/14/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Pancreatitis is the most important risk factor for pancreatic ductal adenocarcinoma (PDAC). Pancreatitis predisposes to PDAC because it induces a process of acinar cell reprogramming known as acinar-to-ductal metaplasia (ADM)-a precursor of pancreatic intraepithelial neoplasia lesions that can progress to PDAC. Mutations in KRAS are found at the earliest stages of pancreatic tumorigenesis, and it appears to be a gatekeeper to cancer progression. We investigated how mutations in KRAS cooperate with pancreatitis to promote pancreatic cancer progression in mice. METHODS We generated mice carrying conditional alleles of Yap1 and Taz and disrupted Yap1 and Taz using a Cre-lox recombination strategy in adult mouse pancreatic acinar cells (Yap1fl/fl;Tazfl/fl;Ela1-CreERT2). We crossed these mice with LSL-KrasG12D mice, which express a constitutively active form of KRAS after Cre recombination. Pancreatic tumor initiation and progression were analyzed after chemically induced pancreatitis. We analyzed pancreatic tissues from patients with pancreatitis or PDAC by immunohistochemistry. RESULTS Oncogenic activation of KRAS in normal, untransformed acinar cells in the pancreatic tissues of mice resulted in increased levels of pancreatitis-induced ADM. Expression of the constitutive active form of KRAS in this system led to activation of the transcriptional regulators YAP1 and TAZ; their function was required for pancreatitis-induced ADM in mice. The JAK-STAT3 pathway was a downstream effector of KRAS signaling via YAP1 and TAZ. YAP1 and TAZ directly mediated transcriptional activation of several genes in the JAK-STAT3 signaling pathway; this could be a mechanism by which acinar cells that express activated KRAS become susceptible to inflammation. CONCLUSIONS We identified a mechanism by which oncogenic KRAS facilitates ADM and thereby generates the cells that initiate neoplastic progression. This process involves activation of YAP1 and TAZ in acinar cells, which up-regulate JAK-STAT3 signaling to promote development of PDAC in mice.
Collapse
Affiliation(s)
- Ralph Gruber
- Mammalian Genetics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Richard Panayiotou
- Transcription Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Emma Nye
- Experimental Histopathology, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Gordon Stamp
- Experimental Histopathology, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Axel Behrens
- Mammalian Genetics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK; School of Medicine, King's College London, London, UK.
| |
Collapse
|
108
|
Sato S, Nakamura F, Hiroshima Y, Nagashima Y, Kato I, Yamashita N, Goshima Y, Endo I. Caerulein-induced pancreatitis augments the expression and phosphorylation of collapsin response mediator protein 4. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2016; 23:422-31. [PMID: 27207309 DOI: 10.1002/jhbp.361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/19/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic pancreatitis is a significant risk factor for pancreatic cancer. Previously, we demonstrated that the pancreatic cancer cells show enhanced expression of collapsin response mediator protein 4 (CRMP4) that strongly correlates with severe venous invasion, liver metastasis, and poor prognosis. However, involvement of CRMP4 in acute or chronic pancreatitis remains unknown. METHODS Acute and chronic pancreatitis mice models were developed by periodic injection of caerulein. The expression levels of CRMP4 and its phosphorylation were examined. RESULTS Elevated CRMP4 levels were observed in the infiltrated lymphocytes as well as in the pancreas parenchyma of both acute and chronic pancreatitis. The expression pattern of phosphorylated CRMP4 was similar to that of CRMP4. Cdk5 partially co-localized with the phosphorylated CRMP4. CONCLUSIONS Pancreatitis induces CRMP4 expression in the pancreas parenchyma and in the infiltrated lymphocytes. Overlapping expression of CRMP4 and Cdk5 may suggest that the Cdk5 is at least, in part, responsible for the phosphorylation of CRMP4. The results suggest that CRMP4 is involved in the inflammatory response in pancreatitis. Understanding the mechanisms of CRMP4 would help us to develop novel therapeutic strategies against acute or chronic pancreatitis, and pancreatic cancer.
Collapse
Affiliation(s)
- Sho Sato
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Yukihiko Hiroshima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoji Nagashima
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
109
|
Seifert L, Werba G, Tiwari S, Giao Ly NN, Nguy S, Alothman S, Alqunaibit D, Avanzi A, Daley D, Barilla R, Tippens D, Torres-Hernandez A, Hundeyin M, Mani VR, Hajdu C, Pellicciotta I, Oh P, Du K, Miller G. Radiation Therapy Induces Macrophages to Suppress T-Cell Responses Against Pancreatic Tumors in Mice. Gastroenterology 2016; 150:1659-1672.e5. [PMID: 26946344 PMCID: PMC4909514 DOI: 10.1053/j.gastro.2016.02.070] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/22/2016] [Accepted: 02/25/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The role of radiation therapy in the treatment of patients with pancreatic ductal adenocarcinoma (PDA) is controversial. Randomized controlled trials investigating the efficacy of radiation therapy in patients with locally advanced unresectable PDA have reported mixed results, with effects ranging from modest benefit to worse outcomes compared with control therapies. We investigated whether radiation causes inflammatory cells to acquire an immune-suppressive phenotype that limits the therapeutic effects of radiation on invasive PDAs and accelerates progression of preinvasive foci. METHODS We investigated the effects of radiation therapy in p48(Cre);LSL-Kras(G12D) (KC) and p48(Cre);LSLKras(G12D);LSL-Trp53(R172H) (KPC) mice, as well as in C57BL/6 mice with orthotopic tumors grown from FC1242 cells derived from KPC mice. Some mice were given neutralizing antibodies against macrophage colony-stimulating factor 1 (CSF1 or MCSF) or F4/80. Pancreata were exposed to doses of radiation ranging from 2 to 12 Gy and analyzed by flow cytometry. RESULTS Pancreata of KC mice exposed to radiation had a higher frequency of advanced pancreatic intraepithelial lesions and more foci of invasive cancer than pancreata of unexposed mice (controls); radiation reduced survival time by more than 6 months. A greater proportion of macrophages from radiation treated invasive and preinvasive pancreatic tumors had an immune-suppressive, M2-like phenotype compared with control mice. Pancreata from mice exposed to radiation had fewer CD8(+) T cells than controls, and greater numbers of CD4(+) T cells of T-helper 2 and T-regulatory cell phenotypes. Adoptive transfer of T cells from irradiated PDA to tumors of control mice accelerated tumor growth. Radiation induced production of MCSF by PDA cells. A neutralizing antibody against MCSF prevented radiation from altering the phenotype of macrophages in tumors, increasing the anti-tumor T-cell response and slowing tumor growth. CONCLUSIONS Radiation treatment causes macrophages murine PDA to acquire an immune-suppressive phenotype and disabled T-cell-mediated anti-tumor responses. MCSF blockade negates this effect, allowing radiation to have increased efficacy in slowing tumor growth.
Collapse
Affiliation(s)
- Lena Seifert
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Gregor Werba
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Shaun Tiwari
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Nancy Ngoc Giao Ly
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Susanna Nguy
- Department of Radiation Oncology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Sara Alothman
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Dalia Alqunaibit
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Antonina Avanzi
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Donnele Daley
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Rocky Barilla
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Daniel Tippens
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Alejandro Torres-Hernandez
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Mautin Hundeyin
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Vishnu R Mani
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Cristina Hajdu
- Department of Pathology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Ilenia Pellicciotta
- Department of Pathology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Philmo Oh
- Department of Radiation Oncology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - Kevin Du
- Department of Radiation Oncology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York
| | - George Miller
- Department of Surgery, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York; Department of Cell Biology, S Arthur Localio Laboratory, New York University School of Medicine, New York, New York.
| |
Collapse
|
110
|
Furuyama T, Tanaka S, Shimada S, Akiyama Y, Matsumura S, Mitsunori Y, Aihara A, Ban D, Ochiai T, Kudo A, Fukamachi H, Arii S, Kawaguchi Y, Tanabe M. Proteasome activity is required for the initiation of precancerous pancreatic lesions. Sci Rep 2016; 6:27044. [PMID: 27244456 PMCID: PMC4886684 DOI: 10.1038/srep27044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Proteasome activity is significantly increased in advanced cancers, but its role in cancer initiation is not clear, due to difficulties in monitoring this process in vivo. We established a line of transgenic mice that carried the ZsGreen-degron(ODC) (Gdeg) proteasome reporter to monitor the proteasome activity. In combination with Pdx-1-Cre;LSL-Kras(G12D) model, proteasome activity was investigated in the initiation of precancerous pancreatic lesions (PanINs). Normal pancreatic acini in Gdeg mice had low proteasome activity. By contrast, proteasome activity was increased in the PanIN lesions that developed in Gdeg;Pdx-1-Cre;LSL-Kras(G12D) mice. Caerulein administration to Gdeg;Pdx-1-Cre;LSL-Kras(G12D) mice induced constitutive elevation of proteasome activity in pancreatic tissues and accelerated PanIN formation. The proteasome inhibitor markedly reduced PanIN formation in Gdeg;Pdx-1-Cre;LSL-Kras(G12D) mice (P = 0.001), whereas it had no effect on PanIN lesions that had already formed. These observations indicated the significance of proteasome activity in the initiation of PanIN but not the maintenance per se. In addition, the expressions of pERK and its downstream factors including cyclin D1, NF-κB, and Cox2 were decreased after proteasome inhibition in PanINs. Our studies showed activation of proteasome is required specifically for the initiation of PanIN. The roles of proteasome in the early stages of pancreatic carcinogenesis warrant further investigation.
Collapse
Affiliation(s)
- Takaki Furuyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Matsumura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Mitsunori
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Arihiro Aihara
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Ochiai
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroshi Fukamachi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeki Arii
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiya Kawaguchi
- Department of Clinical Application, Center for iPS cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
111
|
Abstract
The genetic landscape of pancreatic cancer shows nearly ubiquitous mutations of K-RAS. However, oncogenic K-Rasmt alone is not sufficient to lead to pancreatic ductal adenocarcinoma (PDAC) in either human or in genetically modified adult mouse models. Many stimulants, such as high fat diet, CCK, LPS, PGE2 and others, have physiological effects at low concentrations that are mediated in part through modest increases in K-Ras activity. However, at high concentrations, they induce inflammation that, in the presence of oncogenic K-Ras expression, substantially accelerates PDAC formation. The mechanism involves increased activity of oncogenic K-Rasmt. Unlike what has been proposed in the standard paradigm for the role of Ras in oncogenesis, oncogenic K-Rasmt is now known to not be constitutively active. Rather, it can be activated by standard mechanisms similar to wild-type K-Ras, but its activity is sustained for a prolonged period. Furthermore, if the level of K-Ras activity exceeds a threshold at which it begins to generate its own activators, then a feed-forward loop is formed between K-Ras activity and inflammation and pathological processes including oncogenesis are initiated. Oncogenic K-Rasmt activation, a key event in PDAC initiation and development, is subject to complex regulatory mechanisms. Reagents which inhibit inflammation, such as the Cox2 inhibitor celecoxib, block the feed-forward loop and prevent induction of PDAC in models with endogenous oncogenic K-Rasmt. Increased understanding of the role of activating and inhibitory mechanisms on oncogenic K-Rasmt activity is of paramount importance for the development of preventive and therapeutic strategies to fight against this lethal disease.
Collapse
Affiliation(s)
- Craig D Logsdon
- 1. Department of GI Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA; 2. Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Weiqin Lu
- 1. Department of GI Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| |
Collapse
|
112
|
Smith JP, Fonkoua LK, Moody TW. The Role of Gastrin and CCK Receptors in Pancreatic Cancer and other Malignancies. Int J Biol Sci 2016; 12:283-91. [PMID: 26929735 PMCID: PMC4753157 DOI: 10.7150/ijbs.14952] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The gastrointestinal (GI) peptide gastrin is an important regulator of the release of gastric acid from the stomach parietal cells and it also plays an important role in growth of the gastrointestinal tract. It has become apparent that gastrin and its related peptide cholecystokinin (CCK) are also significantly involved with growth of GI cancers as well as other malignancies through activation of the cholecystokinin-B (CCK-B) receptor. Of interest, gastrin is expressed in the embryologic pancreas but not in the adult pancreas; however, gastrin becomes re-expressed in pancreatic cancer where it stimulates growth of this malignancy by an autocrine mechanism. Strategies to down-regulate gastrin or interfere with its interface with the CCK receptor with selective antibodies or receptor antagonists hold promise for the treatment of pancreatic cancer and other gastrin--responsive tumors.
Collapse
Affiliation(s)
- Jill P Smith
- 1. Department of Medicine, Georgetown University, Washington, DC, USA
| | - Lionel K Fonkoua
- 2. Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Terry W Moody
- 3. National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
113
|
Silencing Mist1 Gene Expression Is Essential for Recovery from Acute Pancreatitis. PLoS One 2015; 10:e0145724. [PMID: 26717480 PMCID: PMC4696804 DOI: 10.1371/journal.pone.0145724] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/08/2015] [Indexed: 12/25/2022] Open
Abstract
Acinar cells of the exocrine pancreas are tasked with synthesizing, packaging and secreting vast quantities of pro-digestive enzymes to maintain proper metabolic homeostasis for the organism. Because the synthesis of high levels of hydrolases is potentially dangerous, the pancreas is prone to acute pancreatitis (AP), a disease that targets acinar cells, leading to acinar-ductal metaplasia (ADM), inflammation and fibrosis—events that can transition into the earliest stages of pancreatic ductal adenocarcinoma. Despite a wealth of information concerning the broad phenotype associated with pancreatitis, little is understood regarding specific transcriptional regulatory networks that are susceptible to AP and the role these networks play in acinar cell and exocrine pancreas responses. In this study, we examined the importance of the acinar-specific maturation transcription factor MIST1 to AP damage and organ recovery. Analysis of wild-type and Mist1 conditional null mice revealed that Mist1 gene transcription and protein accumulation were dramatically reduced as acinar cells underwent ADM alterations during AP episodes. To test if loss of MIST1 function was primarily responsible for the damaged status of the organ, mice harboring a Cre-inducible Mist1 transgene (iMist1) were utilized to determine if sustained MIST1 activity could alleviate AP damage responses. Unexpectedly, constitutive iMist1 expression during AP led to a dramatic increase in organ damage followed by acinar cell death. We conclude that the transient silencing of Mist1 expression is critical for acinar cells to survive an AP episode, providing cells an opportunity to suppress their secretory function and regenerate damaged cells. The importance of MIST1 to these events suggests that modulating key pancreas transcription networks could ease clinical symptoms in patients diagnosed with pancreatitis and pancreatic cancer.
Collapse
|
114
|
NFATc4 Regulates Sox9 Gene Expression in Acinar Cell Plasticity and Pancreatic Cancer Initiation. Stem Cells Int 2015; 2016:5272498. [PMID: 26697077 PMCID: PMC4677249 DOI: 10.1155/2016/5272498] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/12/2015] [Indexed: 11/20/2022] Open
Abstract
Acinar transdifferentiation toward a duct-like phenotype constitutes the defining response of acinar cells to external stress signals and is considered to be the initial step in pancreatic carcinogenesis. Despite the requirement for oncogenic Kras in pancreatic cancer (PDAC) development, oncogenic Kras is not sufficient to drive pancreatic carcinogenesis beyond the level of premalignancy. Instead, secondary events, such as inflammation-induced signaling activation of the epidermal growth factor (EGFR) or induction of Sox9 expression, are required for tumor formation. Herein, we aimed to dissect the mechanism that links EGFR signaling to Sox9 gene expression during acinar-to-ductal metaplasia in pancreatic tissue adaptation and PDAC initiation. We show that the inflammatory transcription factor NFATc4 is highly induced and localizes in the nucleus in response to inflammation-induced EGFR signaling. Moreover, we demonstrate that NFATc4 drives acinar-to-ductal conversion and PDAC initiation through direct transcriptional induction of Sox9. Therefore, strategies designed to disrupt NFATc4 induction might be beneficial in the prevention or therapy of PDAC.
Collapse
|
115
|
Cho JH, Choi JS, Hwang ET, Park JY, Jeon TJ, Kim HM, Cho JH. Usefulness of scheduled follow-up CT in discharged patients with acute pancreatitis. Pancreatology 2015; 15:642-6. [PMID: 26422300 DOI: 10.1016/j.pan.2015.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/17/2015] [Accepted: 09/07/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Follow-up computed tomography (CT) in patients with acute pancreatitis has been advocated but rarely studied. The aim of this study was to determine whether follow-up CT for acute pancreatitis might be helpful in establishing the prognosis or complications, and in determining a selected subgroup of patients for whom computed tomography could be beneficial. METHODS Between January 2010 and December 2012, patients with acute pancreatitis who underwent follow-up CT in the outpatient department between one and three months after discharge were retrospectively enrolled. Events discovered on follow-up CT were defined as newly developed or increased pancreatic collection such as pseudocyst or walled off necrosis, and diagnosis of pancreatic cancer. RESULTS Ultimately, 106 asymptomatic patients were enrolled (mean age 50.24 ± 16, 74.5% male, 31.1% moderately severe and severe acute pancreatitis). The median duration of follow-up CT was 69 (31-90) days. On follow-up CT, 23 patients showed events (2 pancreatic cancer, 21 increasing or developed pancreatic collections). In multivariate analysis, the predictive factors for events on follow-up CT were CTSI ≥3 (OR 4.46, CI 1.08-18.43, p = 0.039) and BISAP ≥ 2 (OR 4.83, CI 1.08-21.55, p = 0.039). CONCLUSIONS Follow-up CT within three months after discharge may be helpful for acute pancreatitis patients with CTSI ≥ 3 points or BISAP score ≥ 2 points.
Collapse
Affiliation(s)
- Jeong Hyeon Cho
- Division of Gastroenterology, Department of Internal Medicine, Myongji Hospital, Goyang, South Korea
| | - Ja Sung Choi
- Division of Gastroenterology, Department of Internal Medicine, Catholic Kwandong University College of Medicine, Gangneung, South Korea
| | - Eui Tae Hwang
- Division of Gastroenterology, Department of Internal Medicine, Myongji Hospital, Goyang, South Korea
| | - Ji Young Park
- Division of Gastroenterology, Department of Internal Medicine, Catholic Kwandong University College of Medicine, Gangneung, South Korea
| | - Tae Joo Jeon
- Division of Gastroenterology, Department of Internal Medicine, Inje University College of Medicine, Sanggye Paik Hospital, Seoul, South Korea
| | - Hee Man Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Jae Hee Cho
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, South Korea.
| |
Collapse
|
116
|
Pin CL, Ryan JF, Mehmood R. Acinar cell reprogramming: a clinically important target in pancreatic disease. Epigenomics 2015; 7:267-81. [PMID: 25942535 DOI: 10.2217/epi.14.83] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acinar cells of the pancreas produce the majority of enzymes required for digestion and make up >90% of the cells within the pancreas. Due to a common developmental origin and the plastic nature of the acinar cell phenotype, these cells have been identified as a possible source of β cells as a therapeutic option for Type I diabetes. However, recent evidence indicates that acinar cells are the main source of pancreatic intraepithelial neoplasias (PanINs), the predecessor of pancreatic ductal adenocarcinoma (PDAC). The conversion of acinar cells to either β cells or precursors to PDAC is dependent on reprogramming of the cells to a more primitive, progenitor-like phenotype, which involves changes in transcription factor expression and activity, and changes in their epigenetic program. This review will focus on the mechanisms that promote acinar cell reprogramming, as well as the factors that may affect these mechanisms.
Collapse
Affiliation(s)
- Christopher L Pin
- Department of Paediatrics, Physiology & Pharmacology, & Oncology, University of Western Ontario, London, ON N6C 2V5, Canada
| | | | | |
Collapse
|
117
|
Yu C, Huo X, Agoston AT, Zhang X, Theiss AL, Cheng E, Zhang Q, Zaika A, Pham TH, Wang DH, Lobie PE, Odze RD, Spechler SJ, Souza RF. Mitochondrial STAT3 contributes to transformation of Barrett's epithelial cells that express oncogenic Ras in a p53-independent fashion. Am J Physiol Gastrointest Liver Physiol 2015; 309:G146-61. [PMID: 26045618 PMCID: PMC4525109 DOI: 10.1152/ajpgi.00462.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/31/2015] [Indexed: 01/31/2023]
Abstract
Metaplastic epithelial cells of Barrett's esophagus transformed by the combination of p53-knockdown and oncogenic Ras expression are known to activate signal transducer and activator of transcription 3 (STAT3). When phosphorylated at tyrosine 705 (Tyr705), STAT3 functions as a nuclear transcription factor that can contribute to oncogenesis. STAT3 phosphorylated at serine 727 (Ser727) localizes in mitochondria, but little is known about mitochondrial STAT3's contribution to carcinogenesis in Barrett's esophagus, which is the focus of this study. We introduced a constitutively active variant of human STAT3 (STAT3CA) into the following: 1) non-neoplastic Barrett's (BAR-T) cells; 2) BAR-T cells with p53 knockdown; and 3) BAR-T cells that express oncogenic H-Ras(G12V). STAT3CA transformed only the H-Ras(G12V)-expressing BAR-T cells (evidenced by loss of contact inhibition, formation of colonies in soft agar, and generation of tumors in immunodeficient mice), and did so in a p53-independent fashion. The transformed cells had elevated levels of both mitochondrial (Ser727) and nuclear (Tyr705) phospho-STAT3. Introduction of a STAT3CA construct with a mutated tyrosine phosphorylation site into H-Ras(G12V)-expressing Barrett's cells resulted in high levels of mitochondrial phospho-STAT3 (Ser727) with little or no nuclear phospho-STAT3 (Tyr705), and the cells still formed tumors in immunodeficient mice. Thus tyrosine phosphorylation of STAT3 is not required for tumor formation in Ras-expressing Barrett's cells. We conclude that mitochondrial STAT3 (Ser727) can contribute to oncogenesis in Barrett's cells that express oncogenic Ras. These findings suggest that agents targeting STAT3 might be useful for chemoprevention in patients with Barrett's esophagus.
Collapse
Affiliation(s)
- Chunhua Yu
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Xiaofang Huo
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Agoston T. Agoston
- 4Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massacusetts;
| | - Xi Zhang
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Arianne L. Theiss
- 6Baylor Research Institute, Baylor University Medical Center, Dallas, Texas;
| | - Edaire Cheng
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,7Department of Pediatrics, Children's Medical Center and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Qiuyang Zhang
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Alexander Zaika
- 8Departments of Surgery and Cancer Biology, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; and
| | - Thai H. Pham
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,3Department of Surgery, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas;
| | - David H. Wang
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,5Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Peter E. Lobie
- 9Cancer Science Institute of Singapore, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Robert D. Odze
- 4Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massacusetts;
| | - Stuart J. Spechler
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,5Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Rhonda F. Souza
- 1Esophageal Diseases Center, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,2Department of Medicine, Veterans Affairs North Texas Health Care System and the University of Texas Southwestern Medical Center, Dallas, Texas; ,5Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas;
| |
Collapse
|
118
|
Mills JC, Sansom OJ. Reserve stem cells: Differentiated cells reprogram to fuel repair, metaplasia, and neoplasia in the adult gastrointestinal tract. Sci Signal 2015; 8:re8. [PMID: 26175494 PMCID: PMC4858190 DOI: 10.1126/scisignal.aaa7540] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It has long been known that differentiated cells can switch fates, especially in vitro, but only recently has there been a critical mass of publications describing the mechanisms adult, postmitotic cells use in vivo to reverse their differentiation state. We propose that this sort of cellular reprogramming is a fundamental cellular process akin to apoptosis or mitosis. Because reprogramming can invoke regenerative cells from mature cells, it is critical to the long-term maintenance of tissues like the pancreas, which encounter large insults during adulthood but lack constitutively active adult stem cells to repair the damage. However, even in tissues with adult stem cells, like the stomach and intestine, reprogramming may allow mature cells to serve as reserve ("quiescent") stem cells when normal stem cells are compromised. We propose that the potential downside to reprogramming is that it increases risk for cancers that occur late in adulthood. Mature, long-lived cells may have years of exposure to mutagens. Mutations that affect the physiological function of differentiated, postmitotic cells may lead to apoptosis, but mutations in genes that govern proliferation might not be selected against. Hence, reprogramming with reentry into the cell cycle might unmask those mutations, causing an irreversible progenitor-like, proliferative state. We review recent evidence showing that reprogramming fuels irreversible metaplastic and precancerous proliferation in the stomach and pancreas. Finally, we illustrate how we think reprogrammed differentiated cells are likely candidates as cells of origin for cancers of the intestine.
Collapse
Affiliation(s)
- Jason C Mills
- Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
119
|
Gall TMH, Wasan H, Jiao LR. Pancreatic cancer: current understanding of molecular and genetic aetiologies. Postgrad Med J 2015; 91:594-600. [PMID: 26124188 DOI: 10.1136/postgradmedj-2014-133161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 06/11/2015] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the few cancers where prognosis has not improved over the past few decades. However, there have been several advances in our understanding of the disease leading to earlier detection and targeted therapeutic treatment. It is now understood that specific somatic and germline mutations lead to the development of the disease, and the risk factors associated with this are clearer. Further, several precursor lesions have been identified which, with early detection and surveillance, allows treatment before the development of carcinoma. PDAC can now be diagnosed with a high sensitivity and specificity following advances in radiology, and treatment can be commenced at an earlier stage of the disease. With continued research we are hopeful that the next decade will see an improved survival rate for all patients with pancreatic cancer.
Collapse
Affiliation(s)
- Tamara M H Gall
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital, London, UK
| | | | - Long R Jiao
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital, London, UK
| |
Collapse
|
120
|
Chen YH, Xie SM, Zhang H, Tan CL, Ke NW, Mai G, Liu XB. Clinical impact of preoperative acute pancreatitis in patients who undergo pancreaticoduodenectomy for periampullary tumors. World J Gastroenterol 2015; 21:6937-6943. [PMID: 26078571 PMCID: PMC4462735 DOI: 10.3748/wjg.v21.i22.6937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/11/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the impact of preoperative acute pancreatitis (PAP) on the surgical management of periampullary tumors. METHODS Fifty-eight patients with periampullary tumors and PAP were retrospectively analyzed. Thirty-four patients who underwent pancreaticoduodenectomy (PD) and 4 patients who underwent total pancreatectomy were compared with a control group of 145 patients without PAP during the same period. RESULTS The preoperative waiting time was significantly shorter for the concomitant PAP patients who underwent a resection (22.4 d vs 54.6 d, P < 0.001) compared to those who did not. The presence of PAP significantly increased the rate of severe complications (Clavien grade 3 or higher) (17.6% vs 4.8%, P = 0.019) and lengthened the hospital stay (19.5 d vs 14.5 d, P = 0.006). A multivariate logistic regression analysis revealed that PAP was an independent risk factor for postoperative pancreatic fistula (OR = 2.91; 95%CI: 1.10-7.68; P = 0.032) and severe complications (OR = 4.70; 95%CI: 1.48-14.96; P = 0.009) after PD. There was no perioperative mortality. CONCLUSION PAP significantly increases the incidence of severe complications and lengthens the hospital stay following PD. PD could be safely performed in highly selective patients with PAP.
Collapse
|
121
|
Leal-Lopes C, Velloso FJ, Campopiano JC, Sogayar MC, Correa RG. Roles of Commensal Microbiota in Pancreas Homeostasis and Pancreatic Pathologies. J Diabetes Res 2015; 2015:284680. [PMID: 26347203 PMCID: PMC4544440 DOI: 10.1155/2015/284680] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/09/2015] [Indexed: 12/12/2022] Open
Abstract
The pancreas plays a central role in metabolism, allowing ingested food to be converted and used as fuel by the cells throughout the body. On the other hand, the pancreas may be affected by devastating diseases, such as pancreatitis, pancreatic adenocarcinoma (PAC), and diabetes mellitus (DM), which generally results in a wide metabolic imbalance. The causes for the development and progression of these diseases are still controversial; therefore it is essential to better understand the underlying mechanisms which compromise the pancreatic homeostasis. The interest in the study of the commensal microbiome increased extensively in recent years, when many discoveries have illustrated its central role in both human physiology and maintenance of homeostasis. Further understanding of the involvement of the microbiome during the development of pathological conditions is critical for the improvement of new diagnostic and therapeutic approaches. In the present review, we discuss recent findings on the behavior and functions played by the microbiota in major pancreatic diseases and provide further insights into its potential roles in the maintenance of pancreatic steady-state activities.
Collapse
Affiliation(s)
- Camila Leal-Lopes
- Department of Biochemistry, Chemistry Institute, University of São Paulo, 05508-000 São Paulo, SP, Brazil
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Fernando J. Velloso
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Julia C. Campopiano
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Mari C. Sogayar
- Department of Biochemistry, Chemistry Institute, University of São Paulo, 05508-000 São Paulo, SP, Brazil
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Ricardo G. Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- *Ricardo G. Correa:
| |
Collapse
|
122
|
Baer R, Cintas C, Dufresne M, Cassant-Sourdy S, Schönhuber N, Planque L, Lulka H, Couderc B, Bousquet C, Garmy-Susini B, Vanhaesebroeck B, Pyronnet S, Saur D, Guillermet-Guibert J. Pancreatic cell plasticity and cancer initiation induced by oncogenic Kras is completely dependent on wild-type PI 3-kinase p110α. Genes Dev 2014; 28:2621-35. [PMID: 25452273 PMCID: PMC4248293 DOI: 10.1101/gad.249409.114] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/27/2014] [Indexed: 12/22/2022]
Abstract
Increased PI 3-kinase (PI3K) signaling in pancreatic ductal adenocarcinoma (PDAC) correlates with poor prognosis, but the role of class I PI3K isoforms during its induction remains unclear. Using genetically engineered mice and pharmacological isoform-selective inhibitors, we found that the p110α PI3K isoform is a major signaling enzyme for PDAC development induced by a combination of genetic and nongenetic factors. Inactivation of this single isoform blocked the irreversible transition of exocrine acinar cells into pancreatic preneoplastic ductal lesions by oncogenic Kras and/or pancreatic injury. Hitting the other ubiquitous isoform, p110β, did not prevent preneoplastic lesion initiation. p110α signaling through small GTPase Rho and actin cytoskeleton controls the reprogramming of acinar cells and regulates cell morphology in vivo and in vitro. Finally, p110α was necessary for pancreatic ductal cancers to arise from Kras-induced preneoplastic lesions by increasing epithelial cell proliferation in the context of mutated p53. Here we identify an in vivo context in which p110α cellular output differs depending on the epithelial transformation stage and demonstrate that the PI3K p110α is required for PDAC induced by oncogenic Kras, the key driver mutation of PDAC. These data are critical for a better understanding of the development of this lethal disease that is currently without efficient treatment.
Collapse
Affiliation(s)
- Romain Baer
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Célia Cintas
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Marlène Dufresne
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France
| | - Stéphanie Cassant-Sourdy
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Nina Schönhuber
- Department of Internal Medicine 2, Technische Universität München, 81675 Munich, Germany
| | - Laetitia Planque
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Hubert Lulka
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France
| | - Bettina Couderc
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France
| | - Corinne Bousquet
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Barbara Garmy-Susini
- UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Inserm, F-31000 Toulouse, France; Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France
| | - Bart Vanhaesebroeck
- Cell Signaling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Stéphane Pyronnet
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| | - Dieter Saur
- Department of Internal Medicine 2, Technische Universität München, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Julie Guillermet-Guibert
- UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France; UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France; Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France
| |
Collapse
|
123
|
Loc WS, Smith JP, Matters G, Kester M, Adair JH. Novel strategies for managing pancreatic cancer. World J Gastroenterol 2014; 20:14717-14725. [PMID: 25356034 PMCID: PMC4209537 DOI: 10.3748/wjg.v20.i40.14717] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/14/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023] Open
Abstract
With the incidence reports of pancreatic cancer increasing every year, research over the last several decades has been focused on the means to achieve early diagnosis in patients that are at a high risk of developing the malignancy. This review covers current strategies for managing pancreatic cancer and further discusses efforts in understanding the role of early onset symptoms leading to tumor progression. Recent investigations in this discussion include type 3c diabetes, selected biomarkers and pathways related to pancreatic intraepithelial neoplasia lesions, drug resistance, and advances in nanomedicine which may provide significant solutions for improving early detection and treatments in future medicine.
Collapse
|
124
|
Abstract
OBJECTIVES Exogenous administration of cholecystokinin (CCK) induces hypertrophy and hyperplasia of the pancreas with an increase in DNA content. We hypothesized that endogenous CCK is involved in the malignant progression of pancreatic intraepithelial neoplasia (PanIN) lesions and the fibrosis associated with pancreatic cancer. METHODS The presence of CCK receptors in early PanIN lesions was examined by immunohistochemistry in mouse and human pancreas. Pdx1-Cre/LSL-Kras transgenic mice were randomized to receive either untreated drinking water or water supplemented with a CCK receptor antagonist (proglumide, 0.1 mg/mL). Pancreas from the mice were removed and examined histologically for number and grade of PanINs after 1, 2, or 4 months of antagonist therapy. RESULTS Both CCK-A and CCK-B receptors were identified in early stage PanINs from mouse and human pancreas. The grade of PanIN lesions was reversed, and progression to advanced lesions arrested in mice treated with proglumide compared with the controls (P = 0.004). Furthermore, pancreatic fibrosis was significantly reduced in antagonist-treated animals compared with vehicle (P < 0.001). CONCLUSIONS These findings demonstrate that endogenous CCK is in part responsible for the development and progression of pancreatic cancer. The use of CCK receptor antagonists may have a role in cancer prophylaxis in high-risk subjects and may reduce fibrosis in the microenvironment.
Collapse
|
125
|
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most lethal diseases with an incidence rate almost equal to the rate of mortality. Chronic pancreatitis (CP) is a common chronic inflammatory disease of unknown etiology that affects the pancreas. Epidemiological studies have identified CP to be a major risk factor for PC. SUMMARY A greater understanding of the molecular mechanisms linking CP and PC has identified several common pathways that provide targets for future interventions. This article reviews those components in the CP-PC connection, including the role of macrophages, the maintenance of genome stability, cytokines, and other nodal factors such as nuclear factor kappa B, COX-2 and reactive oxygen species. KEY MESSAGE The molecular mechanisms that underlie CP and PC provide novel targets for future therapies for PC. PRACTICAL IMPLICATIONS The stromal-desmoplastic reaction plays an important role in initiating and sustaining chronic inflammation and tumor progression. Recently, two targeted anti-tumor agents, erlotinib and nab-paclitaxel, have shown promising therapeutic efficacy. Notably, both these agents target components (EGFR and SPARC) within the inflammatory stroma surrounding malignant cells, underscoring the importance of inflammation in pancreatic carcinogenesis. Identifying the common pathways linking CP and PC may help uncover additional novel targets for future therapies.
Collapse
Affiliation(s)
- Xiangyu Kong
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Tao Sun
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Fanyang Kong
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yiqi Du
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
126
|
Folias AE, Penaranda C, Su AL, Bluestone JA, Hebrok M. Aberrant innate immune activation following tissue injury impairs pancreatic regeneration. PLoS One 2014; 9:e102125. [PMID: 25010227 PMCID: PMC4092101 DOI: 10.1371/journal.pone.0102125] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 06/15/2014] [Indexed: 01/03/2023] Open
Abstract
Normal tissue architecture is disrupted following injury, as resident tissue cells become damaged and immune cells are recruited to the site of injury. While injury and inflammation are critical to tissue remodeling, the inability to resolve this response can lead to the destructive complications of chronic inflammation. In the pancreas, acinar cells of the exocrine compartment respond to injury by transiently adopting characteristics of progenitor cells present during embryonic development. This process of de-differentiation creates a window where a mature and stable cell gains flexibility and is potentially permissive to changes in cellular fate. How de-differentiation can turn an acinar cell into another cell type (such as a pancreatic β-cell), or a cell with cancerous potential (as in cases of deregulated Kras activity) is of interest to both the regenerative medicine and cancer communities. While it is known that inflammation and acinar de-differentiation increase following pancreatic injury, it remains unclear which immune cells are involved in this process. We used a combination of genetically modified mice, immunological blockade and cellular characterization to identify the immune cells that impact pancreatic regeneration in an in vivo model of pancreatitis. We identified the innate inflammatory response of macrophages and neutrophils as regulators of pancreatic regeneration. Under normal conditions, mild innate inflammation prompts a transient de-differentiation of acinar cells that readily dissipates to allow normal regeneration. However, non-resolving inflammation developed when elevated pancreatic levels of neutrophils producing interferon-γ increased iNOS levels and the pro-inflammatory response of macrophages. Pancreatic injury improved following in vivo macrophage depletion, iNOS inhibition as well as suppression of iNOS levels in macrophages via interferon-γ blockade, supporting the impairment in regeneration and the development of chronic inflammation arises from aberrant activation of the innate inflammatory response. Collectively these studies identify targetable inflammatory factors that can be used to influence the development of non-resolving inflammation and pancreatic regeneration following injury.
Collapse
Affiliation(s)
- Alexandra E. Folias
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Cristina Penaranda
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Anthony L. Su
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey A. Bluestone
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
127
|
Keenan BP, Saenger Y, Kafrouni MI, Leubner A, Lauer P, Maitra A, Rucki AA, Gunderson AJ, Coussens LM, Brockstedt DG, Dubensky TW, Hassan R, Armstrong TD, Jaffee EM. A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice. Gastroenterology 2014; 146:1784-94.e6. [PMID: 24607504 PMCID: PMC4035450 DOI: 10.1053/j.gastro.2014.02.055] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/09/2014] [Accepted: 02/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Premalignant lesions and early stage tumors contain immunosuppressive microenvironments that create barriers for cancer vaccines. Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which express an activated form of Kras in pancreatic tissues, develop pancreatic intraepithelial neoplasms (PanIN) that progress to pancreatic ductal adenocarcinoma (PDA). We used these mice to study immune suppression in PDA. METHODS We immunized KPC and Kras(G12D/+);Pdx-1-Cre mice with attenuated intracellular Listeria monocytogenes (which induces CD4(+) and CD8(+) T-cell immunity) engineered to express Kras(G12D) (LM-Kras). The vaccine was given alone or in sequence with an anti-CD25 antibody (PC61) and cyclophosphamide to deplete T-regulatory (Treg) cells. Survival times were measured; pancreatic and spleen tissues were collected and analyzed by histologic, flow cytometry, and immunohistochemical analyses. RESULTS Interferon γ-mediated, CD8(+) T-cell responses were observed in KPC and Kras(G12D/+);Pdx-1-Cre mice given LM-Kras, but not in unvaccinated mice. Administration of LM-Kras to KPC mice 4-6 weeks old (with early stage PanINs), depleted of Treg cells, significantly prolonged survival and reduced PanIN progression (median survival, 265 days), compared with unvaccinated mice (median survival, 150 days; P = .002), mice given only LM-Kras (median survival, 150 days; P = .050), and unvaccinated mice depleted of Treg cells (median survival, 170 days; P = .048). In 8- to 12-week-old mice (with late-stage PanINs), LM-Kras, alone or in combination with Treg cell depletion, did not increase survival time or slow PanIN progression. The combination of LM-Kras and Treg cell depletion reduced numbers of Foxp3(+)CD4(+) T cells in pancreatic lymph nodes, increased numbers of CD4(+) T cells that secrete interleukin 17 and interferon γ, and caused CD11b(+)Gr1(+) cells in the pancreas to acquire an immunostimulatory phenotype. CONCLUSIONS Immunization of KPC mice with Listeria monocytogenes engineered to express Kras(G12D), along with depletion of Treg cells, reduces progression of early stage, but not late-stage, PanINs. This approach increases infiltration of the lesion with inflammatory cells. It might be possible to design immunotherapies against premalignant pancreatic lesions to slow or prevent progression to PDA.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- CD11b Antigen/metabolism
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Carcinoma in Situ/drug therapy
- Carcinoma in Situ/genetics
- Carcinoma in Situ/immunology
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cyclophosphamide/pharmacology
- Disease Models, Animal
- Disease Progression
- Forkhead Transcription Factors/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Inflammation Mediators/metabolism
- Integrases/genetics
- Integrases/metabolism
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/metabolism
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- Receptors, Chemokine/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Bridget P Keenan
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland; Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yvonne Saenger
- Division of Hematology and Oncology, Tisch Cancer Institute and Department of Dermatology, Mount Sinai School of Medicine, New York, New York
| | - Michel I Kafrouni
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Ashley Leubner
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | | | - Anirban Maitra
- Department of Pathology and Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Agnieszka A Rucki
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Andrew J Gunderson
- Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon
| | - Lisa M Coussens
- Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon
| | | | | | - Raffit Hassan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Todd D Armstrong
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Elizabeth M Jaffee
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland.
| |
Collapse
|
128
|
Matters GL, Cooper TK, McGovern CO, Gilius EL, Liao J, Barth BM, Kester M, Smith JP. Cholecystokinin mediates progression and metastasis of pancreatic cancer associated with dietary fat. Dig Dis Sci 2014; 59:1180-91. [PMID: 24817409 PMCID: PMC4096234 DOI: 10.1007/s10620-014-3201-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 04/30/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Obesity and dietary fat are associated with increased risk of several malignancies including pancreatic cancer. The incidence of pancreatic cancer is increased in countries that consume diets high in fat. AIM The purpose of this study was to assess the relationship and mechanism of action between dietary fat and endogenous cholecystokinin (CCK) on pancreatic tumor growth and metastasis in an immunocompetent animal model. METHODS C57BL/6 mice were placed on regular, low-fat, or high-fat diets for 8 weeks before establishment of Panc-02 orthotopic pancreatic tumors. Mice were then treated with a CCK-A receptor antagonist, devazepide, or vehicle for an additional 2.5 weeks. Pancreas tumors were weighed and metastases counted. Blood CCK levels were measured by radioimmunoassay (RIA). Tissues were examined histologically and studied for genes associated with metastasis by RT-PCR array. Effects of the CCK antagonist on Panc-02 cells invasiveness was assessed in a Matrigel invasion assay. RESULTS Mice that received the high-fat diet had larger tumors and tenfold higher serum CCK levels by RIA compared to normal diet controls (p < 0.01). Pancreatic tumors in high-fat diet mice treated with the antagonist had fewer intravascular tumor emboli and metastases compared to controls. The reduction in tumor emboli correlated with decreased vascular endothelial growth factor-A (VEGF-A) expression in tumors (p < 6 × 10(-9)). In vitro invasiveness of Panc-02 cells also was reduced by CCK-A receptor antagonist treatment (p = 1.33 × 10(-6)). CONCLUSION CCK is a mediator of dietary fat-associated pancreatic cancer. CCK is also involved in the invasiveness of pancreatic tumors through a mechanism involving VEGF-A.
Collapse
Affiliation(s)
- Gail L. Matters
- Department of Biochemistry and Molecular Biology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Timothy K. Cooper
- Department of Comparative Medicine, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA. Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Christopher O. McGovern
- Department of Medicine, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Evan L. Gilius
- Department of Medicine, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Jiangang Liao
- Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Brian M. Barth
- Department of Pharmacology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Mark Kester
- Department of Pharmacology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Jill P. Smith
- Department of Medicine, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA. Department of Medicine, Georgetown University Hospital, 3800 Reservoir Rd, NW, 2 Main, 2nd Floor, Washington, DC 20007, USA
| |
Collapse
|
129
|
Ali S, Ahmad A, Aboukameel A, Ahmed A, Bao B, Banerjee S, Philip PA, Sarkar FH. Deregulation of miR-146a expression in a mouse model of pancreatic cancer affecting EGFR signaling. Cancer Lett 2014; 351:134-42. [PMID: 24839931 DOI: 10.1016/j.canlet.2014.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/09/2014] [Accepted: 05/11/2014] [Indexed: 01/20/2023]
Abstract
Aberrant expression of microRNAs (miRNAs) plays important roles in the development and progression of pancreatic cancer (PC). Expression analysis of miR-146a in human PC tissues showed decreased expression in about 80% of samples compared to corresponding non-cancerous tissue. Moreover, expression of miR-146a in eight PC cell lines, and in pancreatic tissues obtained from transgenic mouse models of K-Ras (K), Pdx1-Cre (C), K-Ras;Pdx1-Cre (KC) and K-Ras;Pdx1-Cre;INK4a/Arf (KCI), showed down-regulation of miR-146a expression in KCI mice which was in part led to over-expression of its target gene, epidermal growth factor receptor (EGFR). Treatment of PC cells with CDF, a novel synthetic compound, led to re-expression of miR-146a, resulting in the down-regulation of EGFR expression. Moreover, re-expression of miR-146a by stable transfection or treatment with CDF in vivo (xenograft animal model) resulted in decreased tumor growth which was consistent with reduced EGFR, ERK1, ERK2, and K-Ras expression. Further knock-down of miR-146a in AsPC-1 cells led to the up-regulation of EGFR expression and showed increased clonogenic growth. In addition, knock-down of EGFR by EGFR siRNA transfection of parental AsPC-1 cells and AsPC-1 cells stably transfected with pre-miR-146a resulted in decreased invasive capacity, which was further confirmed by reduced luciferase activity in cells transfected with pMIR-Luc reporter vector containing miR-146a binding site. Collectively, these results suggest that the loss of expression of miR-146a is a fundamental mechanism for over-expression of EGFR signaling and that re-expression of miR-146a by CDF treatment could be useful in designing personalized strategy for the treatment of human PC.
Collapse
Affiliation(s)
- Shadan Ali
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Amro Aboukameel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Alia Ahmed
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fazlul H Sarkar
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States; Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
130
|
Abstract
Over the last decade, it has been discovered that the transcription factor Sox9 plays several critical roles in governing the development of the embryonic pancreas and the homeostasis of the mature organ. While analysis of pancreata from patients affected by the Sox9 haploinsufficiency syndrome campomelic dysplasia initially alluded to a functional role of Sox9 in pancreatic morphogenesis, transgenic mouse models have been instrumental in mechanistically dissecting such roles. Although initially defined as a marker and maintenance factor for pancreatic progenitors, Sox9 is now considered to fulfill additional indispensable functions during pancreogenesis and in the postnatal organ through its interactions with other transcription factors and signaling pathways such as Fgf and Notch. In addition to maintaining both multipotent and bipotent pancreatic progenitors, Sox9 is also required for initiating endocrine differentiation and maintaining pancreatic ductal identity, and it has recently been unveiled as a key player in the initiation of pancreatic cancer. These functions of Sox9 are discussed in this article, with special emphasis on the knowledge gained from various loss-of-function and lineage tracing mouse models. Also, current controversies regarding Sox9 function in healthy and injured adult pancreas and unanswered questions and avenues of future study are discussed.
Collapse
Affiliation(s)
- Philip A Seymour
- The Danish Stem Cell Center (DanStem), University of Copenhagen, Panum Institute, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| |
Collapse
|
131
|
Morris JP, Greer R, Russ HA, von Figura G, Kim GE, Busch A, Lee J, Hertel KJ, Kim S, Mcmanus M, Hebrok M. Dicer regulates differentiation and viability during mouse pancreatic cancer initiation. PLoS One 2014; 9:e95486. [PMID: 24788257 PMCID: PMC4006805 DOI: 10.1371/journal.pone.0095486] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/26/2014] [Indexed: 12/13/2022] Open
Abstract
miRNA levels are altered in pancreatic ductal adenocarcinoma (PDA), the most common and lethal pancreatic malignancy, and intact miRNA processing is essential for lineage specification during pancreatic development. However, the role of miRNA processing in PDA has not been explored. Here we study the role of miRNA biogenesis in PDA development by deleting the miRNA processing enzyme Dicer in a PDA mouse model driven by oncogenic Kras. We find that loss of Dicer accelerates Kras driven acinar dedifferentiation and acinar to ductal metaplasia (ADM), a process that has been shown to precede and promote the specification of PDA precursors. However, unconstrained ADM also displays high levels of apoptosis. Dicer loss does not accelerate development of Kras driven PDA precursors or PDA, but surprisingly, we observe that mouse PDA can develop without Dicer, although at the expense of proliferative capacity. Our data suggest that intact miRNA processing is involved in both constraining pro-tumorigenic changes in pancreatic differentiation as well as maintaining viability during PDA initiation.
Collapse
Affiliation(s)
- John P. Morris
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Renee Greer
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Holger A. Russ
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Guido von Figura
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Grace E. Kim
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Anke Busch
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Jonghyeob Lee
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Klemens J. Hertel
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Seung Kim
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael Mcmanus
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
132
|
Abstract
Pancreatic cancer is one of the most lethal cancers worldwide. No effective screening methods exist, and available treatment modalities do not effectively treat the disease. Inflammatory conditions such as pancreatitis represent a well-known risk factor for pancreatic cancer development. Yet only in the past 2 decades has pancreatic cancer been recognized as an inflammation-driven cancer, and the precise mechanisms underlying the pathogenic role of inflammation are beginning to be explored in detail. A substantial amount of preclinical and clinical evidence suggests that bacteria are likely to influence this process by activating immune receptors and perpetuating cancer-associated inflammation. The recent explosion of investigations of the human microbiome have highlighted how perturbations of commensal bacterial populations can promote inflammation and promote disease processes, including carcinogenesis. The elucidation of the interplay between inflammation and microbiome in the context of pancreatic carcinogenesis will provide novel targets for intervention to prevent and treat pancreatic cancer more efficiently. Further studies toward this direction are urgently needed.
Collapse
Affiliation(s)
- Constantinos P. Zambirinis
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
| | - Smruti Pushalkar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - George Miller
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
- S. Arthur Localio Laboratory, Departments of Cell Biology New York University School of Medicine, New York, NY 10016
| |
Collapse
|
133
|
Baumgart S, Chen NM, Siveke JT, König A, Zhang JS, Singh SK, Wolf E, Bartkuhn M, Esposito I, Heßmann E, Reinecke J, Nikorowitsch J, Brunner M, Singh G, Fernandez-Zapico ME, Smyrk T, Bamlet WR, Eilers M, Neesse A, Gress TM, Billadeau DD, Tuveson D, Urrutia R, Ellenrieder V. Inflammation-induced NFATc1-STAT3 transcription complex promotes pancreatic cancer initiation by KrasG12D. Cancer Discov 2014; 4:688-701. [PMID: 24694735 DOI: 10.1158/2159-8290.cd-13-0593] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Cancer-associated inflammation is a molecular key feature in pancreatic ductal adenocarcinoma. Oncogenic KRAS in conjunction with persistent inflammation is known to accelerate carcinogenesis, although the underlying mechanisms remain poorly understood. Here, we outline a novel pathway whereby the transcription factors NFATc1 and STAT3 cooperate in pancreatic epithelial cells to promote Kras(G12D)-driven carcinogenesis. NFATc1 activation is induced by inflammation and itself accelerates inflammation-induced carcinogenesis in Kras(G12D) mice, whereas genetic or pharmacologic ablation of NFATc1 attenuates this effect. Mechanistically, NFATc1 complexes with STAT3 for enhancer-promoter communications at jointly regulated genes involved in oncogenesis, for example, Cyclin, EGFR and WNT family members. The NFATc1-STAT3 cooperativity is operative in pancreatitis-mediated carcinogenesis as well as in established human pancreatic cancer. Together, these studies unravel new mechanisms of inflammatory-driven pancreatic carcinogenesis and suggest beneficial effects of chemopreventive strategies using drugs that are currently available for targeting these factors in clinical trials. SIGNIFICANCE Our study points to the existence of an oncogenic NFATc1-STAT3 cooperativity that mechanistically links inflammation with pancreatic cancer initiation and progression. Because NFATc1-STAT3 nucleoprotein complexes control the expression of gene networks at the intersection of inflammation and cancer, our study has significant relevance for potentially managing pancreatic cancer and other inflammatory-driven malignancies.
Collapse
Affiliation(s)
- Sandra Baumgart
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Nai-Ming Chen
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New YorkAuthors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Jens T Siveke
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Alexander König
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New YorkAuthors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New YorkAuthors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg
| | - Jin-San Zhang
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Shiv K Singh
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Elmar Wolf
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Marek Bartkuhn
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Irene Esposito
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Elisabeth Heßmann
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New YorkAuthors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Johanna Reinecke
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New YorkAuthors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Julius Nikorowitsch
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Marius Brunner
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Garima Singh
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Martin E Fernandez-Zapico
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Thomas Smyrk
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - William R Bamlet
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Martin Eilers
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Albrecht Neesse
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Thomas M Gress
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Daniel D Billadeau
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - David Tuveson
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Raul Urrutia
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Volker Ellenrieder
- Authors' Affiliations:Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg; Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität; Institute of Pathology, Helmholtz Zentrum, Munich; Theodor Boveri Institute, University of Würzburg, Würzburg; Institute for Genetics, Justus-Liebig-University, Giessen, Germany; Schulze Center for Novel Therapeutics, Division of Oncology Research; Divisions of Anatomic Pathology and Biostatistics, College of Medicine; Laboratory of Epigenetics and Chromatin Dynamics, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; Barrow Brain Tumor Research Center, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| |
Collapse
|
134
|
Flandez M, Cendrowski J, Cañamero M, Salas A, del Pozo N, Schoonjans K, Real FX. Nr5a2 heterozygosity sensitises to, and cooperates with, inflammation in KRas(G12V)-driven pancreatic tumourigenesis. Gut 2014; 63:647-55. [PMID: 23598351 DOI: 10.1136/gutjnl-2012-304381] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Nr5a2 participates in biliary acid metabolism and is a major regulator of the pancreatic exocrine programme. Single nucleotide polymorphisms in the vicinity of NR5A2 are associated with the risk of pancreatic ductal adenocarcinoma (PDAC). AIMS To determine the role of Nr5a2 in pancreatic homeostasis, damage-induced regeneration and mutant KRas-driven pancreatic tumourigenesis. DESIGN Nr5a2+/- and KRas(G12V);Ptf1a-Cre;Nr5a2+/- mice were used to investigate whether a full dose of Nr5a2 is required for normal pancreas development, recovery from caerulein-induced pancreatitis, and protection from tumour development. RESULTS Adult Nr5a2+/- mice did not display histological abnormalities in the pancreas but showed a more severe acute pancreatitis, increased acino-ductal metaplasia and impaired recovery from damage. This was accompanied by increased myeloid cell infiltration and proinflammatory cytokine gene expression, and hyperactivation of nuclear factor κb and signal transducer and activator of transcription 3 signalling pathways. Induction of multiple episodes of acute pancreatitis was associated with more severe damage and delayed regeneration. Inactivation of one Nr5a2 allele selectively in pancreatic epithelial cells was sufficient to cause impaired recovery from pancreatitis. In comparison with Nr5a2+/+ mice, KRas(G12V);Ptf1a(Cre/+);Nr5a2+/- mice showed a non-statistically significant increase in the area affected by preneoplastic lesions. However, a single episode of acute pancreatitis cooperated with loss of one Nr5a2 allele to accelerate KRas(G12V)-driven development of preneoplastic lesions. CONCLUSIONS A full Nr5a2 dose is required to restore pancreatic homeostasis upon damage and to suppress the KRas(G12V)-driven mouse pancreatic intraepithelial neoplasia progression, indicating that Nr5a2 is a novel pancreatic tumour suppressor. Nr5a2 could contribute to PDAC through a role in the recovery from pancreatitis-induced damage.
Collapse
Affiliation(s)
- Marta Flandez
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, CNIO-Spanish National Cancer Research Center, , Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
135
|
von Figura G, Morris JP, Wright CVE, Hebrok M. Nr5a2 maintains acinar cell differentiation and constrains oncogenic Kras-mediated pancreatic neoplastic initiation. Gut 2014; 63:656-64. [PMID: 23645620 PMCID: PMC3883808 DOI: 10.1136/gutjnl-2012-304287] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Emerging evidence from mouse models suggests that mutant Kras can drive the development of pancreatic ductal adenocarcinoma (PDA) precursors from acinar cells by enforcing ductal de-differentiation at the expense of acinar identity. Recently, human genome-wide association studies have identified NR5A2, a key regulator of acinar function, as a susceptibility locus for human PDA. We investigated the role of Nr5a2 in exocrine maintenance, regeneration and Kras driven neoplasia. DESIGN To investigate the function of Nr5a2 in the pancreas, we generated mice with conditional pancreatic Nr5a2 deletion (PdxCre(late); Nr5a2(c/c)). Using this model, we evaluated acinar differentiation, regeneration after caerulein pancreatitis and Kras driven pancreatic neoplasia in the setting of Nr5a2 deletion. RESULTS We show that Nr5a2 is not required for the development of the pancreatic acinar lineage but is important for maintenance of acinar identity. Nr5a2 deletion leads to destabilisation of the mature acinar differentiation state, acinar to ductal metaplasia and loss of regenerative capacity following acute caerulein pancreatitis. Loss of Nr5a2 also dramatically accelerates the development of oncogenic Kras driven acinar to ductal metaplasia and PDA precursor lesions. CONCLUSIONS Nr5a2 is a key regulator of acinar plasticity. It is required for maintenance of acinar identity and re-establishing acinar fate during regeneration. Nr5a2 also constrains pancreatic neoplasia driven by oncogenic Kras, providing functional evidence supporting a potential role as a susceptibility gene for human PDA.
Collapse
Affiliation(s)
- Guido von Figura
- Department of Medicine, Diabetes Center, University of California-San Francisco, San Francisco, California, USA
| | - John P Morris
- Department of Medicine, Diabetes Center, University of California-San Francisco, San Francisco, California, USA
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthias Hebrok
- Department of Medicine, Diabetes Center, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
136
|
Court H, Amoyel M, Hackman M, Lee KE, Xu R, Miller G, Bar-Sagi D, Bach EA, Bergö MO, Philips MR. Isoprenylcysteine carboxylmethyltransferase deficiency exacerbates KRAS-driven pancreatic neoplasia via Notch suppression. J Clin Invest 2014; 123:4681-94. [PMID: 24216479 DOI: 10.1172/jci65764] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 08/08/2013] [Indexed: 12/20/2022] Open
Abstract
RAS is the most frequently mutated oncogene in human cancers. Despite decades of effort, anti-RAS therapies have remained elusive. Isoprenylcysteine carboxylmethyltransferase (ICMT) methylates RAS and other CaaX-containing proteins, but its potential as a target for cancer therapy has not been fully evaluated. We crossed a Pdx1-Cre;LSL-KrasG12D mouse, which is a model of pancreatic ductal adenocarcinoma (PDA), with a mouse harboring a floxed allele of Icmt. Surprisingly, we found that ICMT deficiency dramatically accelerated the development and progression of neoplasia. ICMT-deficient pancreatic ductal epithelial cells had a slight growth advantage and were resistant to premature senescence by a mechanism that involved suppression of cyclin-dependent kinase inhibitor 2A (p16INK4A) expression. ICMT deficiency precisely phenocopied Notch1 deficiency in the Pdx1-Cre;LSL-KrasG12D model by exacerbating pancreatic intraepithelial neoplasias, promoting facial papillomas, and derepressing Wnt signaling. Silencing ICMT in human osteosarcoma cells decreased Notch1 signaling in response to stimulation with cell-surface ligands. Additionally, targeted silencing of Ste14, the Drosophila homolog of Icmt, resulted in defects in wing development, consistent with Notch loss of function. Our data suggest that ICMT behaves like a tumor suppressor in PDA because it is required for Notch1 signaling.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Carcinoma in Situ/genetics
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Disease Models, Animal
- Drosophila melanogaster/genetics
- Drosophila melanogaster/growth & development
- Drosophila melanogaster/metabolism
- Female
- Genes, ras
- Humans
- Male
- Metaplasia
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Mutation
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Methyltransferases/deficiency
- Protein Methyltransferases/genetics
- Receptor, Notch1/metabolism
- Signal Transduction
Collapse
|
137
|
Collins MA, Pasca di Magliano M. Kras as a key oncogene and therapeutic target in pancreatic cancer. Front Physiol 2014; 4:407. [PMID: 24478710 PMCID: PMC3896882 DOI: 10.3389/fphys.2013.00407] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 12/24/2013] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is one of the deadliest human malignancies and little progress has been achieved in its treatment over the past decades. Advances in our understanding of the biology of this disease provide new potential opportunities for treatment. Pancreatic cancer is preceded by precursor lesions, the most common of which are known as Pancreatic Intraepithelial Neoplasia (PanIN). PanIN lesions, which are the focus of this review, have a high incidence of Kras mutations, and Kras mutations are a hallmark of the late-stage disease. We now know from genetically engineered mouse models that oncogenic Kras is not only driving the formation of pancreatic cancer precursor lesions, but it is also required for their progression, and for the maintenance of invasive and metastatic disease. Thus, an enormous effort is being placed in generating Kras inhibitors for clinical use. Additionally, alternative approaches, including understanding the role of Kras effector pathways at different stages of the disease progression, are being devised to target Kras effector pathways therapeutically. In particular, efforts have focused on the MAPK pathway and the PI3K pathway, for which inhibitors are widely available. Finally, recent studies have highlighted the need for oncogenic Kras to establish feedback mechanisms that maintain its levels of activity; the latter might constitute alternative ways to target Kras in pancreatic cancer. Here, we will review recent basic research and discuss potential therapeutic applications.
Collapse
Affiliation(s)
- Meredith A Collins
- Program in Cellular and Molecular Biology, University of Michigan Ann Arbor, MI, USA
| | - Marina Pasca di Magliano
- Program in Cellular and Molecular Biology, University of Michigan Ann Arbor, MI, USA ; Department of Surgery, University of Michigan Ann Arbor, MI, USA ; Department of Cell and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
138
|
Stopczynski RE, Normolle DP, Hartman DJ, Ying H, DeBerry JJ, Bielefeldt K, Rhim AD, DePinho RA, Albers KM, Davis BM. Neuroplastic changes occur early in the development of pancreatic ductal adenocarcinoma. Cancer Res 2014; 74:1718-27. [PMID: 24448244 DOI: 10.1158/0008-5472.can-13-2050] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Perineural tumor invasion of intrapancreatic nerves, neurogenic inflammation, and tumor metastases along extrapancreatic nerves are key features of pancreatic malignancies. Animal studies show that chronic pancreatic inflammation produces hypertrophy and hypersensitivity of pancreatic afferents and that sensory fibers may themselves drive inflammation via neurogenic mechanisms. Although genetic mutations are required for cancer development, inflammation has been shown to be a precipitating event that can accelerate the transition of precancerous lesions to cancer. These observations led us to hypothesize that inflammation that accompanies early phases of pancreatic ductal adenocarcinoma (PDAC) would produce pathologic changes in pancreatic neurons and innervation. Using a lineage-labeled genetically engineered mouse model of PDAC, we found that pancreatic neurotrophic factor mRNA expression and sensory innervation increased dramatically when only pancreatic intraepithelial neoplasia were apparent. These changes correlated with pain-related decreases in exploratory behavior and increased expression of nociceptive genes in sensory ganglia. At later stages, cells of pancreatic origin could be found in the celiac and sensory ganglia along with metastases to the spinal cord. These results demonstrate that the nervous system participates in all stages of PDAC, including those that precede the appearance of cancer.
Collapse
Affiliation(s)
- Rachelle E Stopczynski
- Authors' Affiliations: Departments of Neurobiology, Biostatistics, Pathology, and Medicine, University of Pittsburgh School of Medicine; Departments of Genomic Medicine and Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Smith JP, Solomon TE. Cholecystokinin and pancreatic cancer: the chicken or the egg? Am J Physiol Gastrointest Liver Physiol 2014; 306:G91-G101. [PMID: 24177032 PMCID: PMC4073990 DOI: 10.1152/ajpgi.00301.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal peptide cholecystokinin (CCK) causes the release of pancreatic digestive enzymes and growth of the normal pancreas. Exogenous CCK administration has been used in animal models to study pancreatitis and also as a promoter of carcinogen-induced or Kras-driven pancreatic cancer. Defining CCK receptors in normal human pancreas has been problematic because of its retroperitoneal location, high concentrations of pancreatic proteases, and endogenous RNase. Most studies indicate that the predominant receptor in human pancreas is the CCK-B type, and CCK-A is the predominant form in rodent pancreas. In pancreatic cancer cells and tumors, the role of CCK is better established because receptors are often overexpressed by these cancer cells and stimulation of such receptors promotes growth. Furthermore, in established cancer, endogenous production of CCK and/or gastrin occurs and their actions stimulate the synthesis of more receptors plus growth by an autocrine mechanism. Initially it was thought that the mechanism by which CCK served to potentiate carcinogenesis was by interplay with inflammation in the pancreatic microenvironment. But with the recent findings of CCK receptors on early PanIN (pancreatic intraepithelial neoplasia) lesions and on stellate cells, the question has been raised that perhaps CCK actions are not the result of cancer but an early driving promoter of cancer. This review will summarize what is known regarding CCK, its receptors, and pancreatic cancer, and also what is unknown and requires further investigation to determine which comes first, the chicken or the egg, "CCK or the cancer."
Collapse
Affiliation(s)
- Jill P. Smith
- 1Clinical and Translational Research, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland; and
| | - Travis E. Solomon
- 2Department of Basic Medical Science, University of Missouri-Kansas City, Kansas City, Missouri
| |
Collapse
|
140
|
Greer RL, Staley BK, Liou A, Hebrok M. Numb regulates acinar cell dedifferentiation and survival during pancreatic damage and acinar-to-ductal metaplasia. Gastroenterology 2013; 145:1088-1097.e8. [PMID: 23891977 PMCID: PMC3805717 DOI: 10.1053/j.gastro.2013.07.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDA) is a leading cause of cancer-related death. Through the process of acinar-to-ductal metaplasia (ADM), pancreatic acinar cells give rise to pancreatic intraepithelial neoplasia (PanIN), the most common precursor of PDA. However, even when Kras is activated in a majority of acinar cells, ADM and subsequent development of PanINs is inefficient in the absence of additional stresses. Numb regulates cell junctions, integrins, and the activity of embryonic signaling pathways; therefore, we investigated its effects on acinar cell dedifferentiation, regeneration, and metaplasia. METHODS We used mouse models of pancreatic regeneration and PDA as well as mice with loss-of-function alleles of Numb (p48Cre/p48Cre(ER);Numb(f/f) and p48Cre/p48Cre(ER);Kras(G12D);Numb(f/f) mice) to study the roles of Numb in pancreatic regeneration and ADM. RESULTS Loss of Numb resulted in premature dedifferentiation of acinar cells in response to injury due to administration of the cholecystokinin analogue cerulein and interfered with acinar cell regeneration. Numb was found to regulate multiple signaling pathways in acinar cells during cerulein-induced pancreatitis. Disruption of Numb accelerated and destabilized ADM in the context of oncogenic Kras (in p48Cre;Kras(G12D);Numb(f/f) and p48Cre(ER);Kras(G12D);Numb(f/f) mice). CONCLUSIONS Numb is an important regulator of acinar cell differentiation and viability during metaplasia. In mice with pancreatitis or pancreatic injury, elimination of Numb causes dedifferentiated acinar cells to undergo apoptosis, and this is not mitigated by oncogenic Kras.
Collapse
Affiliation(s)
- Renee L Greer
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | | | | |
Collapse
|
141
|
Abstract
The past several decades have seen great effort devoted to mimicking the key features of pancreatic ductal adenocarcinoma (PDAC) in animals and have produced 2 robust models of this deadly cancer. Carcinogen-treated Syrian hamsters develop PDAC with genetic lesions, which reproduce those of human, including activation of the Kras oncogene, and early studies in this species validated nongenetic risk factors for PDAC including pancreatitis, obesity, and diabetes. More recently, PDAC research has been invigorated by the development of genetically engineered mouse models based on tissue-specific Kras activation and deletion of tumor suppressor genes. Surprisingly, mouse PDAC appears to arise from exocrine acinar rather than ductal cells, via a process of phenotypic reprogramming that is accelerated by inflammation. Studies in both models have uncovered molecular mechanisms by which inflammation promotes and sustains PDAC and identified targets for chemoprevention to suppress PDAC in high-risk individuals. The mouse model, in particular, has also been instrumental in developing new approaches to early detection as well as treatment of advanced disease. Together, animal models enable diverse approaches to basic and preclinical research on pancreatic cancer, the results of which will accelerate progress against this currently intractable cancer.
Collapse
Affiliation(s)
- L Charles Murtaugh
- 1Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
142
|
Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, Pandol SJ, Lugea A, Gukovskaya AS, Li G, Hines OJ, Rozengurt E, Eibl G. High-fat, high-calorie diet promotes early pancreatic neoplasia in the conditional KrasG12D mouse model. Cancer Prev Res (Phila) 2013; 6:1064-73. [PMID: 23943783 DOI: 10.1158/1940-6207.capr-13-0065] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is epidemiologic evidence that obesity increases the risk of cancers. Several underlying mechanisms, including inflammation and insulin resistance, are proposed. However, the driving mechanisms in pancreatic cancer are poorly understood. The goal of the present study was to develop a model of diet-induced obesity and pancreatic cancer development in a state-of-the-art mouse model, which resembles important clinical features of human obesity, for example, weight gain and metabolic disturbances. Offspring of Pdx-1-Cre and LSL-KrasG12D mice were allocated to either a high-fat, high-calorie diet (HFCD; ∼4,535 kcal/kg; 40% of calories from fats) or control diet (∼3,725 kcal/kg; 12% of calories from fats) for 3 months. Compared with control animals, mice fed with the HFCD significantly gained more weight and developed hyperinsulinemia, hyperglycemia, hyperleptinemia, and elevated levels of insulin-like growth factor I (IGF-I). The pancreas of HFCD-fed animals showed robust signs of inflammation with increased numbers of infiltrating inflammatory cells (macrophages and T cells), elevated levels of several cytokines and chemokines, increased stromal fibrosis, and more advanced PanIN lesions. Our results show that a diet high in fats and calories leads to obesity and metabolic disturbances similar to humans and accelerates early pancreatic neoplasia in the conditional KrasG12D mouse model. This model and findings will provide the basis for more robust studies attempting to unravel the mechanisms underlying the cancer-promoting properties of obesity, as well as to evaluate dietary- and chemopreventive strategies targeting obesity-associated pancreatic cancer development.
Collapse
Affiliation(s)
- David W Dawson
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, 10833 LeConte Avenue, 72-236 CHS, Los Angeles, CA 90095.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology 2013; 144:1220-9. [PMID: 23622131 PMCID: PMC3902845 DOI: 10.1053/j.gastro.2013.01.071] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 12/16/2022]
Abstract
The Kras gene is mutated to an oncogenic form in most pancreatic tumors. However, early attempts to use this molecule as a specific biomarker of the disease, or inhibit its activity as a cancer therapy, failed. This left a situation in which everyone was aware of the association between this important oncogene and pancreatic cancer, but no one knew what to do about it. Recent findings have changed this picture-many assumptions made about KRAS and its role in pancreatic cancer were found to be incorrect. Several factors have contributed to increased understanding of the activities of KRAS, including creation of genetically engineered mouse models, which have allowed for detailed analyses of pancreatic carcinogenesis in an intact animal with a competent immune system. Cancer genome sequencing projects have increased our understanding of the heterogeneity of individual tumors. We also have a better understanding of which oncogenes are important for tumor maintenance and are therefore called "drivers." We review the advances and limitations of our knowledge about the role of Kras in development of pancreatic cancers and the important areas for future research.
Collapse
Affiliation(s)
| | - Craig D. Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
144
|
Heparin-binding epidermal growth factor-like growth factor eliminates constraints on activated Kras to promote rapid onset of pancreatic neoplasia. Oncogene 2013; 33:823-31. [PMID: 23376846 PMCID: PMC3929321 DOI: 10.1038/onc.2013.3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer remains as one of the most deadly cancers with few treatment options at late stages and little information about how it develops through earlier stages. Activating mutation of the Kras gene has been implicated in, but is not sufficient for, tumorigenesis. In mouse models of pancreatic cancer, loss of tumor suppressor genes in conjunction with Kras mutation leads to gradual stochastic acquisition of neoplastic precursors and carcinomas, whereas many cells remain phenotypically unaltered in younger mice. Here, we demonstrate that two oncogenic events, mutation of Kras and production of the growth factor heparin-binding epidermal growth factor-like growth factor (HB-EGF), are sufficient for rapid and complete neoplastic transformation of the exocrine pancreas. We found that macrophages are the major source of HB-EGF production in pancreatic cancer tissue samples, and that macrophages are present in high density and in close association with human pancreatic cancer lesions. In a mouse model, high macrophage density was observed at the earliest stages of neoplastic transformation. The consequence of elevated HB-EGF signaling was investigated without the confounding effects of other macrophage-produced factors via transgenic overexpression of the active form of HB-EGF. In this model, HB-EGF was sufficient to promote Kras-initiated tumorigenesis, inducing rapid and complete neoplastic transformation of the entire exocrine pancreas shortly after birth. HB-EGF overexpression and Kras(G12D) together, but neither alone, increased proliferation with increased cyclinD1 and decreased Cdkn2a/2d (p16/p19(Ink4A/Arf)). These findings establish the importance of oncogenic synergy in cancer initiation and promotion, and establish a molecular link between inflammation and the earliest stages of tumor induction.
Collapse
|
145
|
Huang H, Daniluk J, Liu Y, Chu J, Li Z, Ji B, Logsdon CD. Oncogenic K-Ras requires activation for enhanced activity. Oncogene 2013; 33:532-5. [PMID: 23334325 DOI: 10.1038/onc.2012.619] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/05/2012] [Accepted: 11/16/2012] [Indexed: 01/02/2023]
Abstract
Oncogenic Ras mutations are widely considered to be locked in a permanent 'On' state and 'constitutively active'. Yet, many healthy people have cells possessing mutant Ras without apparent harm, and in animal models mutant Ras causes transformation only after upregulation of Ras activity. Here, we demonstrate that oncogenic K-Ras is not constitutively active but can be readily activated by upstream stimulants to lead to prolonged strong Ras activity. These data indicate that in addition to targeting K-Ras downstream effectors, interventions to reduce K-Ras activation may have important cancer-preventive value, especially in patients with oncogenic Ras mutations. As other small G proteins are regulated in a similar manner, this concept is likely to apply broadly to the entire Ras family of molecules.
Collapse
Affiliation(s)
- H Huang
- 1] Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - J Daniluk
- 1] Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Y Liu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Chu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Z Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - B Ji
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - C D Logsdon
- 1] Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
146
|
Kopp JL, von Figura G, Mayes E, Liu FF, Dubois CL, Morris JP, Pan FC, Akiyama H, Wright CVE, Jensen K, Hebrok M, Sander M. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 2012; 22. [PMID: 23201164 PMCID: PMC3568632 DOI: 10.1016/j.ccr.2012.10.025] [Citation(s) in RCA: 529] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tumors are largely classified by histologic appearance, yet morphologic features do not necessarily predict cellular origin. To determine the origin of pancreatic ductal adenocarcinoma (PDA), we labeled and traced pancreatic cell populations after induction of a PDA-initiating Kras mutation. Our studies reveal that ductal and stem-like centroacinar cells are surprisingly refractory to oncogenic transformation, whereas acinar cells readily form PDA precursor lesions with ductal features. We show that formation of acinar-derived premalignant lesions depends on ectopic induction of the ductal gene Sox9. Moreover, when concomitantly expressed with oncogenic Kras, Sox9 accelerates formation of premalignant lesions. These results provide insight into the cellular origin of PDA and suggest that its precursors arise via induction of a duct-like state in acinar cells.
Collapse
Affiliation(s)
- Janel L. Kopp
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California-San Diego, La Jolla, CA 92093-0695
| | - Guido von Figura
- Diabetes Center, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143
| | - Erin Mayes
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California-San Diego, La Jolla, CA 92093-0695
| | - Fen-Fen Liu
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California-San Diego, La Jolla, CA 92093-0695
| | - Claire L. Dubois
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California-San Diego, La Jolla, CA 92093-0695
| | - John P. Morris
- Diabetes Center, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143
| | - Fong Cheng Pan
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240
| | - Haruhiko Akiyama
- Department of Orthopedics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo, Kyoto 606-8507, Japan
| | | | - Kristin Jensen
- Department of Pathology, Veterans Affairs Palo Alto Health Care System and Stanford University Hospital, Stanford, CA 94305
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143
- Corresponding authors: , Telephone: (858) 246-0843, Fax: (858) 246-1579. , Telephone: (415) 514-0820, Fax: (415) 564-5813
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California-San Diego, La Jolla, CA 92093-0695
- Corresponding authors: , Telephone: (858) 246-0843, Fax: (858) 246-1579. , Telephone: (415) 514-0820, Fax: (415) 564-5813
| |
Collapse
|
147
|
Ochi A, Graffeo CS, Zambirinis CP, Rehman A, Hackman M, Fallon N, Barilla RM, Henning JR, Jamal M, Rao R, Greco S, Deutsch M, Medina-Zea MV, Saeed UB, Ego-Osuala MO, Hajdu C, Miller G. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J Clin Invest 2012; 122:4118-29. [PMID: 23023703 PMCID: PMC3484447 DOI: 10.1172/jci63606] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 08/02/2012] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is an aggressive cancer that interacts with stromal cells to produce a highly inflammatory tumor microenvironment that promotes tumor growth and invasiveness. The precise interplay between tumor and stroma remains poorly understood. TLRs mediate interactions between environmental stimuli and innate immunity and trigger proinflammatory signaling cascades. Our finding that TLR7 expression is upregulated in both epithelial and stromal compartments in human and murine pancreatic cancer led us to postulate that carcinogenesis is dependent on TLR7 signaling. In a mouse model of pancreatic cancer, TLR7 ligation vigorously accelerated tumor progression and induced loss of expression of PTEN, p16, and cyclin D1 and upregulation of p21, p27, p53, c-Myc, SHPTP1, TGF-β, PPARγ, and cyclin B1. Furthermore, TLR7 ligation induced STAT3 activation and interfaced with Notch as well as canonical NF-κB and MAP kinase pathways, but downregulated expression of Notch target genes. Moreover, blockade of TLR7 protected against carcinogenesis. Since pancreatic tumorigenesis requires stromal expansion, we proposed that TLR7 ligation modulates pancreatic cancer by driving stromal inflammation. Accordingly, we found that mice lacking TLR7 exclusively within their inflammatory cells were protected from neoplasia. These data suggest that targeting TLR7 holds promise for treatment of human pancreatic cancer.
Collapse
MESH Headings
- Animals
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunity, Innate/genetics
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- MAP Kinase Signaling System/genetics
- MAP Kinase Signaling System/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Mutant Strains
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/metabolism
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Toll-Like Receptor 7/genetics
- Toll-Like Receptor 7/immunology
- Toll-Like Receptor 7/metabolism
Collapse
Affiliation(s)
- Atsuo Ochi
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Christopher S. Graffeo
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Constantinos P. Zambirinis
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Adeel Rehman
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Michael Hackman
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Nina Fallon
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Rocky M. Barilla
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Justin R. Henning
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Mohsin Jamal
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Raghavendra Rao
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Stephanie Greco
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Michael Deutsch
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Marco V. Medina-Zea
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Usama Bin Saeed
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Melvin O. Ego-Osuala
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - Cristina Hajdu
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| | - George Miller
- Department of Surgery,
Department of Cell Biology, and
Department of Pathology, S. Arthur Localio Laboratory, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
148
|
Ali S, Banerjee S, Logna F, Bao B, Philip PA, Korc M, Sarkar FH. Inactivation of Ink4a/Arf leads to deregulated expression of miRNAs in K-Ras transgenic mouse model of pancreatic cancer. J Cell Physiol 2012; 227:3373-80. [PMID: 22213426 DOI: 10.1002/jcp.24036] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human pancreatic cancer (PC) is an aggressive disease, which has been recapitulated in transgenic animal model that provides unique opportunity for mechanistic understanding of disease progression and also for testing the efficacy of novel therapeutics. Emerging evidence suggests deregulated expression of microRNAs (miRNAs) in human PC, and thus we investigated the expression of miRNAs in pancreas tissues obtained from transgenic mouse models of K-Ras (K), Pdx1-Cre (C), K-Ras;Pdx1-Cre (KC), and K-Ras;Pdx1-Cre;INK4a/Arf (KCI), initially from pooled RNA samples using miRNA profiling, and further confirmed in individual specimens by quantitative RT-PCR. We found over-expression of miR-21, miR-221, miR-27a, miR-27b, and miR-155, and down-regulation of miR-216a, miR-216b, miR-217, and miR-146a expression in tumors derived from KC and KCI mouse model, which was consistent with data from KCI-derived RInk-1 cells. Mechanistic investigations revealed a significant induction of EGFR, K-Ras, and MT1-MMP protein expression in tissues from both KC and KCI mouse compared to tissues from K or C, and these results were consistent with similar findings in RInk-1 cells compared to human MIAPaCa-2 cells. Furthermore, miR-155 knock-down in RInk-1 cells resulted in the inhibition of cell growth and colony formation consistent with down-regulation of EGFR, MT1-MMP, and K-Ras expression. In addition, miR-216b which target Ras, and forced re-expression of miR-216b in RInk-1 cells showed inhibition of cell proliferation and colony formation, which was correlated with reduced expression of Ras, EGFR, and MT1-MMP. These findings suggest that these models would be useful for preclinical evaluation of novel miRNA-targeted agents for designing personalized therapy for PC.
Collapse
Affiliation(s)
- Shadan Ali
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | | | | | | | | |
Collapse
|
149
|
Ueda J, Tanaka M, Ohtsuka T, Tokunaga S, Shimosegawa T. Surgery for chronic pancreatitis decreases the risk for pancreatic cancer: a multicenter retrospective analysis. Surgery 2012; 153:357-64. [PMID: 22989892 DOI: 10.1016/j.surg.2012.08.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 08/03/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic pancreatitis is suggested to be one of the risk factors for the development of pancreatic cancer. The aim of this study was to confirm the high incidence of pancreatic cancer in patients with chronic pancreatitis in Japan and to determine the factors associated with the risk for pancreatic cancer in patients with chronic pancreatitis. METHODS The working group of the Research Committee of Intractable Disease supported by the Ministry of Health, Labour and Welfare of Japan carried out a nationwide survey to investigate the relationship between chronic pancreatitis and pancreatic cancer. This retrospective study included patients diagnosed with chronic pancreatitis who had had at least 2 years of follow-up. They were contacted through 22 Japanese referral centers experienced in the management of chronic pancreatitis. RESULTS The standardized incidence ratio (95 CI) of pancreatic cancer was 11.8 (7.1-18.4). The incidence of pancreatic cancer was significantly lower in patients who had received surgery for chronic pancreatitis than in those who had not undergone surgery (hazard ratio estimated by Cox regression 0.11; 95% CI, 0.0014-0.80; P = .03). Patients who continued to drink alcohol after diagnosis of chronic pancreatitis showed a significantly higher incidence of pancreatic cancer than those who stopped drinking after diagnosis of chronic pancreatitis (hazard ratio, 5.07; 95% CI, 1.13-22.73; P = .03). CONCLUSION This study confirmed that chronic pancreatitis is an important risk factor for the development of pancreatic cancer in Japan. Patients who underwent surgery for the treatment of chronic pancreatitis had significantly lower incidences of pancreatic cancer. Surgery for chronic pancreatitis may inhibit the development of pancreatic cancer in patients with chronic pancreatitis.
Collapse
Affiliation(s)
- Junji Ueda
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
150
|
Ardito CM, Grüner BM, Takeuchi KK, Lubeseder-Martellato C, Teichmann N, Mazur PK, DelGiorno KE, Carpenter ES, Halbrook CJ, Hall JC, Pal D, Briel T, Herner A, Trajkovic-Arsic M, Sipos B, Liou GY, Storz P, Murray NR, Threadgill DW, Sibilia M, Washington MK, Wilson CL, Schmid RM, Raines EW, Crawford HC, Siveke JT. EGF receptor is required for KRAS-induced pancreatic tumorigenesis. Cancer Cell 2012; 22:304-17. [PMID: 22975374 PMCID: PMC3443395 DOI: 10.1016/j.ccr.2012.07.024] [Citation(s) in RCA: 418] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/16/2012] [Accepted: 07/31/2012] [Indexed: 12/11/2022]
Abstract
Initiation of pancreatic ductal adenocarcinoma (PDA) is definitively linked to activating mutations in the KRAS oncogene. However, PDA mouse models show that mutant Kras expression early in development gives rise to a normal pancreas, with tumors forming only after a long latency or pancreatitis induction. Here, we show that oncogenic KRAS upregulates endogenous EGFR expression and activation, the latter being dependent on the EGFR ligand sheddase, ADAM17. Genetic ablation or pharmacological inhibition of EGFR or ADAM17 effectively eliminates KRAS-driven tumorigenesis in vivo. Without EGFR activity, active RAS levels are not sufficient to induce robust MEK/ERK activity, a requirement for epithelial transformation.
Collapse
Affiliation(s)
- Christine M. Ardito
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
| | - Barbara M. Grüner
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | | | - Clara Lubeseder-Martellato
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Nicole Teichmann
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Pawel K. Mazur
- Department of Genetics, Department of Pediatrics, Stanford University, Stanford, CA 94305
| | - Kathleen E. DelGiorno
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | - Eileen S. Carpenter
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
| | - Christopher J. Halbrook
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | - Jason C. Hall
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | - Debjani Pal
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
| | - Thomas Briel
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Alexander Herner
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Marija Trajkovic-Arsic
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Bence Sipos
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | - Nicole R. Murray
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
| | | | - Maria Sibilia
- Institute for Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - M. Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Carole L. Wilson
- Department of Pathology, University of Washington, Seattle WA, 98195
| | - Roland M. Schmid
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Elaine W. Raines
- Department of Pathology, University of Washington, Seattle WA, 98195
| | - Howard C. Crawford
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794
- Department of Cancer Biology, Mayo Clinic, Florida, Jacksonville, FL 32224
- Department of Research, Veterans Affairs Medical Center, Northport, NY 11768
- Correspondence: (HCC); (JTS), listed alphabetically
| | - Jens T. Siveke
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
- Correspondence: (HCC); (JTS), listed alphabetically
| |
Collapse
|