Your browser doesn't support javascript.
loading
Oncogenic KRAS Reduces Expression of FGF21 in Acinar Cells to Promote Pancreatic Tumorigenesis in Mice on a High-Fat Diet.
Luo, Yongde; Yang, Yaying; Liu, Muyun; Wang, Dan; Wang, Feng; Bi, Yawei; Ji, Juntao; Li, Suyun; Liu, Yan; Chen, Rong; Huang, Haojie; Wang, Xiaojie; Swidnicka-Siergiejko, Agnieszka K; Janowitz, Tobias; Beyaz, Semir; Wang, Guoqiang; Xu, Sulan; Bialkowska, Agnieszka B; Luo, Catherine K; Pin, Christoph L; Liang, Guang; Lu, Xiongbin; Wu, Maoxin; Shroyer, Kenneth R; Wolff, Robert A; Plunkett, William; Ji, Baoan; Li, Zhaoshen; Li, Ellen; Li, Xiaokun; Yang, Vincent W; Logsdon, Craig D; Abbruzzese, James L; Lu, Weiqin.
Affiliation
  • Luo Y; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Medicine, Stony Brook University, Stony Brook, New York. Electronic address: yongdeluo08@gmail.com.
  • Yang Y; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Liu M; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Wang D; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Wang F; Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
  • Bi Y; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Ji J; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Li S; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Liu Y; Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Chen R; Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Huang H; Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Wang X; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Swidnicka-Siergiejko AK; Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Janowitz T; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
  • Beyaz S; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
  • Wang G; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Xu S; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Bialkowska AB; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Luo CK; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Pin CL; Departments of Pediatrics, Oncology, and Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario Children's Health Research Institute, London, Ontario, Canada.
  • Liang G; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Lu X; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.
  • Wu M; Department of Pathology, Stony Brook University, Stony Brook, New York.
  • Shroyer KR; Department of Pathology, Stony Brook University, Stony Brook, New York.
  • Wolff RA; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Plunkett W; Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Ji B; Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida.
  • Li Z; Department of Gastroenterology, Changhai Hospital, Shanghai, China.
  • Li E; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Li X; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Yang VW; Department of Medicine, Stony Brook University, Stony Brook, New York.
  • Logsdon CD; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas; Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
  • Abbruzzese JL; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas; Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina.
  • Lu W; Department of Medicine, Stony Brook University, Stony Brook, New York; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas. Electronic address: weiqin.lu@stonybrookmedicine.edu.
Gastroenterology ; 157(5): 1413-1428.e11, 2019 11.
Article in En | MEDLINE | ID: mdl-31352001
ABSTRACT
BACKGROUND &

AIMS:

Obesity is a risk factor for pancreatic cancer. In mice, a high-fat diet (HFD) and expression of oncogenic KRAS lead to development of invasive pancreatic ductal adenocarcinoma (PDAC) by unknown mechanisms. We investigated how oncogenic KRAS regulates the expression of fibroblast growth factor 21, FGF21, a metabolic regulator that prevents obesity, and the effects of recombinant human FGF21 (rhFGF21) on pancreatic tumorigenesis.

METHODS:

We performed immunohistochemical analyses of FGF21 levels in human pancreatic tissue arrays, comprising 59 PDAC specimens and 45 nontumor tissues. We also studied mice with tamoxifen-inducible expression of oncogenic KRAS in acinar cells (KrasG12D/+ mice) and fElasCreERT mice (controls). KrasG12D/+ mice were placed on an HFD or regular chow diet (control) and given injections of rhFGF21 or vehicle; pancreata were collected and analyzed by histology, immunoblots, quantitative polymerase chain reaction, and immunohistochemistry. We measured markers of inflammation in the pancreas, liver, and adipose tissue. Activity of RAS was measured based on the amount of bound guanosine triphosphate.

RESULTS:

Pancreatic tissues of mice expressed high levels of FGF21 compared with liver tissues. FGF21 and its receptor proteins were expressed by acinar cells. Acinar cells that expressed KrasG12D/+ had significantly lower expression of Fgf21 messenger RNA compared with acinar cells from control mice, partly due to down-regulation of PPARG expression-a transcription factor that activates Fgf21 transcription. Pancreata from KrasG12D/+ mice on a control diet and given injections of rhFGF21 had reduced pancreatic inflammation, infiltration by immune cells, and acinar-to-ductal metaplasia compared with mice given injections of vehicle. HFD-fed KrasG12D/+ mice given injections of vehicle accumulated abdominal fat, developed extensive inflammation, pancreatic cysts, and high-grade pancreatic intraepithelial neoplasias (PanINs); half the mice developed PDAC with liver metastases. HFD-fed KrasG12D/+ mice given injections of rhFGF21 had reduced accumulation of abdominal fat and pancreatic triglycerides, fewer pancreatic cysts, reduced systemic and pancreatic markers of inflammation, fewer PanINs, and longer survival-only approximately 12% of the mice developed PDACs, and none of the mice had metastases. Pancreata from HFD-fed KrasG12D/+ mice given injections of rhFGF21 had lower levels of active RAS than from mice given vehicle.

CONCLUSIONS:

Normal acinar cells from mice and humans express high levels of FGF21. In mice, acinar expression of oncogenic KRAS significantly reduces FGF21 expression. When these mice are placed on an HFD, they develop extensive inflammation, pancreatic cysts, PanINs, and PDACs, which are reduced by injection of FGF21. FGF21 also reduces the guanosine triphosphate binding capacity of RAS. FGF21 might be used in the prevention or treatment of pancreatic cancer.
Subject(s)
Acinar Cells/metabolism; Carcinoma, Pancreatic Ductal/metabolism; Cell Transformation, Neoplastic/metabolism; Diet, High-Fat; Fibroblast Growth Factors/metabolism; Pancreatic Intraductal Neoplasms/metabolism; Pancreatic Neoplasms/metabolism; Proto-Oncogene Proteins p21(ras)/metabolism; Acinar Cells/pathology; Animals; Carcinoma, Pancreatic Ductal/genetics; Carcinoma, Pancreatic Ductal/pathology; Carcinoma, Pancreatic Ductal/prevention & control; Cell Transformation, Neoplastic/genetics; Cell Transformation, Neoplastic/pathology; Down-Regulation; Fibroblast Growth Factors/genetics; Gene Expression Regulation, Neoplastic; Humans; Klotho Proteins; Membrane Proteins/genetics; Membrane Proteins/metabolism; Mice, Transgenic; Mutation; PPAR gamma/genetics; PPAR gamma/metabolism; Pancreatic Cyst/genetics; Pancreatic Cyst/metabolism; Pancreatic Cyst/pathology; Pancreatic Intraductal Neoplasms/genetics; Pancreatic Intraductal Neoplasms/pathology; Pancreatic Intraductal Neoplasms/prevention & control; Pancreatic Neoplasms/genetics; Pancreatic Neoplasms/pathology; Pancreatic Neoplasms/prevention & control; Pancreatitis/genetics; Pancreatitis/metabolism; Pancreatitis/pathology; Proto-Oncogene Proteins p21(ras)/genetics; Receptor, Fibroblast Growth Factor, Type 1/genetics; Receptor, Fibroblast Growth Factor, Type 1/metabolism; Signal Transduction; Transcription Factors/genetics; Transcription Factors/metabolism; Tumor Suppressor Protein p53/genetics; Tumor Suppressor Protein p53/metabolism
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Cell Transformation, Neoplastic / Proto-Oncogene Proteins p21(ras) / Carcinoma, Pancreatic Ductal / Acinar Cells / Diet, High-Fat / Fibroblast Growth Factors / Pancreatic Intraductal Neoplasms Type of study: Risk_factors_studies Language: En Journal: Gastroenterology Year: 2019 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Cell Transformation, Neoplastic / Proto-Oncogene Proteins p21(ras) / Carcinoma, Pancreatic Ductal / Acinar Cells / Diet, High-Fat / Fibroblast Growth Factors / Pancreatic Intraductal Neoplasms Type of study: Risk_factors_studies Language: En Journal: Gastroenterology Year: 2019 Document type: Article