Mitochondrial Dysfunction, Through Impaired Autophagy, Leads to Endoplasmic Reticulum Stress, Deregulated Lipid Metabolism, and Pancreatitis in Animal Models

Gastroenterology. 2018 Feb;154(3):689-703. doi: 10.1053/j.gastro.2017.10.012. Epub 2017 Oct 23.

Abstract

Background & aims: Little is known about the signaling pathways that initiate and promote acute pancreatitis (AP). The pathogenesis of AP has been associated with abnormal increases in cytosolic Ca2+, mitochondrial dysfunction, impaired autophagy, and endoplasmic reticulum (ER) stress. We analyzed the mechanisms of these dysfunctions and their relationships, and how these contribute to development of AP in mice and rats.

Methods: Pancreatitis was induced in C57BL/6J mice (control) and mice deficient in peptidylprolyl isomerase D (cyclophilin D, encoded by Ppid) by administration of L-arginine (also in rats), caerulein, bile acid, or an AP-inducing diet. Parameters of pancreatitis, mitochondrial function, autophagy, ER stress, and lipid metabolism were measured in pancreatic tissue, acinar cells, and isolated mitochondria. Some mice with AP were given trehalose to enhance autophagic efficiency. Human pancreatitis tissues were analyzed by immunofluorescence.

Results: Mitochondrial dysfunction in pancreas of mice with AP was induced by either mitochondrial Ca2+ overload or through a Ca2+ overload-independent pathway that involved reduced activity of ATP synthase (80% inhibition in pancreatic mitochondria isolated from rats or mice given L-arginine). Both pathways were mediated by cyclophilin D and led to mitochondrial depolarization and fragmentation. Mitochondrial dysfunction caused pancreatic ER stress, impaired autophagy, and deregulation of lipid metabolism. These pathologic responses were abrogated in cyclophilin D-knockout mice. Administration of trehalose largely prevented trypsinogen activation, necrosis, and other parameters of pancreatic injury in mice with L-arginine AP. Tissues from patients with pancreatitis had markers of mitochondrial damage and impaired autophagy, compared with normal pancreas.

Conclusions: In different animal models, we find a central role for mitochondrial dysfunction, and for impaired autophagy as its principal downstream effector, in development of AP. In particular, the pathway involving enhanced interaction of cyclophilin D with ATP synthase mediates L-arginine-induced pancreatitis, a model of severe AP the pathogenesis of which has remained unknown. Strategies to restore mitochondrial and/or autophagic function might be developed for treatment of AP.

Keywords: Acinar Cell; Inflammatory Response; Lamellar Bodies; Pancreas.

Publication types

  • Comparative Study
  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Acute Disease
  • Animals
  • Arginine
  • Autophagy* / drug effects
  • Bile Acids and Salts
  • Calcium Signaling
  • Ceruletide
  • Choline Deficiency / complications
  • Cyclophilins / deficiency
  • Cyclophilins / genetics
  • Disease Models, Animal
  • Endoplasmic Reticulum Stress* / drug effects
  • Ethionine
  • Genetic Predisposition to Disease
  • Humans
  • Lipid Metabolism* / drug effects
  • Membrane Potential, Mitochondrial
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Mitochondria / drug effects
  • Mitochondria / metabolism*
  • Mitochondria / pathology
  • Mitochondrial Proton-Translocating ATPases / metabolism
  • Pancreas / drug effects
  • Pancreas / metabolism*
  • Pancreas / pathology
  • Pancreatitis / chemically induced
  • Pancreatitis / drug therapy
  • Pancreatitis / metabolism*
  • Pancreatitis / pathology
  • Peptidyl-Prolyl Isomerase F
  • Phenotype
  • Rats
  • Time Factors
  • Trehalose / pharmacology

Substances

  • Bile Acids and Salts
  • Peptidyl-Prolyl Isomerase F
  • PPIF protein, mouse
  • Ceruletide
  • Arginine
  • Trehalose
  • Mitochondrial Proton-Translocating ATPases
  • Cyclophilins
  • Ethionine