Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33491670

ABSTRACT

Severe acute pancreatitis (AP) is a life-threatening disease with up to 30% mortality. Therefore, prevention of AP aggravation and promotion of pancreatic regeneration are critical during the course and treatment of AP. Hypertriglyceridemia (HTG) is an established aggravating factor for AP that hinders pancreatic regeneration; however, its exact mechanism remains unclear. Using miRNA sequencing and further verification, we found that miRNA-153 (miR-153) was upregulated in the pancreas of HTG animal models and in the plasma of patients with HTG-AP. Increased miR-153 aggravated HTG-AP and delayed pancreatic repair via targeting TRAF3. Furthermore, miR-153 was transcriptionally suppressed by sterol regulatory element-binding transcription factor 1c (SREBP1c), which was suppressed by lipoprotein lipase malfunction-induced HTG. Overexpressing SREBP1c suppressed miR-153 expression, alleviated the severity of AP, and facilitated tissue regeneration in vivo. Finally, therapeutic administration of insulin also protected against HTG-AP via upregulating SREBP1c. Collectively, our results not only provide evidence that HTG leads to the development of more severe AP and hinders pancreatic regeneration via inducing persistent dysregulation of SREBP1c/miR-153 signaling, but also demonstrate that SREBP1c activators, including insulin, might be used to treat HTG-AP in patients.


Subject(s)
Hypertriglyceridemia/complications , MicroRNAs/genetics , Pancreatitis/complications , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Disease Models, Animal , Humans , Hypertriglyceridemia/genetics , Hypertriglyceridemia/physiopathology , Insulin/administration & dosage , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Pancreas/drug effects , Pancreas/pathology , Pancreas/physiopathology , Pancreatitis/genetics , Pancreatitis/physiopathology , Rats , Rats, Sprague-Dawley , Regeneration/genetics , Regeneration/physiology , Signal Transduction , TNF Receptor-Associated Factor 3/metabolism , Up-Regulation
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165971, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32950676

ABSTRACT

Acute pancreatitis (AP) is associated with impaired acinar cell autophagic flux, intracellular zymogen activation, cell necrosis and inflammation. Activation of the cholinergic system of vagus nerve has been shown to attenuate AP, but the effect of organ-intrinsic cholinergic system on pancreatitis remains unknown. In this study, we aim to examine the effect of α7 nicotinic acetylcholine receptor (α7nAChR) stimulation within the pancreas during AP. In vivo, AP was induced by caerulein plus LPS or ethanol plus palmitoleic acid in mice. In vitro, pancreatic acini were isolated and subjected to cholecystokinin (CCK) stimulation. Mice or acini were pre-treated with PNU-282987 (selective α7nAChR agonist) or methyllycaconitine citrate salt (selective α7nAChR antagonist). Pancreatitis severity, acinar cell injury, autophagic flux, and transcription factor EB (TFEB) pathway were analyzed. Both caerulein plus LPS in vivo and CCK in vitro led to an up-regulation of α7nAChR, indicating activation of pancreas-intrinsic α7nAChR signaling during AP. PNU-282987 decreased acinar cell injury, trypsinogen activation and pancreatitis severity. Conversely, methyllycaconitine citrate salt increased acinar cell injury and aggravated AP. Moreover, activation of α7nAChR by PNU-282987 promoted autophagic flux as indicated by reduced p62, increased LysoTracker staining and decreased number of autolysosomes with undegraded contents. Furthermore, PNU-282987 treatment significantly increased TFEB activity in pancreatic acinar cells. α7nAChR activation also attenuated pancreatic inflammation and NF-κB activation. Our results showed that activation of α7nAChR protected against experimental pancreatitis through enhancing TFEB-mediated acinar cell autophagy, suggesting that activation of pancreas-intrinsic α7nAChR may serve as an endogenous protective mechanism during AP.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Pancreatitis/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Aconitine/administration & dosage , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Autophagy/drug effects , Benzamides/administration & dosage , Benzamides/pharmacology , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/pharmacology , Ceruletide/administration & dosage , Ethanol/administration & dosage , Fatty Acids, Monounsaturated/administration & dosage , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred BALB C , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Signal Transduction/drug effects , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
3.
Front Pharmacol ; 11: 618, 2020.
Article in English | MEDLINE | ID: mdl-32457617

ABSTRACT

Impaired autophagy has been shown to play a critical role in experimental and human acute pancreatitis (AP). However, the mechanism for transcriptional regulation of autophagy remains largely unknown. In this study, we aim to explore the role of BRD4 (bromodomain-containing protein 4), a transcriptional repressor of autophagy, during AP. Changes in pancreatic BRD4 expression and the effect of BRD4 inhibition were measured in mice with AP (induced by caerulein and ethanol and palmitoleic acid) and in isolated pancreatic acinar cells stimulated with cholecystokinin (CCK). Pancreatitis severity was evaluated by serum amylase and pancreatic histopathology. The autophagic flux, the fusion of autophagosome and lysosome, and lysosomal degradation were evaluated. Sirtuin 1 (SIRT1) expression and the effect of SIRT1 inhibition were assessed. We found that pancreatic BRD4 expression was upregulated during various models of AP. BRD4 inhibition reduced CCK-stimulated pancreatic acinar cell injury and pro-inflammatory expression in vitro and protected against two models of experimental AP. Mechanistically, BRD4 inhibition restored impaired autophagic flux via promoting autophagosome-lysosome fusion and lysosomal degradation. BRD4 inhibition also upregulated SIRT1 and inhibition of SIRT1 reversed the effects of BRD4 inhibition on autophagic flux. Our data suggest that BRD4 is a potential therapeutic target for treating AP.

SELECTION OF CITATIONS
SEARCH DETAIL
...