Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Biol Chem ; 293(21): 8032-8047, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29626097

ABSTRACT

Mitochondrial dysfunction lies at the core of acute pancreatitis (AP). Diverse AP stimuli induce Ca2+-dependent formation of the mitochondrial permeability transition pore (MPTP), a solute channel modulated by cyclophilin D (CypD), the formation of which causes ATP depletion and necrosis. Oxidative stress reportedly triggers MPTP formation and is elevated in clinical AP, but how reactive oxygen species influence cell death is unclear. Here, we assessed potential MPTP involvement in oxidant-induced effects on pancreatic acinar cell bioenergetics and fate. H2O2 application promoted acinar cell apoptosis at low concentrations (1-10 µm), whereas higher levels (0.5-1 mm) elicited rapid necrosis. H2O2 also decreased the mitochondrial NADH/FAD+ redox ratio and ΔΨm in a concentration-dependent manner (10 µm to 1 mm H2O2), with maximal effects at 500 µm H2O2 H2O2 decreased the basal O2 consumption rate of acinar cells, with no alteration of ATP turnover at <50 µm H2O2 However, higher H2O2 levels (≥50 µm) diminished spare respiratory capacity and ATP turnover, and bioenergetic collapse, ATP depletion, and cell death ensued. Menadione exerted detrimental bioenergetic effects similar to those of H2O2, which were inhibited by the antioxidant N-acetylcysteine. Oxidant-induced bioenergetic changes, loss of ΔΨm, and cell death were not ameliorated by genetic deletion of CypD or by its acute inhibition with cyclosporine A. These results indicate that oxidative stress alters mitochondrial bioenergetics and modifies pancreatic acinar cell death. A shift from apoptosis to necrosis appears to be associated with decreased mitochondrial spare respiratory capacity and ATP production, effects that are independent of CypD-sensitive MPTP formation.


Subject(s)
Apoptosis , Cyclophilins/physiology , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/physiology , Necrosis , Oxidative Stress , Pancreas/pathology , Acinar Cells/metabolism , Acinar Cells/pathology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Energy Metabolism , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Permeability Transition Pore , Pancreas/metabolism , Reactive Oxygen Species/metabolism
2.
Pancreatology ; 19(2): 209-216, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30611702

ABSTRACT

Ethyl pyruvate (EP) has been shown to improve outcomes from multiple organ dysfunction syndrome (MODS) in experimental animal models of critical illness. This review aimed to summarise in vitro and in vivo effects of EP analogs on acute pancreatitis (AP) with the objective of proposing medicinal chemistry modifications of EP for future research. In vitro studies showed that both sodium pyruvate and EP significantly reduced pancreatic acinar necrotic cell death pathway activation induced by multiple pancreatic toxins. In vivo studies using different murine AP models showed that EP (usually at a dose of 40 mg/kg every 6 h) consistently reduced pain, markers of pancreatic injury, systemic inflammation and MODS. There was also a significant increase in survival rate, even when EP was administered 12 h after disease induction (compared with untreated groups or those treated with Ringer's lactate solution). Experimental studies suggest that EP and analogs are promising drug candidates for treating AP. EP or analogs can undergo medicinal chemistry modifications to improve its stability and deliverability. EP or analogs could be evaluated as a supplement to intravenous fluid therapy in AP.


Subject(s)
Pancreatitis/drug therapy , Pyruvates/therapeutic use , Animals , Biomarkers , Humans , Inflammation , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology
3.
J Physiol ; 596(13): 2547-2564, 2018 07.
Article in English | MEDLINE | ID: mdl-29717784

ABSTRACT

KEY POINTS: Giant trypsin-containing endocytic vacuoles are formed in pancreatic acinar cells stimulated with inducers of acute pancreatitis. F-actin envelops endocytic vacuoles and regulates their properties. Endocytic vacuoles can rupture and release their content into the cytosol of acinar cells. Endocytic vacuoles can fuse with the plasma membrane of acinar cells and exocytose their content. ABSTRACT: Intrapancreatic activation of trypsinogen is an early event in and hallmark of the development of acute pancreatitis. Endocytic vacuoles, which form by disconnection and transport of large post-exocytic structures, are the only resolvable sites of the trypsin activity in live pancreatic acinar cells. In the present study, we characterized the dynamics of endocytic vacuole formation induced by physiological and pathophysiological stimuli and visualized a prominent actin coat that completely or partially surrounded endocytic vacuoles. An inducer of acute pancreatitis taurolithocholic acid 3-sulphate and supramaximal concentrations of cholecystokinin triggered the formation of giant (more than 2.5 µm in diameter) endocytic vacuoles. We discovered and characterized the intracellular rupture of endocytic vacuoles and the fusion of endocytic vacuoles with basal and apical regions of the plasma membrane. Experiments with specific protease inhibitors suggest that the rupture of endocytic vacuoles is probably not induced by trypsin or cathepsin B. Perivacuolar filamentous actin (observed on the surface of ∼30% of endocytic vacuoles) may play a stabilizing role by preventing rupture of the vacuoles and fusion of the vacuoles with the plasma membrane. The rupture and fusion of endocytic vacuoles allow trypsin to escape the confinement of a membrane-limited organelle, gain access to intracellular and extracellular targets, and initiate autodigestion of the pancreas, comprising a crucial pathophysiological event.


Subject(s)
Acinar Cells/pathology , Exocytosis , Pancreas, Exocrine/pathology , Pancreatitis/pathology , Transport Vesicles/pathology , Vacuoles/physiology , Acinar Cells/metabolism , Acute Disease , Animals , Male , Mice , Pancreas, Exocrine/metabolism , Pancreatitis/etiology , Transport Vesicles/metabolism
4.
Gut ; 66(2): 301-313, 2017 02.
Article in English | MEDLINE | ID: mdl-26642860

ABSTRACT

OBJECTIVE: Caffeine reduces toxic Ca2+ signals in pancreatic acinar cells via inhibition of inositol 1,4,5-trisphosphate receptor (IP3R)-mediated signalling, but effects of other xanthines have not been evaluated, nor effects of xanthines on experimental acute pancreatitis (AP). We have determined effects of caffeine and its xanthine metabolites on pancreatic acinar IP3R-mediated Ca2+ signalling and experimental AP. DESIGN: Isolated pancreatic acinar cells were exposed to secretagogues, uncaged IP3 or toxins that induce AP and effects of xanthines, non-xanthine phosphodiesterase (PDE) inhibitors and cyclic adenosine monophosphate and cyclic guanosine monophosphate (cAMP/cGMP) determined. The intracellular cytosolic calcium concentration ([Ca2+]C), mitochondrial depolarisation and necrosis were assessed by confocal microscopy. Effects of xanthines were evaluated in caerulein-induced AP (CER-AP), taurolithocholic acid 3-sulfate-induced AP (TLCS-AP) or palmitoleic acid plus ethanol-induced AP (fatty acid ethyl ester AP (FAEE-AP)). Serum xanthines were measured by liquid chromatography-mass spectrometry. RESULTS: Caffeine, dimethylxanthines and non-xanthine PDE inhibitors blocked IP3-mediated Ca2+ oscillations, while monomethylxanthines had little effect. Caffeine and dimethylxanthines inhibited uncaged IP3-induced Ca2+ rises, toxin-induced Ca2+ release, mitochondrial depolarisation and necrotic cell death pathway activation; cAMP/cGMP did not inhibit toxin-induced Ca2+ rises. Caffeine significantly ameliorated CER-AP with most effect at 25 mg/kg (seven injections hourly); paraxanthine or theophylline did not. Caffeine at 25 mg/kg significantly ameliorated TLCS-AP and FAEE-AP. Mean total serum levels of dimethylxanthines and trimethylxanthines peaked at >2 mM with 25 mg/kg caffeine but at <100 µM with 25 mg/kg paraxanthine or theophylline. CONCLUSIONS: Caffeine and its dimethylxanthine metabolites reduced pathological IP3R-mediated pancreatic acinar Ca2+ signals but only caffeine ameliorated experimental AP. Caffeine is a suitable starting point for medicinal chemistry.


Subject(s)
Acinar Cells/drug effects , Caffeine/pharmacology , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Pancreas/pathology , Pancreatitis/prevention & control , Phosphodiesterase Inhibitors/pharmacology , Acinar Cells/metabolism , Animals , Caffeine/therapeutic use , Cell Death/drug effects , Cells, Cultured , Ceruletide , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cytosol/metabolism , Ethanol , Fatty Acids, Monounsaturated , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/physiology , Necrosis/diagnostic imaging , Pancreatitis/blood , Pancreatitis/chemically induced , Phosphodiesterase Inhibitors/therapeutic use , Signal Transduction/drug effects , Taurolithocholic Acid/analogs & derivatives , Xanthines/blood , Xanthines/pharmacology
5.
Pancreatology ; 17(6): 884-892, 2017.
Article in English | MEDLINE | ID: mdl-29102149

ABSTRACT

BACKGROUND: Clinical and experimental acute pancreatitis feature histone release within the pancreas from innate immune cells and acinar cell necrosis. In this study, we aimed to detail the source of circulating histones and assess their role in the pathogenesis of acute pancreatitis. METHODS: Circulating nucleosomes were measured in patient plasma, taken within 24 and 48 h of onset of acute pancreatitis and correlated with clinical outcomes. Using caerulein hyperstimulation, circulating histones were measured in portal, systemic venous and systemic arterial circulation in mice, and the effects of systemic administration of histones in this model were assessed. The sites of actions of circulating histones were assessed by administration of FITC-labelled histones. The effects of histones on isolated pancreatic acinar cells were further assessed by measuring acinar cell death and calcium permeability in vitro. RESULTS: Cell-free histones were confirmed to be abundant in human acute pancreatitis and found to derive from pancreatitis-associated liver injury in a rodent model of the disease. Fluorescein isothianate-labelled histones administered systemically targeted the pancreas and exacerbated injury in experimental acute pancreatitis. Histones induce charge- and concentration-dependent plasmalemma leakage and necrosis in isolated pancreatic acinar cells, independent of extracellular calcium. CONCLUSION: We conclude that histones released systemically in acute pancreatitis concentrate within the inflamed pancreas and exacerbate injury. Circulating histones may provide meaningful biomarkers and targets for therapy in clinical acute pancreatitis.


Subject(s)
Histones/blood , Histones/metabolism , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/blood , Pancreatitis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Necrosis/metabolism , Pancreatitis/chemically induced , Young Adult
6.
Gut ; 65(8): 1333-46, 2016 08.
Article in English | MEDLINE | ID: mdl-26071131

ABSTRACT

OBJECTIVE: Acute pancreatitis is caused by toxins that induce acinar cell calcium overload, zymogen activation, cytokine release and cell death, yet is without specific drug therapy. Mitochondrial dysfunction has been implicated but the mechanism not established. DESIGN: We investigated the mechanism of induction and consequences of the mitochondrial permeability transition pore (MPTP) in the pancreas using cell biological methods including confocal microscopy, patch clamp technology and multiple clinically representative disease models. Effects of genetic and pharmacological inhibition of the MPTP were examined in isolated murine and human pancreatic acinar cells, and in hyperstimulation, bile acid, alcoholic and choline-deficient, ethionine-supplemented acute pancreatitis. RESULTS: MPTP opening was mediated by toxin-induced inositol trisphosphate and ryanodine receptor calcium channel release, and resulted in diminished ATP production, leading to impaired calcium clearance, defective autophagy, zymogen activation, cytokine production, phosphoglycerate mutase 5 activation and necrosis, which was prevented by intracellular ATP supplementation. When MPTP opening was inhibited genetically or pharmacologically, all biochemical, immunological and histopathological responses of acute pancreatitis in all four models were reduced or abolished. CONCLUSIONS: This work demonstrates the mechanism and consequences of MPTP opening to be fundamental to multiple forms of acute pancreatitis and validates the MPTP as a drug target for this disease.


Subject(s)
Acinar Cells , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/metabolism , Pancreas , Pancreatitis, Acute Necrotizing , Phosphoprotein Phosphatases/metabolism , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Autophagy/drug effects , Calcium/metabolism , Cell Culture Techniques , Disease Models, Animal , Humans , Inositol Phosphates/metabolism , Inositol Phosphates/pharmacology , Mice , Mitochondria/enzymology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Necrosis , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/metabolism , Pancreatitis, Acute Necrotizing/pathology
7.
Gastroenterology ; 149(2): 481-92.e7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25917787

ABSTRACT

BACKGROUND & AIMS: Sustained activation of the cytosolic calcium concentration induces injury to pancreatic acinar cells and necrosis. The calcium release-activated calcium modulator ORAI1 is the most abundant Ca(2+) entry channel in pancreatic acinar cells; it sustains calcium overload in mice exposed to toxins that induce pancreatitis. We investigated the roles of ORAI1 in pancreatic acinar cell injury and the development of acute pancreatitis in mice. METHODS: Mouse and human acinar cells, as well as HEK 293 cells transfected to express human ORAI1 with human stromal interaction molecule 1, were hyperstimulated or incubated with human bile acid, thapsigargin, or cyclopiazonic acid to induce calcium entry. GSK-7975A or CM_128 were added to some cells, which were analyzed by confocal and video microscopy and patch clamp recordings. Acute pancreatitis was induced in C57BL/6J mice by ductal injection of taurolithocholic acid 3-sulfate or intravenous' administration of cerulein or ethanol and palmitoleic acid. Some mice then were given GSK-7975A or CM_128, which inhibit ORAI1, at different time points to assess local and systemic effects. RESULTS: GSK-7975A and CM_128 each separately inhibited toxin-induced activation of ORAI1 and/or activation of Ca(2+) currents after Ca(2+) release, in a concentration-dependent manner, in mouse and human pancreatic acinar cells (inhibition >90% of the levels observed in control cells). The ORAI1 inhibitors also prevented activation of the necrotic cell death pathway in mouse and human pancreatic acinar cells. GSK-7975A and CM_128 each inhibited all local and systemic features of acute pancreatitis in all 3 models, in dose- and time-dependent manners. The agents were significantly more effective, in a range of parameters, when given at 1 vs 6 hours after induction of pancreatitis. CONCLUSIONS: Cytosolic calcium overload, mediated via ORAI1, contributes to the pathogenesis of acute pancreatitis. ORAI1 inhibitors might be developed for the treatment of patients with pancreatitis.


Subject(s)
Acinar Cells/drug effects , Benzamides/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium/metabolism , Pancreatitis/drug therapy , Pyrazoles/pharmacology , Acinar Cells/cytology , Acute Disease , Animals , Bile Acids and Salts/toxicity , Calcium/toxicity , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Indoles/toxicity , Mice , Mice, Inbred C57BL , ORAI1 Protein , Pancreatitis/chemically induced , Pancreatitis/metabolism , Thapsigargin/toxicity , Time Factors , Treatment Outcome
8.
Biochem J ; 465(3): 405-12, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25370603

ABSTRACT

The inducers of acute pancreatitis trigger a prolonged increase in the cytosolic Ca(2+) concentration ([Ca(2+)]c), which is responsible for the damage to and eventual death of pancreatic acinar cells. Vacuolization is an important indicator of pancreatic acinar cell damage. Furthermore, activation of trypsinogen occurs in the endocytic vacuoles; therefore the vacuoles can be considered as 'initiating' organelles in the development of the cell injury. In the present study, we investigated the relationship between the formation of endocytic vacuoles and Ca(2+) influx developed in response to the inducers of acute pancreatitis [bile acid taurolithocholic acid 3-sulfate (TLC-S) and supramaximal concentration of cholecystokinin-8 (CCK)]. We found that the inhibitor of STIM (stromal interaction molecule)/Orai channels, GSK-7975A, effectively suppressed both the Ca(2+) influx (stimulated by inducers of pancreatitis) and the formation of endocytic vacuoles. Cell death induced by TLC-S or CCK was also inhibited by GSK-7975A. We documented the formation of endocytic vacuoles in response to store-operated Ca(2+) entry (SOCE) induced by thapsigargin [TG; inhibitor of sarcoplasmic/endoplasmic reticulum (ER) Ca(2+) pumps] and observed strong inhibition of TG-induced vacuole formation by GSK-7975A. Finally, we found that structurally-unrelated inhibitors of calpain suppress formation of endocytic vacuoles, suggesting that this Ca2+-dependent protease is a mediator between Ca(2+) elevation and endocytic vacuole formation.


Subject(s)
Acinar Cells/metabolism , Calcium/metabolism , Pancreas/cytology , Pancreas/metabolism , Transport Vesicles/metabolism , Vacuoles/metabolism , Animals , Cells, Cultured , Mice
9.
Mediators Inflamm ; 2015: 901780, 2015.
Article in English | MEDLINE | ID: mdl-25878403

ABSTRACT

Although oxidative stress has been strongly implicated in the development of acute pancreatitis (AP), antioxidant therapy in patients has so far been discouraging. The aim of this study was to assess potential protective effects of a mitochondria-targeted antioxidant, MitoQ, in experimental AP using in vitro and in vivo approaches. MitoQ blocked H2O2-induced intracellular ROS responses in murine pancreatic acinar cells, an action not shared by the control analogue dTPP. MitoQ did not reduce mitochondrial depolarisation induced by either cholecystokinin (CCK) or bile acid TLCS, and at 10 µM caused depolarisation per se. Both MitoQ and dTPP increased basal and CCK-induced cell death in a plate-reader assay. In a TLCS-induced AP model MitoQ treatment was not protective. In AP induced by caerulein hyperstimulation (CER-AP), MitoQ exerted mixed effects. Thus, partial amelioration of histopathology scores was observed, actions shared by dTPP, but without reduction of the biochemical markers pancreatic trypsin or serum amylase. Interestingly, lung myeloperoxidase and interleukin-6 were concurrently increased by MitoQ in CER-AP. MitoQ caused biphasic effects on ROS production in isolated polymorphonuclear leukocytes, inhibiting an acute increase but elevating later levels. Our results suggest that MitoQ would be inappropriate for AP therapy, consistent with prior antioxidant evaluations in this disease.


Subject(s)
Antioxidants/chemistry , Mitochondria/metabolism , Organophosphorus Compounds/chemistry , Pancreatitis/metabolism , Ubiquinone/analogs & derivatives , Acinar Cells/metabolism , Acute Disease , Animals , Apoptosis , Ceruletide/chemistry , Cholecystokinin/chemistry , Disease Models, Animal , Inflammation/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Necrosis/metabolism , Oxidative Stress , Pancreas/metabolism , Reactive Oxygen Species/metabolism , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/chemistry , Ubiquinone/chemistry
10.
Gut ; 63(8): 1313-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24162590

ABSTRACT

OBJECTIVE: Non-oxidative metabolism of ethanol (NOME) produces fatty acid ethyl esters (FAEEs) via carboxylester lipase (CEL) and other enzyme action implicated in mitochondrial injury and acute pancreatitis (AP). This study investigated the relative importance of oxidative and non-oxidative pathways in mitochondrial dysfunction, pancreatic damage and development of alcoholic AP, and whether deleterious effects of NOME are preventable. DESIGN: Intracellular calcium ([Ca(2+)](C)), NAD(P)H, mitochondrial membrane potential and activation of apoptotic and necrotic cell death pathways were examined in isolated pancreatic acinar cells in response to ethanol and/or palmitoleic acid (POA) in the presence or absence of 4-methylpyrazole (4-MP) to inhibit oxidative metabolism. A novel in vivo model of alcoholic AP induced by intraperitoneal administration of ethanol and POA was developed to assess the effects of manipulating alcohol metabolism. RESULTS: Inhibition of OME with 4-MP converted predominantly transient [Ca(2+)](C) rises induced by low ethanol/POA combination to sustained elevations, with concurrent mitochondrial depolarisation, fall of NAD(P)H and cellular necrosis in vitro. All effects were prevented by 3-benzyl-6-chloro-2-pyrone (3-BCP), a CEL inhibitor. 3-BCP also significantly inhibited rises of pancreatic FAEE in vivo and ameliorated acute pancreatic damage and inflammation induced by administration of ethanol and POA to mice. CONCLUSIONS: A combination of low ethanol and fatty acid that did not exert deleterious effects per se became toxic when oxidative metabolism was inhibited. The in vitro and in vivo damage was markedly inhibited by blockade of CEL, indicating the potential for development of specific therapy for treatment of alcoholic AP via inhibition of FAEE generation.


Subject(s)
Acyltransferases/antagonists & inhibitors , Calcium/metabolism , Carboxylesterase/metabolism , Ethanol/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Pancreatitis, Alcoholic/metabolism , Pyrones/pharmacology , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Apoptosis/drug effects , Calcium Signaling , Carboxylesterase/antagonists & inhibitors , Cells, Cultured , Disease Models, Animal , Ethanol/toxicity , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fomepizole , Mice , NADP/metabolism , Necrosis , Pancreatitis, Alcoholic/chemically induced , Pancreatitis, Alcoholic/pathology , Pyrazoles/pharmacology
11.
Autophagy ; : 1-13, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39216469

ABSTRACT

The WD40 domain (WDD) of ATG16L1 plays a pivotal role in non-canonical autophagy. This study examined the role of recently identified LAP-like non-canonical autophagy (LNCA) in acute pancreatitis. LNCA involves rapid single-membrane LC3 conjugation to endocytic vacuoles in pancreatic acinar cells. The rationale for this study was the previously observed presence of trypsin in the organelles undergoing LNCA; aberrant trypsin formation is an important factor in pancreatitis development. Here we report that the deletion of WDD (attained in ATG16L1[E230] mice) eliminated LNCA, aggravated caerulein-induced acute pancreatitis and suppressed the fast trypsin degradation observed in both a rapid caerulein-induced disease model and in caerulein-treated isolated pancreatic acinar cells. These experiments indicate that LNCA is a WDD-dependent mechanism and suggest that it plays not an activating but a protective role in acute pancreatitis. Furthermore, palmitoleic acid, another inducer of experimental acute pancreatitis, strongly inhibited LNCA, suggesting a novel mechanism of pancreatic lipotoxicity.Abbreviation: AMY: amylase; AP: acute pancreatitis; CASM: conjugation of Atg8 to single membranes; CCK: cholecystokinin; FAEE model: fatty acid and ethanol model; IL6: interleukin 6; LA: linoleic acid; LAP: LC3-associated phagocytosis; LMPO: lung myeloperoxidase; LNCA: LAP-like non-canonical autophagy; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MPO: myeloperoxidase; PMPO: pancreatic myeloperoxidase; POA: palmitoleic acid; WDD: WD40 domain; WT: wild type.

12.
Cells ; 11(16)2022 08 12.
Article in English | MEDLINE | ID: mdl-36010591

ABSTRACT

Recent studies have highlighted the importance of autophagy and particularly non-canonical autophagy in the development and progression of acute pancreatitis (a frequent disease with considerable morbidity and significant mortality). An important early event in the development of acute pancreatitis is the intrapancreatic activation of trypsinogen, (i.e., formation of trypsin) leading to the autodigestion of the organ. Another prominent phenomenon associated with the initiation of this disease is vacuolisation and specifically the formation of giant endocytic vacuoles in pancreatic acinar cells. These organelles develop in acinar cells exposed to several inducers of acute pancreatitis (including taurolithocholic acid and high concentrations of secretagogues cholecystokinin and acetylcholine). Notably, early trypsinogen activation occurs in the endocytic vacuoles. These trypsinogen-activating organelles undergo activation, long-distance trafficking, and non-canonical autophagy. In this review, we will discuss the role of autophagy in acute pancreatitis and particularly focus on the recently discovered LAP-like non-canonical autophagy (LNCA) of endocytic vacuoles.


Subject(s)
Pancreatitis , Trypsinogen , Acute Disease , Autophagy , Humans , Vacuoles
13.
Cells ; 10(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925729

ABSTRACT

Acute pancreatitis (AP) is a severe and potentially fatal disease caused predominantly by alcohol excess and gallstones, which lacks a specific therapy. The role of Receptor-Interacting Protein Kinase 1 (RIPK1), a key component of programmed necrosis (Necroptosis), is unclear in AP. We assessed the effects of RIPK1 inhibitor Necrostatin-1 (Nec-1) and RIPK1 modification (RIPK1K45A: kinase dead) in bile acid (TLCS-AP), alcoholic (FAEE-AP) and caerulein hyperstimulation (CER-AP) mouse models. Involvement of collateral Nec-1 target indoleamine 2,3-dioxygenase (IDO) was probed with the inhibitor Epacadostat (EPA). Effects of Nec-1 and RIPK1K45A were also compared on pancreatic acinar cell (PAC) fate in vitro and underlying mechanisms explored. Nec-1 markedly ameliorated histological and biochemical changes in all models. However, these were only partially reduced or unchanged in RIPK1K45A mice. Inhibition of IDO with EPA was protective in TLCS-AP. Both Nec-1 and RIPK1K45A modification inhibited TLCS- and FAEE-induced PAC necrosis in vitro. Nec-1 did not affect TLCS-induced Ca2+ entry in PACs, however, it inhibited an associated ROS elevation. The results demonstrate protective actions of Nec-1 in multiple models. However, RIPK1-dependent necroptosis only partially contributed to beneficial effects, and actions on targets such as IDO are likely to be important.


Subject(s)
Imidazoles/therapeutic use , Indoles/therapeutic use , Pancreatitis/drug therapy , Pancreatitis/enzymology , Protective Agents/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acinar Cells/metabolism , Alcohols , Animals , Bile Acids and Salts , Calcium/metabolism , Ceruletide , Imidazoles/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoles/pharmacology , Male , Mice, Inbred C57BL , Pancreas/pathology , Pancreatitis/chemically induced , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
14.
Biochim Biophys Acta ; 1787(11): 1374-82, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19344663

ABSTRACT

In this review we will attempt to summarise the complex and sometimes contradictory effects that mitochondria have on different forms of calcium signalling. Mitochondria can influence Ca(2+) signalling indirectly by changing the concentration of ATP, NAD(P)H, pyruvate and reactive oxygen species - which in turn modulate components of the Ca(2+) signalling machinery i.e. buffering, release from internal stores, influx from the extracellular solution, uptake into cellular organelles and extrusion by plasma membrane Ca(2+) pumps. Mitochondria can directly influence the calcium concentration in the cytosol of the cell by importing Ca(2+) via the mitochondrial Ca(2+) uniporter or transporting Ca(2+) from the interior of the organelle into the cytosol by means of Na+/Ca(2+) or H+/Ca(2+) exchangers. Considerable progress in understanding the relationship between Ca(2+) signalling cascades and mitochondrial physiology has been accumulated over the last few years due to the development of more advanced optical techniques and electrophysiological approaches.


Subject(s)
Calcium Signaling , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans , Inositol 1,4,5-Trisphosphate Receptors/physiology , Ryanodine Receptor Calcium Release Channel/physiology
15.
Biochem J ; 425(1): 159-68, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-19843011

ABSTRACT

Ca2+ entry through store-operated Ca2+ channels involves the interaction at ER-PM (endoplasmic reticulum-plasma membrane) junctions of STIM (stromal interaction molecule) and Orai. STIM proteins are sensors of the luminal ER Ca2+ concentration and, following depletion of ER Ca2+, they oligomerize and translocate to ER-PM junctions where they form STIM puncta. Direct binding to Orai proteins activates their Ca2+ channel function. It has been suggested that an additional interaction of the C-terminal polybasic domain of STIM1 with PM phosphoinositides could contribute to STIM1 puncta formation prior to binding to Orai. In the present study, we investigated the role of phosphoinositides in the formation of STIM1 puncta and SOCE (store-operated Ca2+ entry) in response to store depletion. Treatment of HeLa cells with inhibitors of PI3K (phosphatidylinositol 3-kinase) and PI4K (phosphatidylinositol 4-kinase) (wortmannin and LY294002) partially inhibited formation of STIM1 puncta. Additional rapid depletion of PtdIns(4,5)P2 resulted in more substantial inhibition of the translocation of STIM1-EYFP (enhanced yellow fluorescent protein) into puncta. The inhibition was extensive at a concentration of LY294002 (50 microM) that should primarily inhibit PI3K, consistent with a major role for PtdIns(4,5)P2 and PtdIns(3,4,5)P3 in puncta formation. Depletion of phosphoinositides also inhibited SOCE based on measurement of the rise in intracellular Ca2+ concentration after store depletion. Overexpression of Orai1 resulted in a recovery of translocation of STMI1 into puncta following phosphoinositide depletion and, under these conditions, SOCE was increased to above control levels. These observations support the idea that phosphoinositides are not essential but contribute to STIM1 accumulation at ER-PM junctions with a second translocation mechanism involving direct STIM1-Orai interactions.


Subject(s)
Calcium/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositols/metabolism , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/metabolism , Adenosine Triphosphate/metabolism , Androstadienes/pharmacology , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Membrane/metabolism , Chromones/pharmacology , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Microscopy, Confocal , Morpholines/pharmacology , Neoplasm Proteins/genetics , ORAI1 Protein , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Binding , Protein Transport/drug effects , Stromal Interaction Molecule 1 , Transfection , Wortmannin
16.
Cells ; 9(6)2020 06 05.
Article in English | MEDLINE | ID: mdl-32516955

ABSTRACT

Acute pancreatitis is a frequent disease that lacks specific drug treatment. Unravelling the molecular mechanisms of acute pancreatitis is essential for the development of new therapeutics. Several inducers of acute pancreatitis trigger sustained Ca2+ increases in the cytosol and mitochondria of pancreatic acinar cells. The mitochondrial calcium uniporter (MCU) mediates mitochondrial Ca2+ uptake that regulates bioenergetics and plays an important role in cell survival, damage and death. Aberrant Ca2+ signaling and mitochondrial damage in pancreatic acinar cells have been implicated in the initiation of acute pancreatitis. The primary aim of this study was to assess the involvement of the MCU in experimental acute pancreatitis. We found that pancreatic acinar cells from MCU-/- mice display dramatically reduced mitochondrial Ca2+ uptake. This is consistent with the drastic changes of stimulus-metabolism coupling, manifested by the reduction of mitochondrial NADH/FAD+ responses to cholecystokinin and in the decrease of cholecystokinin-stimulated oxygen consumption. However, in three experimental models of acute pancreatitis (induced by caerulein, taurolithocholic acid 3-sulfate or palmitoleic acid plus ethanol), MCU knockout failed to reduce the biochemical and histological changes characterizing the severity of local and systemic damage. A possible explanation of this surprising finding is the redundancy of damaging mechanisms activated by the inducers of acute pancreatitis.


Subject(s)
Acinar Cells/metabolism , Calcium Channels/metabolism , Pancreas/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Severity of Illness Index , Animals , Calcium/metabolism , Calcium Signaling , Cytosol/metabolism , Disease Models, Animal , Flavin-Adenine Dinucleotide/metabolism , Mice, Knockout , Mitochondria/metabolism , NAD/metabolism
17.
Autophagy ; 16(7): 1314-1331, 2020 07.
Article in English | MEDLINE | ID: mdl-31651224

ABSTRACT

Activation of trypsinogen (formation of trypsin) inside the pancreas is an early pathological event in the development of acute pancreatitis. In our previous studies we identified the activation of trypsinogen within endocytic vacuoles (EVs), cellular organelles that appear in pancreatic acinar cells treated with the inducers of acute pancreatitis. EVs are formed as a result of aberrant compound exocytosis and subsequent internalization of post-exocytic structures. These organelles can be up to 12 µm in diameter and can be actinated (i.e. coated with F-actin). Notably, EVs can undergo intracellular rupture and fusion with the plasma membrane, providing trypsin with access to cytoplasmic and extracellular targets. Unraveling the mechanisms involved in cellular processing of EVs is an interesting cell biological challenge with potential benefits for understanding acute pancreatitis. In this study we have investigated autophagy of EVs and discovered that it involves a non-canonical LC3-conjugation mechanism, reminiscent in its properties to LC3-associated phagocytosis (LAP); in both processes LC3 was recruited to single, outer organellar membranes. Trypsinogen activation peptide was observed in approximately 55% of LC3-coated EVs indicating the relevance of the described process to the early cellular events of acute pancreatitis. We also investigated relationships between actination and non-canonical autophagy of EVs and concluded that these processes represent sequential steps in the evolution of EVs. Our study expands the known roles of LAP and indicates that, in addition to its well-established functions in phagocytosis and macropinocytosis, LAP is also involved in the processing of post-exocytic organelles in exocrine secretory cells. ABBREVIATIONS: AP: acute pancreatitis; CCK: cholecystokinin; CLEM: correlative light and electron microscopy; DPI: diphenyleneiodonium; EV: endocytic vacuole; LAP: LC3-associate phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; PACs: pancreatic acinar cells; PFA: paraformaldehyde; PtdIns3K: phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; Res: resveratrol; TAP: trypsinogen activation peptide; TEM: transmission electron microscopy; TLC-S: taurolithocholic acid 3-sulfate; TRD: Dextran Texas Red 3000 MW Neutral; ZGs: zymogen granules.


Subject(s)
Acinar Cells/metabolism , Autophagy , Endocytosis , Microtubule-Associated Proteins/metabolism , Pancreas/cytology , Phagocytosis , Vacuoles/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Acinar Cells/drug effects , Acinar Cells/ultrastructure , Actins/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/metabolism , Chloroquine/pharmacology , Cholecystokinin/pharmacology , Mice, Inbred C57BL , Onium Compounds/pharmacology , Phagocytosis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology , Taurolithocholic Acid/analogs & derivatives , Trypsinogen/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/drug effects
18.
J Ethnopharmacol ; 257: 112861, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32315735

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Dachengqi decoction (DCQD) belongs to a family of purgative herbal formulas widely used in China for the treatment of acute pancreatitis (AP). AP is a prevalent digestive disease currently without an effective pharmacological intervention. Formula granules have become the preferred method for delivery of herbal formulation in China given its benefit of potency retention, dosing precision and ease of use. The efficacy of DCQD formula granules (DFGs) in experimental AP models has not been investigated. AIM OF THE STUDY: To analyse and compare the differences in chemical composition of DFGs, with their aqueous extraction (AE) and chloroform extraction (CE) derivatives. To assess their efficacy on severity and targeted pancreatic pro-inflammatory signalling pathways in freshly isolated acinar cells and two models of experimental AP. MATERIAL AND METHODS: UPLC-Q-TOF-MS was used to analyse chemical components of DFGs and their extractions. Freshly isolated mouse pancreatic acinar cells were treated with taurolithocholic acid 3-sulphate disodium salt (TLCS, 500 µM) with or without DFGs, AE and CE. Apoptotic and necrotic cell death pathway activation was measured by caspase 3/7 (10 µl/mL) and propidium iodide (PI, 1 µM), respectively, using a fluorescent plate reader. Necrotic acinar cells were also counted by epifluorescence microscopy. Mice received either 7 intraperitoneal injections of caerulein (50 µg/kg) at hourly intervals or retrograde infusion of TLCS (3 mM, 50 µl) to induce AP (CER-AP and TLCS-AP, respectively). In CER-AP, mice received oral gavage of DFGs (2.1, 4.2 and 5.2 g/kg), AE (0.6, 1.2, and 2.4 g/kg) and CE (4, 9 and 17 mg/kg), or matched DFGs (1.8 g/kg) and AE (1 g/kg) for 3 times at 2-hourly intervals, or a single intraperitoneal injection of DCQD-related monomers rhein (20 mg/kg), narigeinine (25 mg/kg), and honokiol (5 mg/kg) begun at the 3rd injection of caerulein. In TLCS-AP, DFGs (4.2 g/kg) were given orally at 1, 3 and 5 h post-surgery. Disease severity and pancreatic pro-inflammatory markers were determined. RESULTS: The main effective anthraquinones and their glycosides, flavonoids and their glycosides, polyphenols and lignans were found in the DFGs. A higher proportion of polar components including glycosides attached to anthraquinones, phenols and flavonoids was found in AE. Conversely, lower polar components containing methoxy substituted flavonoids and anthraquinones were more abundant in CE. DFGs were given at 4.2 g/kg, a consistent reduction in the pancreatic histopathology score and severity indices was observed in both CER-AP and TLCS-AP. In vitro, AE significantly reduced both apoptotic and necrotic cell death pathway activation, while CE increased TLCS-induced acinar cell necrosis. In vivo, AE at dose of 1.2 g/kg consistently reduced pancreatic histopathological scores and myeloperoxidase in the CER-AP that were associated with suppressed expression of pro-inflammatory meditator mRNAs and proteins. CE increased lung myeloperoxidase and failed to protect against CER-AP in all dosages. AE was demonstrated to be more effective than DFGs in reducing pancreatic histopathological scores and myeloperoxidase. CONCLUSIONS: AE from DFGs alleviated the severity of mouse AP models via an inhibition of pancreatic pro-inflammatory signalling pathways. Efficacy of AE on experimental AP was more potent than its original DFGs and DCQD monomers.


Subject(s)
Acinar Cells/drug effects , Anti-Inflammatory Agents/pharmacology , Inflammation Mediators , Pancreas, Exocrine/drug effects , Pancreatitis/prevention & control , Plant Extracts/pharmacology , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Apoptosis/drug effects , Chloroform/chemistry , Disease Models, Animal , Male , Mice, Inbred C57BL , Necrosis , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Signal Transduction , Solvents/chemistry , Water/chemistry
19.
Gastroenterology ; 135(2): 632-41, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18555802

ABSTRACT

BACKGROUND & AIMS: Cholecystokinin (CCK) has been thought to act only indirectly on human pancreatic acinar cells via vagal nerve stimulation, rather than by direct CCK receptor activation as on rodent pancreatic acinar cells. We tested whether CCK (CCK-8 and human CCK-58) can act directly on human pancreatic acinar cells. METHODS: Human acinar cells were freshly isolated from pancreatic transection line samples, loaded with Fluo4-AM or quinacrine, and examined for Ca(2+), metabolic and secretory responses to CCK-8, human CCK-58, or acetylcholine with confocal microscopy. RESULTS: CCK-8 and human CCK-58 at physiologic concentrations (1-20 pmol/L) elicited rapid, robust, oscillatory increases of the cytosolic Ca(2+) ion concentration, showing apical to basal progression, in acinar cells from 14 patients with unobstructed pancreata. The cytosolic Ca(2+) ion concentration increases were followed by increases in mitochondrial adenosine triphosphate production and secretion. CCK-elicited Ca(2+) signals and exocytosis were not inhibited by atropine (1 mumol/L) or tetrodotoxin (100 nmol/L), showing that CCK was unlikely to have acted via neurotransmitter release. CCK-elicited Ca(2+) signals were inhibited reversibly by caffeine (5-20 mmol/L), indicating involvement of intracellular inositol trisphosphate receptor Ca(2+) release channels. Acetylcholine (50 nmol/L) elicited similar Ca(2+) signals. CONCLUSIONS: CCK at physiologic concentrations in the presence of atropine and tetrodotoxin elicits cytosolic Ca(2+) signaling, activates mitochondrial function, and stimulates enzyme secretion in isolated human pancreatic acinar cells. We conclude that CCK acts directly on acinar cells in the human pancreas.


Subject(s)
Amylases/metabolism , Calcium Signaling , Cholecystokinin/metabolism , Cytosol/metabolism , Exocytosis , Pancreas, Exocrine/metabolism , Acetylcholine/pharmacology , Adenosine Triphosphate/metabolism , Adult , Aged , Aged, 80 and over , Anesthetics, Local/pharmacology , Aniline Compounds , Atropine/pharmacology , Caffeine/pharmacology , Calcium Signaling/drug effects , Cell Polarity , Cholinergic Agents/pharmacology , Exocytosis/drug effects , Female , Fluorescent Dyes , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Microscopy, Confocal , Middle Aged , Mitochondria/metabolism , Muscarinic Antagonists/pharmacology , NAD/metabolism , Pancreas, Exocrine/cytology , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/enzymology , Quinacrine/pharmacology , Sincalide/metabolism , Tetrodotoxin/pharmacology , Time Factors , Xanthenes
20.
Pancreas ; 47(1): 18-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29200128

ABSTRACT

OBJECTIVES: Mitochondrial permeability transition pore inhibition is a promising approach to treat acute pancreatitis (AP). We sought to determine (i) the effects of the mitochondrial permeability transition pore inhibitor 3,5-seco-4-nor-cholestan-5-one oxime-3-ol (TRO40303) on murine and human pancreatic acinar cell (PAC) injury induced by fatty acid ethyl esters (FAEEs) or taurolithocholic acid-3-sulfate and (ii) TRO40303 pharmacokinetics and efficacy in experimental alcoholic AP (FAEE-AP). METHODS: Changes in mitochondrial membrane potential (Δψm), cytosolic Ca ([Ca]c), and cell fate were examined in freshly isolated murine or human PACs by confocal microscopy. TRO40303 pharmacokinetics were assessed in cerulein-induced AP and therapeutic efficacy in FAEE-AP induced with palmitoleic acid and ethanol. Severity of AP was assessed by standard biomarkers and blinded histopathology. RESULTS: TRO40303 prevented loss of Δψm and necrosis induced by 100 µM palmitoleic acid ethyl ester or 500 µM taurolithocholic acid-3-sulfate in murine and human PACs. Pharmacokinetic analysis found TRO40303 accumulated in the pancreas. A single dose of 3 mg/kg TRO40303 significantly reduced serum amylase (P = 0.043), pancreatic trypsin (P = 0.018), and histopathology scores (P = 0.0058) in FAEE-AP. CONCLUSIONS: TRO40303 protects mitochondria and prevents necrotic cell death pathway activation in murine and human PACs, ameliorates the severity of FAEE-AP, and is a candidate drug for human AP.


Subject(s)
Esters/pharmacology , Fatty Acids/pharmacology , Mitochondria/drug effects , Oximes/pharmacology , Pancreatitis, Alcoholic/prevention & control , Secosteroids/pharmacology , Acinar Cells/drug effects , Acinar Cells/metabolism , Acute Disease , Animals , Ceruletide , Esters/metabolism , Fatty Acids/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/metabolism , Necrosis/prevention & control , Oximes/pharmacokinetics , Pancreatitis/chemically induced , Pancreatitis/prevention & control , Pancreatitis, Alcoholic/metabolism , Pancreatitis, Alcoholic/pathology , Secosteroids/pharmacokinetics , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL