Next Article in Journal
Bioactivity of Olive Oil Phenols in Neuroprotection
Next Article in Special Issue
The Role of Omega-3 Fatty Acids in Developmental Psychopathology: A Systematic Review on Early Psychosis, Autism, and ADHD
Previous Article in Journal
Reduced Abundance and Subverted Functions of Proteins in Prion-Like Diseases: Gained Functions Fascinate but Lost Functions Affect Aetiology
Previous Article in Special Issue
High Endogenous Accumulation of ω-3 Polyunsaturated Fatty Acids Protect against Ischemia-Reperfusion Renal Injury through AMPK-Mediated Autophagy in Fat-1 Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Mini-Review on the Effect of Docosahexaenoic Acid (DHA) on Cerulein-Induced and Hypertriglyceridemic Acute Pancreatitis

Department of Food and Nutrition, Brian Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(11), 2239; https://doi.org/10.3390/ijms18112239
Submission received: 20 September 2017 / Revised: 21 October 2017 / Accepted: 23 October 2017 / Published: 25 October 2017
(This article belongs to the Special Issue Omega-3 Fatty Acids in Health and Disease: New Knowledge)

Abstract

:
Acute pancreatitis refers to the sudden inflammation of the pancreas. It is associated with premature activation and release of digestive enzymes into the pancreatic interstitium and systemic circulation, resulting in pancreatic tissue autodigestion and multiple organ dysfunction, as well as with increased cytokine production, ultimately leading to deleterious local and systemic effects. Although mechanisms involved in pathogenesis of acute pancreatitis have not been completely elucidated, oxidative stress is regarded as a major risk factor. In human acute pancreatitis, lipid peroxide levels in pancreatic tissues increase. Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid (C22:6n-3), exerts anti-inflammatory and antioxidant effects on various cells. Previous studies have shown that DHA activates peroxisome proliferator-activated receptor-γ and induces catalase, which inhibits oxidative stress-mediated inflammatory signaling required for cytokine expression in experimental acute pancreatitis using cerulein. Cerulein, a cholecystokinin analog, induces intra-acinar activation of trypsinogen in the pancreas, which results in human acute pancreatitis-like symptoms. Therefore, DHA supplementation may be beneficial for preventing or inhibiting acute pancreatitis development. Since DHA reduces serum triglyceride levels, addition of DHA to lipid-lowering drugs like statins has been investigated to reduce hypertriglyceridemic acute pancreatitis. However, high DHA concentrations increase cytosolic Ca2+, which activates protein kinase C and may induce hyperlipidemic acute pancreatitis. In this review, effect of DHA on cerulein-induced and hypertriglyceridemic acute pancreatitis has been discussed. The relation of high concentration of DHA to hyperlipidemic acute pancreatitis has been included.

1. Introduction

Acute pancreatitis is an inflammatory disease of the pancreas, which may result in multiple organ dysfunction and increased cytokine release [1,2]. About 20–30% patients develop severe forms of this disease, involving local and systemic complications. The mortality rate of acute pancreatitis patients has decreased over the last decade due to improvements in critical care; however, the worldwide incidence of acute pancreatitis is still high [3].
There have been various experimental acute pancreatitis models including ischemia-reperfusion, retrograde administration of sodium taurocholate into the pancreatic duct, and cerulein-induced edematous pancreatitis models [4]. Cerulein-induced pancreatitis is the most well-characterized and widely used experimental model for acute edematous pancreatitis. Supramaximal stimulation of the pancreas with cerulein, a cholecystokinin (CCK) analog, induces intra-acinar activation of trypsinogen in rat pancreas [5]. Moreover, cerulein doses, higher than those that cause maximum pancreatic secretion of amylase and lipase [6,7], dysregulate the production and secretion of digestive enzymes, resulting in increasing their levels in serum, along with causing cytoplasmic vacuolization, death of acinar cells, edema formation, and infiltration of inflammatory cells into the pancreas [8,9]. Ederle et al. [10] reported that the mean CCK level in pancreatitis patients (260.3 ± 300.8 pg/mL) was significantly higher than that in the control subjects (56.6 ± 61.7 pg/mL). Shirohara and Otsuki demonstrated that plasma CCK levels increased during acute pancreatitis, including gallstone pancreatitis, since edema of the bile duct causes transient disturbances in bile flow into the duodenum. They suggested the usefulness of CCK receptor antagonists for the treatment of acute pancreatitis [11].
Ghrelin (GHRL) is an endogenous ligand for the growth hormone secretagogue receptor (GHS-R). A recent study showed that cerulein inhibited GHS-R and GHRL expression in the rat pancreatic acinar cells. GHRL stimulates its own expression and expression of its receptor in isolated pancreatic acinar cells and AR42J cells on the positive feedback pathway. This mechanism may explain the pancreatoprotective effect of GHRL in the process of acute pancreatitis [12]. Obestatin, 23-amino-acid-peptide, colocalized with GHRL in human pancreas, decreased serum level of proinflammatory IL-1β and improved pancreatic blood flow in rats with cerulein-induced acute pancreatitis [13] as well as ischemia/reperfusion-induced acute pancreatitis of rats [14]. These studies suggest that decreased GHRL may contribute to the development of acute pancreatitis.
Although the mechanisms involved in the pathogenesis of acute pancreatitis are not completely understood, oxidative stress is regarded as a major risk factor [15,16,17]. It has been reported that reactive oxygen species (ROS) are important mediators for the initiation and development of pancreatitis [18]. Cerulein-induced activation of nuclear factor-κB (NF-κB) and cytokine expression is potentially mediated by ROS that are produced by NADPH oxidase in the pancreatic acinar cells [19]. ROS generation induced by cerulein is mainly responsible for cytokine production in acinar cells via direct activation of inflammatory signaling, involving protein kinase C-δ (PKC-δ), NF-κB, activator protein-1 (AP-1), and janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) [19,20,21].
Previously, we demonstrated that the omega-3 polyunsaturated fatty acids (PUFAs) may prevent oxidative stress-induced inflammation in the pancreas [22]. Studies involving human subjects also indicated that the use of enteral formula enriched with omega-3 PUFAs for the treatment of acute pancreatitis may be beneficial, evident by the shortened time for jejunal feeding and hospital stay [23]. Moreover, it also reduced the histological severity of acute pancreatitis [24,25,26]. Parenteral therapy with omega-3 PUFAs decreased histopathologic severity in acute necrotizing pancreatitis via early inhibition of prostaglandin synthesis and reduction of lipid peroxidation [27].
DHA shows antioxidant and anti-inflammatory effects on various cells and tissues. Therefore, the beneficial effects of DHA for prevention and treatment of various diseases have been extensively investigated [28]. Understanding the underlying mechanism responsible for the inhibition of acute pancreatitis by DHA could help in the identification of novel therapeutic treatment options, thereby preventing undesirable complications or fatal outcomes.
Hypertriglyceridemia is associated with acute pancreatitis. A range of 3–38% of patients with acute pancreatitis is related to hypertriglyceridemic pancreatitis [29,30,31,32,33,34]. Both primary (genetic) and secondary disorders of lipoprotein metabolism (e.g., uncontrolled diabetes, alcoholism, medications, and pregnancy) may be associated with hypertriglyceridemic pancreatitis [35]. Perfusion of the pancreas with fatty acid (FA) has induced pancreatic edema [36] and the activation of trypsinogen to initiate acute pancreatitis in mice [37]. Hypertriglyceridemia (HTG) contributes to and accelerates the inflammatory cascade and pancreatic tissue damage in rats [38]. Furthermore, large amounts of FA inhibited mitochondrial complexes I and V and decreased ATP levels in acinar cells, which induced mitochondrial toxicity in pancreatic acinar cells [39]. Ca2+ signals are necessary for normal acinar cell secretory function [40]. However, abnormal, prolonged elevation of cytosolic Ca2+ is a critical trigger of pancreatitis via PKC activation [40]. Therefore, high levels of triglyceride (TG), FA, and Ca2+ may trigger pancreatic inflammation.
Since DHA reduces serum TG levels, it may prevent acute pancreatitis associated with HTG and FFA. On the other hand, a high concentration of DHA induces Ca2+-mediated activation of protein kinase C (PKC) isoforms (PKC-α, PKC-δ, PKC-ε, and PKC-ζ) and zymogen activation in the pancreatic acinar cells, which may promote acute pancreatitis [41].
Here, we review the antioxidant and anti-inflammatory effects of DHA on cerulein-induced experimental acute pancreatitis. In addition, the controversy associated with the effect of DHA on hypertriglyceridemic and hyperlipidemic acute pancreatitis has also been discussed.

2. Oxidative Stress and Inflammatory Signaling in Cerulein-Induced Acute Pancreatitis

Oxidative stress is a major risk factor associated with the pathogenesis of human acute pancreatitis. It has been demonstrated that pancreatic oxidative stress occurs during early stages of ROS induction [42]. Once produced, ROS can act as molecular triggers for inducing pancreatitis. They may attack biological membranes directly and/or trigger the accumulation of neutrophils and aid in their adherence to the capillary wall [43,44]. Therefore, it is likely that ROS play a central role in pancreatic inflammation and the development of additional pancreatic complications. In human acute pancreatitis, lipid peroxide levels in the bile and pancreatic tissues increase, while antioxidant vitamins decrease [45]. During pancreatitis, the increase in ROS may be related to reduced levels and activities of antioxidant enzymes, including superoxide dismutase (SOD) and catalase [46]. Depletion of pancreatic glutathione (GSH) occurs in the early phases of acute pancreatitis [47] and influences disease severity in rat models [48]. The activities of several antioxidant enzymes, including glutathione peroxidase, SOD, and catalase, and levels of antioxidant vitamins decrease in patients with human acute pancreatitis [49,50]. Serum lipid peroxide levels increased in human pancreatitis patients [51,52].
Evidences suggest that proinflammatory cytokines, such as IL-1β, TNF-α, and IL-6, act as mediators of acute pancreatitis [53,54,55]. IL-6 is a proinflammatory cytokine associated with acute phase responses during inflammation [56]. Elevated levels of IL-6 have been observed in patients with acute pancreatitis and are determinants of disease severity [57].
TNF-α is produced in pancreatic acinar cells in experimental acute pancreatitis model. It is an activator of immune cells and regulates the synthesis of other pro-inflammatory cytokines [58]. Pretreatment with GHRL in rats with intact sensory nerves reduced the pancreatitis-induced increase in plasma concentration of TNF-α [59]. GHRL induced anti-inflammatory cytokine IL-4, which suppressed IL-1β expression in cerulein-induced acute pancreatitis [59].
IL-1β is produced as a pro-enzyme and requires proteolytic cleavage by IL-1 converting enzyme (ICE) or by neutrophil proteases to develop maximal activity. IL-1β and ICE are expressed at low levels in mouse pancreas, but increase rapidly on cerulein stimulation or on a choline deficient, ethionine-supplemented diet [60]. Therefore, inhibition of IL-1β expression may prevent the development of acute pancreatitis [61].
Cerulein induces high ROS production and activates oxidation-sensitive NF-κB, thereby inducing high cytokine expression in freshly isolated pancreatic acinar cells (without inflammatory cells) in vitro [62]. NF-κB is known to regulate the expression of inflammatory cytokines such as IL-1β, TNF-α, and IL-6, which induce the acute and edematous form of pancreatitis. PKC-δ was shown to activate NF-κB in a mouse model of cerulein-induced acute pancreatitis. Activation of PKC-δ is necessary for NF-κB activation, which is responsible for the pathogenesis of acute pancreatitis [20]. Therefore, reducing ROS may prevent ROS-mediated activation of inflammatory signaling including NF-κB and expression of inflammatory cytokines in pancreas.

3. Antioxidant and Anti-Inflammatory Effects of DHA on Cerulein-Induced Acute Pancreatitis

DHA, at 20 and 50 μM, reduced ROS levels, leading to the inhibition of the JAK 2/STAT3 pathway in cerulein-stimulated pancreatic acinar cells [63]. In response to oxidative stress, activation of the JAK2/STAT3 pathway induces the expression of inflammatory cytokines [64,65]. Moreover, in pancreatic AR42J cells treated with cerulein, DHA acts as an agonist of peroxisome proliferator-activated receptor-γ (PPAR-γ). PPAR-γ induces the expression of glutathione and thioredoxin antioxidant systems in the murine hippocampal HT22 cells [66]. PPAR-γ inactivates STAT3 by directly interacting with STAT3 in cerulein-stimulated pancreatic acinar cells [67]. Since STAT3 regulate cytokine expression, PPAR-γ induction by DHA may be beneficial for inhibiting inflammation in pancreas. DHA upregulates the expression of antioxidant enzymes, including catalase, glutathione peroxidase, and manganese superoxide dismutase (SOD2), in the murine hippocampal HT22 cells and in rats during post-natal development [66,68,69]. In the cerulein-stimulated AR42J cells, DHA induces catalase expression [63]. Recent studies reported that PPAR-γ-specific agonists upregulate the expression of copper-zinc superoxide dismutase (SOD1) in primary endothelial cells [70], and SOD2 and glutathione peroxidase in the skeletal muscle cells, heart, and neurons [71]. Therefore, DHA may induce the expression of SOD1, SOD2, catalase, and glutathione peroxidase in cerulein-stimulated AR42J cells. Since oxidative stress is involved in the pathogenesis of acute pancreatitis, DHA may prevent and/or inhibit the development of acute pancreatitis by suppressing the inflammatory signaling pathways, such as JAK 2/STAT 3, in the pancreatic tissues.
It has been reported that cerulein upregulates IL-6 by activating NADPH oxidase to produce excess ROS in the pancreatic acinar cells [72]. Moreover, cerulein induces the nuclear transcription factor, activator protein-1 (AP-1), which regulates inflammatory cytokine gene expression [73]. DHA inhibited AP-1 activation and mRNA expression of IL-1β and IL-6 in the pancreatic acinar AR42J cells stimulated with cerulein [22]. In cerulein-treated rats, pretreatment with DHA (intraperitoneal injection, 13 mg/kg body) suppressed pancreatic edema formation, and increased lipid peroxidation, myeloperoxidase activity, and NF-κB activation in pancreatic tissues. DHA inhibited PKC-δ activation and increased the expression of antioxidant enzyme SOD1 in pancreatic tissues of cerulein-treated rats [74]. These results suggested that DHA may be beneficial for preventing the development of pancreatitis by suppressing the activation of PKC-δ and NF-κB, and inhibiting the expression of inflammatory cytokines.
Taken together, DHA activates PPAR-γ and induces the expression of PPAR-γ-target gene, SOD1 and catalase, thereby inhibiting ROS-mediated activation of PKC-δ, NF-κB, AP-1, JAK2/STAT3, and inflammatory cytokine expression, in the in vitro cerulein-stimulated pancreatic acinar cells and in vivo rat models (Figure 1).

4. Hypertriglyceridemia and Acute Pancreatitis

Hypertriglyceridemia with serum TG levels ≥500 mg/dL (≥5.65 mmol/L), increases the risk of acute pancreatitis [75]. Therefore, lowering TG levels reduces the risk of pancreatitis. Both genetic and secondary disorders of lipoprotein metabolism are associated with hypertriglyceridemic pancreatitis. Pancreatic lipase-induced hydrolysis of TG and the subsequent formation of free fatty acids (FAs) trigger inflammation [35]. Earlier, Havel [76] proposed that, when high concentrations of FAs are present (concentrations exceed the binding capacity of plasma albumin), the FA molecules self-aggregate to form micellar structures with detergent-like properties. These FA micelles initially attack platelets and the vascular endothelium. Finally, they attack the acinar cells, thereby resulting in ischemia and pancreatic injury.
In the pathogenesis of acute pancreatitis, endothelial dysregulation, vascular leakage, and coagulation activation have been shown [77]. These pathologic events may be related to FA-induced vascular damage. Therefore, angiopoietin-2, which is associated with endothelial dysfunction, has been recently proposed as a marker of severity in acute pancreatitis [78]. Anticoagulant such as acenocoumarol has been used for treating experimental ischemia/reperfusion-induced acute pancreatitis in rats [79,80] and cerulein-induced pancreatitis rats [81].

5. Effect of DHA on Hypertriglyceridemic Acute Pancreatitis

Pharmacological interventions for lowering TG levels include statins, fibrates, nicotinic acid, and omega-3 PUFAs [82]. Omega-3 PUFAs reduce hepatic lipogenesis by inhibiting diacylglycerol acetyltransferase and phosphatidic acid phosphohydrolase, involved in TG synthesis, thereby decreasing TG production in the liver [83]. Omega-3 PUFAs inhibit hepatic FA synthesis by suppressing sterol regulatory element-binding protein (SREBP) 1c, a transcription factor that plays a key role in lipogenesis [84,85]. In addition, omega-3 PUFAs increase lipoprotein lipase activity in the extrahepatic tissues, including the adipose, heart, and skeletal muscles, and increase β-oxidation of FAs in the liver and skeletal muscles, thereby contributing to the reduction of FA delivery to the liver and reducing plasma TG levels [86]. Since FA aggregates induce pancreatic inflammation and injury, the TG-lowering effect of DHA may inhibit development of hypertriglyceridemic acute pancreatitis (Figure 2). At pharmacological doses (3–4 g/day), omega-3 PUFAs acids significantly reduce TG levels in hypertriglyceridemia patients [87]. Compared to EPA, DHA demonstrated a higher efficacy for the reduction of TG (DHA, −8 to −43.7%; EPA, +1.8 to −34.9%) [88,89]. Omega-3 PUFAs are effective in reducing the levels of TG and other lipids in hypertriglyceridemic patients treated with statins [90]. Currently, omega-3 PUFA-based formulations are being evaluated to ascertain whether the addition of omega-3 PUFAs to statin prevents the development of acute pancreatitis in patients with hypertriglyceridemia.

6. Effect of High Concentration of DHA on Hyperlipidemic Acute Pancreatitis

A high concentration (1 mM) of DHA induced a persistent increase in cytosolic Ca2+ concentration and upregulated the expression of PKC isoforms (PKC-α, PKC-δ, PKC-ε, and PKC-ζ) in mouse pancreatic acinar cells [41]. This increase in Ca2+ level and activation of PKC-α, PKC-δ, and PKC-ζ was similar to that observed using supramaximal concentrations of CCK in isolated pancreatic acinar cells [91]. Petersen and Sutton [92] reported that sustained elevation of Ca2+ concentration causes abnormal enzyme activation, vacuolization, and necrosis in acinar cells. In pancreatic acinar cells, PKC isoforms, namely, PKC-α, PKC-δ, PKC-ε, and PKC-ζ, have been identified [93]. PKC-δ incudes the premature activation of zymogen and NF-κB, and modulates the expression of inflammatory molecules in pancreatic acinar cells during experimental pancreatitis [94,95,96,97]. Therefore, high concentrations of DHA may promote the development of acute pancreatitis via activation of the PKC isoforms (Figure 2). Low concentrations of DHA (0.1 mM) had no effect of Ca2+ concentration in acinar cells [41]. These results support the proposed mechanism of pathogenesis of hyperlipidemic acute pancreatitis. Therefore, high DHA doses should be avoided for hyperlipidemia patients, for preventing the development of acute pancreatitis.

7. Other Omega-3 Fatty Acids and Pancreatitis

Cell membrane fatty acids are precursors of lipid mediators such as eicosanoids (prostaglandins: PG; thromboxanes: TX; leukotrienes: LT) [98]. Lipid mediators produced by DHA and eicosapentaenoic acid (EPA, C20:5n3) show anti-inflammatory properties [99]. EPA and DHA inhibit production of arachidonic acid-derived inflammatory eicosanoids [100]. Interestingly, the amounts of omega-3 PUFAs DHA, EPA, and docosapentaenoic acid (DPA, C22:5n3) decreased in the erythrocyte membrane phospholipids of acute pancreatitis patients compared to control subjects [101]. These studies suggest that reduction of omega-3 PUFAs may be related to the development of acute pancreatitis. In addition, the mixture of omega-3 PUFAs decreased Toll-like receptor 4, NF-κB p56, and inflammatory cytokine expression in the pancreas in the severe acute pancreatitis model of rats received retrograde infusion of sodium taurocholate into the pancreatic duct [102]. This study suggest that omega-3 PUFAs inhibit the TLR4/NF-κBp56 signaling pathway to suppress inflammatory cytokines in pancreas. Parenteral infusion of fish-oil-based lipid emulsion reduced systemic inflammatory cytokines and inflammatory eicosanoid levels in sodium taurocholate-induced acute pancreatitis [103].
Serum TG levels increase during pregnancy, which may elicit acute pancreatitis. Therefore, it is important to abrogate the rapid rise of TG levels in pregnancy [104]. Oral EPA prevented rapid increase in serum TG, suggesting that EPA administration may be a useful treatment for hypertriglyceridemic acute pancreatitis during pregnancy.

8. Conclusions and Future Directions

DHA treatment inhibited inflammatory mediators by reducing ROS generation and inflammatory cytokine expression in cerulein-induced experimental model for acute pancreatitis. DHA induces the expression of PPAR-γ-target gene, SOD1, and catalase, thereby inhibiting ROS-mediated activation of inflammatory signaling (PKC-δ, NF-κB, AP-1, JAK2/STAT3) and inflammatory cytokine expression in cerulein-stimulated pancreatic acinar cells and rat models. DHA-induced activation of PPAR-γ and catalase expression may be responsible for the anti-inflammatory effects of DHA in cerulein-induced acute pancreatitis. In addition, DHA inhibits FA and TG synthesis in liver. DHA activates LPL activity in extrahepatic tissues and FA oxidation in liver. Therefore, DHA reduces plasma levels of TG and FFA released from TG. Since FA aggregates induces pancreatic damage as well as vascular endothelial dysfunction, TG-lowering effect of DHA may inhibit hypertriglyceridemic acute pancreatitis. Combination of DHA and lipid-lowering drugs like statins may reduce the development of hypertriglyceridemic acute pancreatitis by decreasing TG levels. On the other hand, a high concentration of DHA may promote PKC-mediated inflammation, since PKC activates NF-κB and zymogen, which may cause hyperlipidemic acute pancreatitis. An understanding of how DHA interferes with inflammatory mediators is important for determining the effects of DHA when it is used in combination with traditional therapy for inhibiting inflammation in the pancreatic tissues.

Acknowledgments

This study was supported by a grant from the National Research Foundation (NRF) of Korea, which is funded by the Korean Government (NRF-2015 R1A2A2A01004855).

Author Contributions

All authors contributed substantially to the conception of this review. Yoo Kyung Jeong drafted the manuscript. Hyeyoung Kim edited the manuscript. All authors approved the manuscript in its current form.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AP-1activator protein-1
CCKcholecystokinin
DHAdocosahexaenoic acid
DPAdocosapentaenoic acid
EPAeicosapentaenoic acid
FAfatty acid
GSHglutathione
GHRLghrelin
GHS-Rgrowth hormone secretagogue receptor
ICEIL-1 converting enzyme
JAKjanus kinase
LPLlipoprotein lipase
LTleukotrienes
NF-κBnuclear factor-κB
PGprostaglandins
PKCprotein kinase C
PPAR-γperoxisome proliferator-activated receptor-γ
PUFAspolyunsaturated acids
ROSreactive oxygen species
SODsuperoxide dismutase
SREBPsterol regulatory element-binding protein
STAT3signal transducer and activator of transcription 3
TGtriglyceride
TXthromboxanes

References

  1. Bhatia, M.; Wong, F.L.; Cao, Y.; Lau, H.Y.; Huang, J.; Puneet, P.; Chevali, L. Pathophysiology of acute pancreatitis. Pancreatology 2005, 5, 132–144. [Google Scholar] [CrossRef] [PubMed]
  2. Frossard, J.L.; Hadengue, A.; Pastor, C.M. New serum markers for the detection of severe acute pancreatitis in humans. Am. J. Respir. Crit. Care Med. 2001, 164, 162–170. [Google Scholar] [CrossRef] [PubMed]
  3. Heinrich, S.; Schäfer, M.; Rousson, V.; Clavien, P.A. Evidence-based treatment of acute pancreatitis: A look at established paradigms. Ann. Surg. 2006, 243, 154–168. [Google Scholar] [CrossRef] [PubMed]
  4. Ceranowicz, P.; Cieszkowski, J.; Warzecha, Z.; Dembiński, A. Experimental models of acute pancreatitis. Postepy Higieny I Medycyny Doswiadczalnej 2015, 69, 264–269. [Google Scholar] [CrossRef] [PubMed]
  5. Hofbauer, B.; Saluja, A.K.; Lerch, M.M.; Bhagat, L.; Bhatia, M.; Lee, H.S.; Frossard, J.L.; Adler, G.; Steer, M.L. Intra-acinar cell activation of trypsinogen during caerulein-induced pancreatitis in rats. Am. J. Physiol. 1998, 275, G352–G362. [Google Scholar] [PubMed]
  6. Jensen, R.T.; Wank, S.A.; Rowley, W.H.; Sato, S.; Gardner, J.D. Interaction of CCK with pancreatic acinar cells. Trends Pharmacol. Sci. 1989, 10, 418–423. [Google Scholar] [CrossRef]
  7. Sato, S.; Stark, H.A.; Martinez, J.; Beaven, M.A.; Jensen, R.T.; Gardner, J.D. Receptor occupation, calcium mobilization, and amylase release in pancreatic acini: Effect of CCK-JMV-180. Am. J. Physiol. 1989, 257, G202–G209. [Google Scholar] [PubMed]
  8. Willemer, S.; Elsasser, H.P.; Adler, G. Hormone-induced pancreatitis. Eur. Surg. Res. 1992, 24, S29–S49. [Google Scholar] [CrossRef]
  9. Lerch, M.M.; Adler, G. Experiemental animal models of acute pancreatitis. Int. J. Pancreatol. 1994, 15, 159–170. [Google Scholar] [PubMed]
  10. Ederle, A.; Vantini, I.; Harvey, R.F.; Cavallini, G.; Piubello, W.; Benini, L.; Scuro, L.A. Fasting serum cholecystokinin immunoreactivity in chronic relapsing pancreatitis. Int. J. Clin. Lab. Res. 1978, 8, 199–206. [Google Scholar]
  11. Shirohara, H.; Otsuki, M. Plasma cholecystokinin levels in acute pancreatitis. Pancreas 1997, 14, 249–254. [Google Scholar] [CrossRef] [PubMed]
  12. Bonior, J.; Ceranowicz, P.; Gajdosz, R.; Kuśnierz-Cabala, B.; Pierzchalski, P.; Warzecha, Z.; Dembiński, A.; Pędziwiatr, M.; Kot, M.; Leja-Szpak, A.; et al. Molecular ghrelin system in the pancreatic acinar cells: The role of the polypeptide, caerulein and sensory nerves. Int. J. Mol. Sci. 2017, 18, 929. [Google Scholar] [CrossRef] [PubMed]
  13. Bukowczan, J.; Cieszkowski, J.; Warzecha, Z.; Ceranowicz, P.; Kusnierz-Cabala, B.; Tomaszewska, R.; Dembinski, A. Therapeutic effect of obestatin in the course of cerulein-induced acute pancreatitis. Pancreas 2016, 45, 700–706. [Google Scholar] [CrossRef] [PubMed]
  14. Bukowczan, J.; Warzecha, Z.; Ceranowicz, P.; Kuśnierz-Cabala, B.; Tomaszewska, R. Obestatin accelerates the recovery in the course of ischemia/reperfusion-induced acute pancreatitis in rats. PLoS ONE 2015, 10, e0134380. [Google Scholar] [CrossRef] [PubMed]
  15. Gorelick, F.S.; Thrower, E. The acinar cell and early pancreatitis responses. Clin. Gastroenterol. Hepatol. 2009, 7, S10–S14. [Google Scholar] [CrossRef] [PubMed]
  16. Chakraborty, M.; Hickey, A.J.; Petrov, M.S.; Macdonald, J.R.; Thompson, N.; Newby, L.; Sim, D.; Windsor, J.A.; Phillips, A.R. Mitochondrial dysfunction in peripheral blood mononuclear cells in early experimental and clinical acute pancreatitis. Pancreatology 2016, 16, 739–747. [Google Scholar] [CrossRef] [PubMed]
  17. Saluja, A.; Steer, M. Pathophysiology of pancreatitis. Digestion 1999, 60, 27–33. [Google Scholar] [CrossRef] [PubMed]
  18. Leung, P.S.; Chan, Y.C. Role of oxidative stress in pancreatic inflammation. Antioxid. Redox Signal. 2009, 11, 135–165. [Google Scholar] [CrossRef] [PubMed]
  19. Kim, H. Cerulien pancreatitis: Oxidative stress, inflammation, and apoptosis. Gut Liver 2008, 2, 74–80. [Google Scholar] [CrossRef] [PubMed]
  20. Ramnath, R.D.; Sun, J.; Bhatia, M. PKC δ mediates pro-inflammatory responses in a mouse model of caerulein-induced acute pancreatitis. J. Mol. Med. (Berl.) 2010, 88, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
  21. Ju, K.D.; Lim, J.W.; Kim, K.H.; Kim, H. Potential role of NADPH oxidase-mediated activation of Jak2/Stat3 and mitogen-activated protein kinases and expression of TGF-β1 in the pathophysiology of acute pancreatitis. Inflamm. Res. 2011, 60, 791–800. [Google Scholar] [CrossRef] [PubMed]
  22. Park, K.S.; Lim, J.W.; Kim, H. Inhibitory mechanism of omega-3 fatty acids in pancreatic inflammation and apoptosis. Ann. N. Y. Acad. Sci. 2009, 1171, 421–427. [Google Scholar] [CrossRef] [PubMed]
  23. Lasztity, N.; Hamvas, J.; Biró, L.; Németh, E.; Marosvölgyi, T.; Decsi, T.; Pap, A.; Antal, M. Effect of enterally administered n-3 polyunsaturated fatty acids in acute pancreatitis—A prospective randomized clinical trial. Clin. Nutr. 2005, 24, 198–205. [Google Scholar] [CrossRef] [PubMed]
  24. Kilian, M.; Heukamp, I.; Gregor, J.I.; Schimke, I.; Kristiansen, G.; Wenger, F.A. Fish oil, but not soy bean or olive oil enriched infusion decreases histopathological severity of acute pancreatitis in rats without affecting eicosanoid synthesis. Inflammation 2011, 34, 597–602. [Google Scholar] [CrossRef] [PubMed]
  25. Weylandt, K.H.; Nadolny, A.; Kahlke, L.; Köhnke, T.; Schmöcker, C.; Wang, J.; Lauwers, G.Y.; Glickman, J.N.; Kang, J.X. Reduction of inflammation and chronic tissue damage by omega-3 fatty acids in fat-1 transgenic mice with pancreatitis. Biochim. Biophys. Acta 2008, 1782, 634–641. [Google Scholar] [CrossRef] [PubMed]
  26. Kilian, M.; Heukamp, I.; Gregor, J.I.; Bretthauer, C.; Walz, M.K.; Jacobi, C.A.; Lochs, H.; Schimke, I.; Guski, H.; Wenger, F.A. n-3, n-6, and n-9 polyunsaturated fatty acids—Which composition in parenteral nutrition decreases severity of acute hemorrhagic necrotizing pancreatitis in rats? Int. J. Colorectal. Dis. 2006, 21, 57–63. [Google Scholar] [CrossRef] [PubMed]
  27. Kilian, M.; Gregor, J.I.; Heukamp, I.; Wagner, C.; Walz, M.K.; Schimke, I.; Kristiansen, G.; Wenger, F.A. Early inhibition of prostaglandin synthesis by n-3 fatty acids determinates histologic severity of necrotizing pancreatitis. Pancreas 2009, 38, 436–441. [Google Scholar] [CrossRef] [PubMed]
  28. Moyad, M.A. An introduction to dietary/supplemental omega-3 fatty acids for general health and prevention: Part II. Urol. Oncol. 2005, 23, 36–48. [Google Scholar] [CrossRef] [PubMed]
  29. Farmer, R.G.; Winkelman, E.I.; Brown, H.B.; Lewis, L.A. Hyperlipoproteinemia and pancreatitis. Am. J. Med. 1973, 54, 161–165. [Google Scholar] [CrossRef]
  30. Yadav, D.; Pitchumoni, C.S. Issues in hyperlipidemic pancreatitis. J. Clin. Gastroenterol. 2003, 36, 54–62. [Google Scholar] [CrossRef] [PubMed]
  31. Cameron, J.L.; Capuzzi, D.M.; Zuidema, G.D.; Margolis, S. Acute pancreatitis with hyperlipemia: The incidence of lipid abnormalities in acute pancreatitis. Ann. Surg. 1973, 177, 483–489. [Google Scholar] [CrossRef] [PubMed]
  32. Dominguez-Muñoz, J.E.; Malfertheiner, P.; Ditschuneit, H.H.; Blanco-Chavez, J.; Uhl, W.; Büchler, M.; Ditschuneit, H. Hyperlipidemia in acute pancreatitis. Relationship with etiology, onset, and severity of the disease. Int. J. Pancreatol. 1991, 10, 261–267. [Google Scholar] [PubMed]
  33. Toskes, P.P. Hyperlipidemic pancreatitis. Gastroenterol. Clin. N. Am. 1990, 19, 783–791. [Google Scholar]
  34. Valdivielso, P.; Ramírez-Bueno, A.; Ewald, N. Current knowledge of hypertriglyceridemic pancreatitis. Eur. J. Intern. Med. 2014, 25, 689–694. [Google Scholar] [CrossRef] [PubMed]
  35. Ewald, N.; Hardt, P.D.; Kloer, H.U. Severe hypertriglyceridemia and pancreatitis: Presentation and management. Curr. Opin. Lipidol. 2009, 20, 497–504. [Google Scholar] [CrossRef] [PubMed]
  36. Saharia, P.; Margolis, S.; Zuidema, G.D.; Cameron, J.L. Acute pancreatitis with hyperlipemia: Studies with an isolated perfused canine pancreas. Surgery 1977, 82, 60–67. [Google Scholar] [PubMed]
  37. Halangk, W.; Lerch, M.M.; Brandt-Nedelev, B.; Roth, W.; Ruthenbuerger, M.; Reinheckel, T.; Domschke, W.; Lippert, H.; Peters, C.; Deussing, J. Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis. J. Clin. Investig. 2000, 106, 773–781. [Google Scholar] [CrossRef] [PubMed]
  38. Kimura, W.; Mössner, J. Role of hypertriglyceridemia in the pathogenesis of experimental acute pancreatitis in rats. Int. J. Pancreatol. 1996, 20, 177–184. [Google Scholar] [CrossRef] [PubMed]
  39. Navina, S.; Acharya, C.; DeLany, J.P.; Orlichenko, L.S.; Baty, C.J.; Shiva, S.S.; Durgampudi, C.; Karlsson, J.M.; Lee, K.; Bae, K.T.; et al. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci. Transl. Med. 2011, 3, 107–110. [Google Scholar] [CrossRef] [PubMed]
  40. Gerasimenko, J.V.; Gerasimenko, O.V.; Petersen, O.H. The role of Ca2+ in the pathophysiology of pancreatitis. J. Physiol. 2014, 592, 269–280. [Google Scholar] [CrossRef] [PubMed]
  41. Chang, Y.T.; Chang, M.C.; Tung, C.C.; Wei, S.C.; Wong, J.M. Distinctive roles of unsaturated and saturated fatty acids in hyperlipidemic pancreatitis. World J. Gastroenterol. 2015, 21, 9534–9543. [Google Scholar] [CrossRef] [PubMed]
  42. Schoenberg, M.H.; Büchler, M.; Gaspar, M.; Stinner, A.; Younes, M.; Melzner, I.; Bültmann, B.; Beger, H.G. Oxygen free radicals in acute pancreatitis of the rat. Gut 1990, 31, 1138–1143. [Google Scholar] [CrossRef] [PubMed]
  43. Petrone, W.F.; English, D.K.; Wong, K.; McCord, J.M. Free radicals and inflammation: Superoxide-dependent activation of a neutrophil chemotactic factor in plasma. Proc. Natl. Acad. Sci. USA 1980, 77, 1159–1163. [Google Scholar] [CrossRef] [PubMed]
  44. Bjork, J.; Arfors, K.E. Oxygen free radicals and leukotriene B4 induced increase in vascular leakage is mediated by polymorphonuclear leukocytes. Agents Actions Suppl. 1981, 11, 63–72. [Google Scholar]
  45. Guyan, P.M.; Uden, S.; Braganza, J.M. Heightened free radical activity in pancreatitis. Free Radic. Biol. Med. 1990, 8, 347–354. [Google Scholar] [CrossRef]
  46. Bopanna, S.; Nayak, B.; Prakash, S.; Mahapatra, S.J.; Garg, P.K. Increased oxidative stress and deficient antioxidant levels may be involved in the pathogenesis of idiopathic recurrent acute pancreatitis. Pancreatology 2017, 17, 529–533. [Google Scholar] [CrossRef] [PubMed]
  47. Gómez-Cambronero, L.; Camps, B.; de La Asunción, J.G.; Cerdá, M.; Pellín, A.; Pallardó, F.V.; Calvete, J.; Sweiry, J.H.; Mann, G.E.; Viña, J.; et al. Pentoxifylline ameliorates cerulein-induced pancreatitis in rats: Role of glutathione and nitric oxide. J. Pharmacol. Exp. Ther. 2000, 293, 670–676. [Google Scholar] [PubMed]
  48. Alsfasser, G.; Gock, M.; Herzog, L.; Gebhard, M.M.; Herfarth, C.; Klar, E.; Schmidt, J. Glutathione depletion with l-buthionine-(S,R)-sulfoximine demonstrates deleterious effects in acute pancreatitis of the rat. Dig. Dis. Sci. 2002, 47, 1793–1799. [Google Scholar] [CrossRef] [PubMed]
  49. Curran, F.J.; Sattar, N.; Talwar, D.; Baxter, J.N.; Imrie, C.W. Relationship of carotenoid and vitamins A and E with the acute inflammatory response in acute pancreatitis. Br. J. Surg. 2000, 87, 301–305. [Google Scholar] [CrossRef] [PubMed]
  50. Cullen, J.J.; Mitros, F.A.; Oberley, L.W. Expression of antioxidant enzymes in diseases of the human pancreas: Another link between chronic pancreatitis and pancreatic cancer. Pancreas 2003, 26, 23–27. [Google Scholar] [CrossRef] [PubMed]
  51. Grigor’eva, I.N.; Romanova, T.I.; Ragino, I.U.I. Lipid peroxidation in patients with acute and chronic pancreatitis. Exp. Clin. Gastroenterol. 2011, 7, 24–27. [Google Scholar]
  52. Singh, N.; Bhardwaj, P.; Pandey, R.M.; Saraya, A. Oxidative stress and antioxidant capacity in patients with chronic pancreatitis with and without diabetes mellitus. Indian J. Gastroenterol. 2012, 31, 226–231. [Google Scholar] [CrossRef] [PubMed]
  53. Viedma, J.A.; Perez-Mateo, M.; Dominguez, J.E.; Carballo, F. Role of interleukin-6 in acute pancreatitis. Comparison with C-reactive protein and phospholipase A. Gut 1992, 33, 1264–1267. [Google Scholar] [CrossRef] [PubMed]
  54. Heath, D.I.; Cruickshank, A.; Gudgeon, M.; Jehanli, A.; Shenkin, A.; Imrie, C.W. Role of interleukin-6 in mediating the acute phase protein response and potential as an early means of severity assessment in acute pancreatitis. Gut 1993, 34, 41–45. [Google Scholar] [CrossRef] [PubMed]
  55. Sameshima, H.; Ikei, S.; Mori, K.; Yamaguchi, Y.; Egami, H.; Misumi, M.; Moriyasu, M.; Ogawa, M. The role of tumor necrosis factor-alpha in the aggravation of cerulein-induced pancreatitis in rats. Int. J. Pancreatol. 1993, 14, 107–115. [Google Scholar] [PubMed]
  56. Schölmerich, J. Interleukins in acute pancreatitis. Scand. J. Gastroenterol. Suppl. 1996, 219, 37–42. [Google Scholar] [CrossRef] [PubMed]
  57. Leser, H.G.; Gross, V.; Scheibenbogen, C.; Heinisch, A.; Salm, R.; Lausen, M.; Rückauer, K.; Andreesen, R.; Farthmann, E.H.; Schölmerich, J. Elevation of serum interleukin-6 concentration precedes acute-phase response and reflects severity in acute pancreatitis. Gastroenterology 1991, 101, 782–785. [Google Scholar] [CrossRef]
  58. Malleo, G.; Mazzon, E.; Siriwardena, A.K.; Cuzzocrea, S. TNF-α as a therapeutic target in acute pancreatitis-lessons from experimental models. Sci. World J. 2007, 7, 431–448. [Google Scholar] [CrossRef] [PubMed]
  59. Bonior, J.; Warzecha, Z.; Ceranowicz, P.; Gajdosz, R.; Pierzchalski, P.; Kot, M.; Leja-Szpak, A.; Nawrot-Porąbka, K.; Link-Lenczowski, P.; Pędziwiatr, M.; et al. Capsaicin-sensitive sensory nerves are necessary for the protective effect of ghrelin in cerulein-induced acute pancreatitis in rats. Int. J. Mol. Sci. 2017, 18, 1402. [Google Scholar] [CrossRef] [PubMed]
  60. Fink, G.W.; Norman, J.G. Specific changes in the pancreatic expression of the interleukin 1 family of genes during experimental acute pancreatitis. Cytokine 1997, 9, 1023–1027. [Google Scholar] [CrossRef] [PubMed]
  61. Romac, J.M.; Shahid, R.A.; Choi, S.S.; Karaca, G.F.; Westphalen, C.B.; Wang, T.C.; Liddle, R.A. Pancreatic secretory trypsin inhibitor I reduces the severity of chronic pancreatitis in mice overexpressing interleukin-1beta in the pancreas. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G535–G541. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, J.H.; Lim, J.W.; Namkung, W.; Kim, K.H. Suppression of cerulein-induced cytokine expression by antioxidants in pancreatic acinar cells. Lab. Investig. 2002, 82, 1359–1368. [Google Scholar] [CrossRef] [PubMed]
  63. Song, E.A.; Lim, J.W.; Kim, H. Docosahexaenoic acid inhibits IL-6 expression via PPAR-gamma mediated expression of catalase in cerulein-stimulated pancreatic acinar cells. Int. J. Biochem. Cell Biol. 2017, 88, 60–68. [Google Scholar] [CrossRef] [PubMed]
  64. Carballo, M.; Conde, M.; El Bekay, R.; Martı́n-Nieto, J.; Camacho, M.J.; Monteseirín, J.; Conde, J.; Bedoya, F.J.; Sobrino, F. Oxidative stress triggers STAT3 tyrosine phosphorylation and nuclear translocation in human lymphocytes. J. Biol. Chem. 1999, 274, 17580–17586. [Google Scholar] [CrossRef] [PubMed]
  65. Darnell, J.E.; Kerr, I.M.; Stark, G.R. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994, 264, 1415–1421. [Google Scholar] [CrossRef] [PubMed]
  66. Casañas-Sánchez, V.; Pérez, J.A.; Fabelo, N.; Herrera-Herrera, A.V.; Fernández, A.V.; Marín, R.; González-Montelongo, M.C.; Díaz, M. Addition of docosahexaenoic acid, but not arachidonic acid, activates glutathione and thioredoxin antioxidant systems in murine hippocampal HT22 cells: Potential implications in neuroprotection. J. Neurochem. 2014, 131, 470–483. [Google Scholar] [CrossRef] [PubMed]
  67. Ju, K.D.; Lim, J.W.; Kim, H. Peroxisome proliferator-activated receptor-gamma inhibits the activation of STAT3 in cerulein-stimulated pancreatic acinar cells. J. Cancer Prev. 2017, 22, 189–194. [Google Scholar] [CrossRef] [PubMed]
  68. Garrel, C.; Alessandri, J.M.; Guesnet, P.; Al-Gubory, K.H. Omega-3 fatty acids enhance mitochondrial superoxide dismutase activity in rat organs during post-natal development. Int. J. Biochem. Cell Biol. 2012, 44, 123–131. [Google Scholar] [CrossRef] [PubMed]
  69. Hossain, M.S.; Hashimoto, M.; Gamoh, S.; Masumura, S. Antioxidative effects of docosahexaenoic acid in the cerebrum versus cerebellum and brainstem of aged hypercholesterolemic rats. J. Neurochem. 1999, 72, 1133–1138. [Google Scholar] [CrossRef] [PubMed]
  70. Inoue, I.; Goto, S.; Matsunaga, T.; Nakajima, T.; Awata, T.; Hokari, S.; Komoda, T.; Katayama, S. The ligands/activators for peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARgamma increase Cu2+, Zn2+-superoxide dismutase and decrease p22phox message expressions in primary endothelial cells. Metabolism 2001, 50, 3–11. [Google Scholar] [CrossRef] [PubMed]
  71. Polvani, S.; Tarocchi, M.; Galli, A. PPARγ and oxidative stress: Con(β) catenating NRF2 and FOXO. PPAR Res. 2012. [Google Scholar] [CrossRef] [PubMed]
  72. Yu, J.H.; Kim, H. Oxidative stress and inflammatory signaling in cerulein pancreatitis. World J. Gastroenterol. 2014, 20, 17324–17329. [Google Scholar] [CrossRef] [PubMed]
  73. Ju, K.D.; Yu, J.H.; Kim, H.; Kim, K.H. Role of mitogen-activated protein kinases, NF-kB and Ap-1 on cerulein-induced IL-8 expression in pancreatic acinar cells. Ann. N. Y. Acad. Sci. 2006, 1090, 368–374. [Google Scholar] [CrossRef] [PubMed]
  74. Jeong, Y.K.; Lee, S.; Lim, J.W.; Kim, H. Docosahexaenoic acid inhibits cerulein-induced acute pancreatitis in rats. Nutrients 2017, 9, 744. [Google Scholar] [CrossRef] [PubMed]
  75. Scherer, J.; Singh, V.P.; Pitchumoni, C.S.; Yadav, D. Issues in hypertriglyceridemic pancreatitis: An update. J. Clin. Gastroenterol. 2014, 48, 195–203. [Google Scholar] [CrossRef] [PubMed]
  76. Havel, R.J. Pathogenesis, differentiation and management of hypertriglyceridemia. Adv. Intern. Med. 1969, 15, 117–145. [Google Scholar] [PubMed]
  77. Dumnicka, P.; Maduzia, D.; Ceranowicz, P.; Olszanecki, R.; Drożdż, R.; Kuśnierz-Cabala, B. The interplay between inflammation, coagulation and endothelial injury in the early phase of acute pancreatitis: Clinical implications. Int. J. Mol. Sci. 2017, 18, 354. [Google Scholar] [CrossRef] [PubMed]
  78. Dumnicka, P.; Kuśnierz-Cabala, B.; Sporek, M.; Mazur-Laskowska, M.; Gil, K.; Kuźniewski, M.; Ceranowicz, P.; Warzecha, Z.; Dembiński, A.; Bonior, J.; et al. Serum concentrations of angiopoietin-2 and soluble fms-like tyrosine kinase 1 (sFlt-1) are associated with coagulopathy among patients with acute pancreatitis. Int. J. Mol. Sci. 2017, 18, 753. [Google Scholar] [CrossRef] [PubMed]
  79. Warzecha, Z.; Sendur, P.; Ceranowicz, P.; Cieszkowski, J.; Dembiński, M.; Sendur, R.; Bonior, J.; Jaworek, J.; Ambroży, T.; Olszanecki, R.; et al. Therapeutic effect of low doses of acenocoumarol in the course of ischemia/reperfusion-induced acute pancreatitis in rats. Int. J. Mol. Sci. 2017, 18, 882. [Google Scholar] [CrossRef] [PubMed]
  80. Warzecha, Z.; Sendur, P.; Ceranowiczm, P.; Dembinskim, M.; Cieszkowskim, J.; Kusnierz-Cabalam, B.; Tomaszewskam, R.; Dembinskim, A. Pretreatment with low doses of acenocoumarol inhibits the development of acute ischemia/reperfusion-induced pancreatitis. J. Physiol. Pharmacol. 2015, 66, 731–740. [Google Scholar] [PubMed]
  81. Warzecha, Z.; Sendur, P.; Ceranowicz, P.; Dembiński, M.; Cieszkowski, J.; Kuśnierz-Cabala, B.; Olszanecki, R.; Tomaszewska, R.; Ambroży, T.; Dembiński, A. Protective effect of pretreatment with acenocoumarol in cerulein-induced acute pancreatitis. Int. J. Mol. Sci. 2016, 17, 1709. [Google Scholar] [CrossRef] [PubMed]
  82. Catapano, A.L.; Reiner, Z.; De Backer, G.; Graham, I.; Taskinen, M.R.; Wiklund, O.; Agewall, S.; Alegria, E.; Chapman, M.; Durrington, P.; et al. ESC/EAS guidelines for the management of dyslipidaemias the task force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and the European Atherosclerosis Society (EAS). Atherosclerosis 2011, 217, 3–46. [Google Scholar] [CrossRef] [PubMed]
  83. Al-Shurbaji, A.; Larsson-Backström, C.; Berglund, L.; Eggertsen, G.; Björkhem, I. Effect of n-3 fatty acids on the key enzymes involved in cholesterol and triglyceride turnover in rat liver. Lipids 1991, 26, 385–389. [Google Scholar] [CrossRef] [PubMed]
  84. Sampath, H.; Ntambi, J.M. Polyunsaturated fatty acid regulation of gene expression. Nutr. Rev. 2004, 62, 333–339. [Google Scholar] [CrossRef] [PubMed]
  85. Jump, D.B.; Botolin, D.; Wang, Y.; Xu, J.; Christian, B.; Demeure, O. Fatty acid regulation of hepatic gene transcription. J. Nutr. 2005, 135, 2503–2506. [Google Scholar] [PubMed]
  86. Shearer, G.C.; Savinova, O.V.; Harris, W.S. Fish oil—How does it reduce plasma triglycerides? Biochim. Biophys. Acta 2012, 1821, 843–851. [Google Scholar] [CrossRef] [PubMed]
  87. Jacobson, T.A. Role of n-3 fatty acids in the treatment of hypertriglyceridemia and cardiovascular disease. Am. J. Clin. Nutr. 2008, 87, 1981S–1990S. [Google Scholar] [PubMed]
  88. Jacobson, T.A.; Glickstein, S.B.; Rowe, J.D.; Soni, P.N. Effects of eicosapentaenoic acid and docosahexaenoic acid on low-density lipoprotein cholesterol and other lipids: A review. J. Clin. Lipidol. 2012, 6, 5–18. [Google Scholar] [CrossRef] [PubMed]
  89. Wei, M.Y.; Jacobson, T.A. Effects of eicosapentaenoic acid versus docosahexaenoic acid on serum lipids: A systematic review and meta-analysis. Curr. Atheroscler. Rep. 2011, 13, 474–483. [Google Scholar] [CrossRef] [PubMed]
  90. Pirillo, A.; Catapano, A.L. Update on the management of severe hypertriglyceridemia—Focus on free fatty acid forms of omega-3. Drug Des. Dev. Ther. 2015, 9, 2129–2137. [Google Scholar]
  91. Gorelick, F.; Pandol, S.; Thrower, E. Protein kinase C in the pancreatic acinar cell. J. Gastroenterol. Hepatol. 2008, 23 (Suppl. 1), S37–S41. [Google Scholar] [CrossRef] [PubMed]
  92. Petersen, O.H.; Sutton, R. Ca2+ signalling and pancreatitis: Effects of alcohol, bile and coffee. Trends Pharmacol. Sci. 2006, 27, 113–120. [Google Scholar] [CrossRef] [PubMed]
  93. Bastani, B.; Yang, L.; Baldassare, J.J.; Pollo, D.A.; Gardner, J.D. Cellular distribution of isoforms of protein kinase C (PKC) in pancreatic acini. Biochim. Biophys. Acta 1995, 1269, 307–315. [Google Scholar] [CrossRef]
  94. Satoh, A.; Gukovskaya, A.S.; Nieto, J.M.; Cheng, J.H.; Gukovsky, I.; Reeve, J.R.; Shimosegawa, T.; Pandol, S.J. PKC-delta and -epsilon regulate NF-kappaB activation induced by cholecystokinin and TNF-alpha in pancreatic acinar cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G582–G591. [Google Scholar] [CrossRef] [PubMed]
  95. Thrower, E.C.; Osgood, S.; Shugrue, C.A.; Kolodecik, T.R.; Chaudhuri, A.M.; Reeve, J.R.; Pandol, S.J.; Gorelick, F.S. The novel protein kinase C isoforms -delta and-epsilon modulate caerulein-induced zymogen activation in pancreatic acinar cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G1344–G1353. [Google Scholar] [CrossRef] [PubMed]
  96. Li, C.; Chen, X.; Williams, J.A. Regulation of CCK-induced amylase release by PKC-delta in rat pancreatic acinar cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G764–G771. [Google Scholar] [CrossRef] [PubMed]
  97. Ramnath, R.D.; Sun, J.; Adhikari, S.; Zhi, L.; Bhatia, M. Role of PKC-delta on substance P-induced chemokine synthesis in pancreatic acinar cells. Am. J. Physiol. Cell Physiol. 2008, 294, C683–C692. [Google Scholar] [CrossRef] [PubMed]
  98. Kremmyda, L.S.; Tvrzicka, E.; Stankova, B.; Zak, A. Fatty acids as biocompounds: Their role in human metabolism, health and disease: A review. Part 2: Fatty acid physiological roles and applications in human health and disease. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2011, 155, 195–218. [Google Scholar] [CrossRef] [PubMed]
  99. Bannenberg, G.; Serhan, C.N. Specialized pro-resolving lipid mediators in the inflammatory response: An update. Biochim. Biophys. Acta 2010, 1801, 1260–1273. [Google Scholar] [CrossRef] [PubMed]
  100. Calder, P.C. Polyunsaturated fatty acids and inflammatory processes: New twists in an old tale. Biochimie 2009, 91, 791–795. [Google Scholar] [CrossRef] [PubMed]
  101. Kuliaviene, I.; Gulbinas, A.; Cremers, J.; Pundzius, J.; Kupcinskas, L.; Dambrauskas, Z.; Jansen, E. Fatty acids of erythrocyte membrane in acute pancreatitis patients. World J. Gastroenterol. 2013, 19, 5678–5684. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, B.; Xu, X.B.; Jin, X.X.; Wu, X.W.; Li, M.L.; Guo, M.X.; Zhang, X.H. Effects of ω-3 Fatty Acids on toll-like receptor 4 and nuclear factor κB p56 in the pancreas of rats with severe acute pancreatitis. Pancreas 2017, 46, 1267–1274. [Google Scholar] [CrossRef] [PubMed]
  103. Garla, P.; Garib, R.; Torrinhas, R.S.; Machado, M.C.; Calder, P.C.; Waitzberg, D.L. Effect of parenteral infusion of fish oil-based lipid emulsion on systemic inflammatory cytokines and lung eicosanoid levels in experimental acute pancreatitis. Clin. Nutr. 2017, 36, 302–308. [Google Scholar] [CrossRef] [PubMed]
  104. Nakao, J.; Ohba, T.; Takaishi, K.; Katabuchi, H. Omega-3 fatty acids for the treatment of hypertriglyceridemia during the second trimester. Nutrition 2015, 31, 409–412. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposed mechanisms of action of docosahexaenoic acid (DHA) in cerulein-induce acute pancreatitis. Cerulein induced the activation of NADPH oxidase, which produces large amounts of reactive oxygen species (ROS) in pancreatic acinar cells. ROS activate protein kinase C-δ (PKC-δ), which activates nuclear factor-κB (NF-κB). ROS also directly activate NF-κB, activator protein-1 (AP-1), janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3), and inflammatory cytokine expression in the cerulein-stimulated pancreatic acinar cells and rat models. The inflammatory events result in development of acute edematous pancreatitis. DHA induces activation of peroxisome proliferator-activated receptor-γ (PPAR-γ) and expression of the PPARγ-target gene, SOD1, and catalase. Since SOD1 and catalase scavenge ROS, DHA inhibits the ROS-mediated activation of inflammatory signaling (PKC-δ, NF-κB, AP-1, JAK2/STAT3) and inflammatory cytokine expression in cerulein-stimulated pancreatic acinar cells and animal models. Antioxidant and anti-inflammatory effects of DHA may be responsible for preventing the development of acute pancreatitis. The bars represent inhibition, while the arrows represent stimulation.
Figure 1. Proposed mechanisms of action of docosahexaenoic acid (DHA) in cerulein-induce acute pancreatitis. Cerulein induced the activation of NADPH oxidase, which produces large amounts of reactive oxygen species (ROS) in pancreatic acinar cells. ROS activate protein kinase C-δ (PKC-δ), which activates nuclear factor-κB (NF-κB). ROS also directly activate NF-κB, activator protein-1 (AP-1), janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3), and inflammatory cytokine expression in the cerulein-stimulated pancreatic acinar cells and rat models. The inflammatory events result in development of acute edematous pancreatitis. DHA induces activation of peroxisome proliferator-activated receptor-γ (PPAR-γ) and expression of the PPARγ-target gene, SOD1, and catalase. Since SOD1 and catalase scavenge ROS, DHA inhibits the ROS-mediated activation of inflammatory signaling (PKC-δ, NF-κB, AP-1, JAK2/STAT3) and inflammatory cytokine expression in cerulein-stimulated pancreatic acinar cells and animal models. Antioxidant and anti-inflammatory effects of DHA may be responsible for preventing the development of acute pancreatitis. The bars represent inhibition, while the arrows represent stimulation.
Ijms 18 02239 g001
Figure 2. Proposed mechanisms of action of docosahexaenoic acid (DHA) in hypertriglyceridemic and hyperlipidemic acute pancreatitis. DHA inhibits triglyceride (TG) and fatty acid (FA) synthesis in the liver. DHA increases lipoprotein lipase (LPL) activity in the extrahepatic tissues and β-oxidation of FA in the liver and skeletal muscles, thereby contributing to the reduction of FA delivery to the liver and reducing plasma TG levels. High amounts of FAs induce pancreatic inflammation and injury. Therefore, the TG-lowering effect of DHA may prevent hypertriglyceridemic acute pancreatitis. On the other hand, a high concentration of DHA increases Ca2+ and activates PKC isoforms (PKC-α, PKC-δ, PKC-ε, and PKC-ζ) in pancreatic acinar cells, which may induce zymogene activation and pancreatic injury associated with hyperlipidemic acute pancreatitis. In addition, PKC activates NF-κB and induces inflammatory cytokine expression in pancreatic acinar cells. The bars represent inhibition, while the arrows represent stimulation.
Figure 2. Proposed mechanisms of action of docosahexaenoic acid (DHA) in hypertriglyceridemic and hyperlipidemic acute pancreatitis. DHA inhibits triglyceride (TG) and fatty acid (FA) synthesis in the liver. DHA increases lipoprotein lipase (LPL) activity in the extrahepatic tissues and β-oxidation of FA in the liver and skeletal muscles, thereby contributing to the reduction of FA delivery to the liver and reducing plasma TG levels. High amounts of FAs induce pancreatic inflammation and injury. Therefore, the TG-lowering effect of DHA may prevent hypertriglyceridemic acute pancreatitis. On the other hand, a high concentration of DHA increases Ca2+ and activates PKC isoforms (PKC-α, PKC-δ, PKC-ε, and PKC-ζ) in pancreatic acinar cells, which may induce zymogene activation and pancreatic injury associated with hyperlipidemic acute pancreatitis. In addition, PKC activates NF-κB and induces inflammatory cytokine expression in pancreatic acinar cells. The bars represent inhibition, while the arrows represent stimulation.
Ijms 18 02239 g002

Share and Cite

MDPI and ACS Style

Jeong, Y.K.; Kim, H. A Mini-Review on the Effect of Docosahexaenoic Acid (DHA) on Cerulein-Induced and Hypertriglyceridemic Acute Pancreatitis. Int. J. Mol. Sci. 2017, 18, 2239. https://doi.org/10.3390/ijms18112239

AMA Style

Jeong YK, Kim H. A Mini-Review on the Effect of Docosahexaenoic Acid (DHA) on Cerulein-Induced and Hypertriglyceridemic Acute Pancreatitis. International Journal of Molecular Sciences. 2017; 18(11):2239. https://doi.org/10.3390/ijms18112239

Chicago/Turabian Style

Jeong, Yoo Kyung, and Hyeyoung Kim. 2017. "A Mini-Review on the Effect of Docosahexaenoic Acid (DHA) on Cerulein-Induced and Hypertriglyceridemic Acute Pancreatitis" International Journal of Molecular Sciences 18, no. 11: 2239. https://doi.org/10.3390/ijms18112239

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop