Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 1.
Published in final edited form as: Nutr Clin Pract. 2014 Mar 31;29(3):291–294. doi: 10.1177/0884533614527774

Potential Influence of Intravenous Lipids on the Outcomes of Acute Pancreatitis

Krutika S Patel 1, Pawan Noel 1, Vijay P Singh 1
PMCID: PMC4040228  NIHMSID: NIHMS587824  PMID: 24687866

Abstract

Parenteral nutrition (PN) has been associated with a higher rate of adverse outcomes compared with enteral feeding in patients with acute pancreatitis (AP). However, PN may be necessary when feeding via the enteral route is poorly tolerated or impossible, and PN is recommended as a second-line nutrition therapy in AP. Intravenous (IV) lipids are commonly used as a part of PN in patients with AP. While the adverse outcomes related to the use of PN in AP have commonly been attributed to infectious complications, data suggest that the unsaturated fatty acids in the triglycerides used in IV lipids may contribute to the development of organ failure. We discuss the clinical and experimental data on this issue and the alternative lipid emulsions that are being studied.

Keywords: pancreatic diseases, pancreatitis, nutritional support, parenteral nutrition, total parenteral nutrition, intravenous fat emulsions


Parenteral nutrition (PN) is a frequently needed nutrition resource in patients with severe acute pancreatitis (AP) who are intolerant to enteral feeding or in whom it is not feasible. In the following paragraphs, we briefly discuss the rationale for use of PN, including intravenous (IV) lipids, and analyze how the composition of IV lipids may influence adverse outcomes noted in severe AP in light of published literature. In addition, we comment on the alternate formulations and what is needed to make more conclusive analyses regarding the use of IV lipids in AP.

Recent studies from North America show the usage of PN to be as high as 40%–60%1,2 in patients with AP. Intravenous (IV) lipids have been used in most studies analyzing the effect of PN in AP.310 Their use may minimize risks associated with high doses of IV dextrose such as hyperglycemia and fatty liver. In addition, the typical rationale for using PN during AP is to provide nutrition while not stimulating pancreatic enzyme secretion.11,12 PN has been shown to achieve this by reducing the levels of cholecystokinin13,14 and exocrine pancreatic stimulation by the vagus15,16 in patients administered IV lipids, which in contrast to enteral lipids do not elevate plasma cholecystokinin levels.17

While there are abundant data on the superiority of enteral nutrition (EN)6,1820 over PN in AP and recent guidelines recommend the use of PN as a second-line nutrition therapy,21 in several situations, PN is indicated in severe AP. These include the lack of enteral access, tube malpositioning, discomfort and dislodgement,4 intolerance to enteral feeding, ileus, complex pancreatic fistulae, abdominal compartment syndrome,22 abdominal pain, and diarrhea.18 Factors that may contribute to the superiority of EN include improved gut barrier function23 and improved glycemic control,24 resulting in a lower risk of pancreatic6 and systemic infections25; fewer operative interventions; a lower incidence of multiple organ failure6 and mortality6; and a trend favoring reduced hospital stay.25

IV lipid emulsions available in the United States contain soybean oil and are enriched in the long-chain unsaturated fatty acids (UFAs) oleic acid, linoleic acid, and linolenic acid, which form 70%–85% of the triglyceride (TG) content.26 Some of these fatty acids have been shown to be proinflammatory.2628 Several studies in patients with AP show the prevalence of organ failure to be >50% in the PN group.3,6,8,29 While part of the organ failure could be from sepsis,25 central line–related infections,30 and infected pancreatic necrosis, the possibility of organ failure related to the IV lipid in PN, independent of infections, may exist. While no trials have compared outcomes of AP in patients receiving IV lipids with those receiving PN without lipids, or the outcomes of different rates of infusion of IV lipids, several randomized clinical trials have compared outcomes of patients with AP receiving IV lipids as a part of the PN38,24 with those receiving EN (Table 1). A significant proportion among these show a higher prevalence of organ failure and mortality in the PN group.38 Basic science literature shows UFAs contribute to organ failure associated with AP.28,31 In addition, hypertriglyceridemia (>1000 mg/dL) is also typically associated with severe AP,3238 and while guidelines permit the use of IV lipids if baseline TGs are <400 mg/dL,39 the prevalence of hypertriglyceridemia may vary from 7.6% (TG cutoff >4.5 mmol)40 to 26% (TG cutoff >3 mmol)41 during PN. IV lipids may sometimes result in serum triglyceride levels >1000 mg/dL42 and fatty acids 6- to 8-fold above normal,42 which are in or above the range of serum levels associated with severe AP.43,44 In the following paragraphs, we discuss the mechanistic findings regarding how AP may be worsened by hyperlipidemia and the nature of studies needed in the future.

Table 1.

Randomized Controlled Trials Comparing Outcomes in Patients With Acute Pancreatitis Receiving EN vs PN That Included IV Lipids.

Author (Year) IV Lipid Brand (Conc.),
Target Dose/24 h
No. of Deaths/Organ Failure/Patients

EN Group IV Lipid Group
Wu et al3 (2010) Intralipid (20%), 250 mL 6/11/53 23/44/54
Eckerwall et al24 (2006) Kabiven (?), 25 kcal/kg 1/1/24 0/1/26
Petrov et al6 (2006) ? (10%), 30 kcal/kg 2/11/35 12/27/34
Louie et al4 (2005) Intralipid (10%), 25 kcal/kg 0/7/10 3/13/18
Gupta et al8 (2003) ? (10%), 500 mL 0/0/8 0/6/9
Olah et al7 (2002) Intralipid (10%), 30 kcal/kg 2/2/41 4/5/48
Kalfarentzos et al5 (1997) Lipofudin (20%), 30–35 kcal/kg 1/2/18 2/4/20

The second column mentions the brand of intravenous (IV) lipid used in the study, the concentration (Conc.) of the lipid emulsion, and the method of dose calculation described in the study (ie, volume of the IV lipid in milliliters/24 hours or target total kcal/kg/24 hours, including those from other sources). The 2 columns on the right mention the outcomes in each group as the number of deaths/number of patients with organ failure/total number of patients in the group. The “?” indicates that the information was not specified.

EN, enteral nutrition; PN, parenteral nutrition.

A significant body of literature supports the contribution of high UFAs to adverse outcomes in AP and organ failure. While the studies do not mention whether the patients received PN, elevated UFAs have been noted in the sera43,44 and necrotic collections of patients with severe AP.28,45 Studies in pancreatic acinar cells have shown that UFAs may contribute to local injury,28,46,47 and experimental models of pancreatitis show that administration of unsaturated TGs into the pancreas or elevated serum UFAs are respectively associated with worse pancreatic necrosis and multisystem organ failure.28,48 Elevated C18 UFAs have been noted to predict acute respiratory distress syndrome (ARDS)49 in humans, and administration of UFAs replicate components of multisystem organ failure in experimental studies. These include hypocalcemia,50 acute renal failure,51 ARDS,52 and an exaggerated inflammatory state.53 TG emulsions have also been shown to worsen the severity of ARDS and lung inflammation54 in humans. The mechanisms by which these may be mediated include release of phospholipase A2, platelet activating factor,54 reactive oxygen species,55 and metabolites of UFAs56,57 that may cause cellular injury.58,59 Moreover, the propensity of TGs in IV lipid emulsions to form UFAs may be enhanced by the hyperlipasemia of AP. This may be mediated by the extracellular lipases cleaving TGs to their constituent fatty acids, the majority of which would be UFAs, further contributing to adverse outcomes associated with them. Studies showing that critically ill patients with unexplained lipase elevation have a greater need for mechanical ventilation60 and patients with AP who died receiving PN had serum TGs 2- to 4-fold higher than (but <1000 mg/dL) those of survivors35 support this notion.

Some studies, however, support the use of PN during AP. These include early studies showing PN safely improving the nutrition status of patients with AP61,62 and ones showing EN being associated with a higher rate of overall and pulmonary complications.24 Studies in rodents have also shown that there was no difference in mortality of rodents with severe AP whether or not they were given IV lipids.63 Rats with mild caerulein pancreatitis administered IV TGs have been shown to have no worsening of local severity compared with caerulein alone64 or an improvement in edema, inflammation, and the histological appearance of the pancreas.65 However, a careful look at the design and parameters measured in these studies brings their relevance to severe AP outcomes into question. In the study showing that IV lipids did not worsen severe AP,63 the mortality in both groups (ie, with and without lipids) was 64%, but the study was done on only about half the original animals due to a high mortality and technical issues prior to administration of the lipid. This high mortality in the control or untreated groups makes the implications of this study hard to interpret. The other 2 studies on IV TGs in caerulein pancreatitis64,65 did not study renal or lung injury and were terminated within 6 hours of induction of pancreatitis—an interval too short to study the systemic implications of lipotoxicity.66

Realizing the concerns associated with current formulations, clinical trials have explored versions of IV lipid that may be less toxic than what is currently available. These include medium-chain TGs and ω-3 fatty acids. A prospective crossover study found a 1:1 ratio of medium-chain TGs and soybean oil to be associated with improved PaO2 /FiO2 compared with soybean oil alone10 in patients with AP. Fish oil is enriched in ω-3 fatty acids, which are known to form less toxic metabolites and a reduced inflammatory state67 than the ω-6 fatty acids comprising soybean oil.26,27 Randomized controlled trials have noted ω-3 fatty acid supplementation to be associated with improved oxygenation and reduced renal injury in human AP,9 and while most studies in experimental AP show better outcomes with these,6870 others have noted that fish oil supplementation did not alter the proinflammatory milieu or the parameters of AP severity.71

In summary, when using the enteral route is not feasible, IV lipids are an important nutrition resource in AP. More definitive studies comparing the efficacy of conventional IV lipid formulations with isocaloric PN without lipids or alternate formulations of IV lipids (such as those enriched in medium-chain TGs or ω-3 fatty acids) in reducing the local and systemic compilations associated with AP are needed.

Acknowledgments

Financial disclosure: This project was supported by grant RO1DK092460 from the National Institutes of Health (V.P.S.).

References

  • 1.Sun E, Tharakan M, Kapoor S, et al. Poor compliance with ACG guidelines for nutrition and antibiotics in the management of acute pancreatitis: a North American survey of gastrointestinal specialists and primary care physicians. J Pancreas. 2013;14:221–227. doi: 10.6092/1590-8577/871. [DOI] [PubMed] [Google Scholar]
  • 2.Vlada AC, Schmit B, Perry A, Trevino JG, Behrns KE, Hughes SJ. Failure to follow evidence-based best practice guidelines in the treatment of severe acute pancreatitis. HPB. 2013;15:822–827. doi: 10.1111/hpb.12140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wu XM, Ji KQ, Wang HY, Li GF, Zang B, Chen WM. Total enteral nutrition in prevention of pancreatic necrotic infection in severe acute pancreatitis. Pancreas. 2010;39:248–251. doi: 10.1097/MPA.0b013e3181bd6370. [DOI] [PubMed] [Google Scholar]
  • 4.Louie BE, Noseworthy T, Hailey D, Gramlich LM, Jacobs P, Warnock GL. 2004 MacLean-Mueller prize enteral or parenteral nutrition for severe pancreatitis: a randomized controlled trial and health technology assessment. Can J Surg. 2005;48:298–306. [PMC free article] [PubMed] [Google Scholar]
  • 5.Kalfarentzos F, Kehagias J, Mead N, Kokkinis K, Gogos CA. Enteral nutrition is superior to parenteral nutrition in severe acute pancreatitis: results of a randomized prospective trial. Br J Surg. 1997;84:1665–1669. [PubMed] [Google Scholar]
  • 6.Petrov MS, Kukosh MV, Emelyanov NV. A randomized controlled trial of enteral versus parenteral feeding in patients with predicted severe acute pancreatitis shows a significant reduction in mortality and in infected pancreatic complications with total enteral nutrition. Dig Surg. 2006;23:336–345. doi: 10.1159/000097949. [DOI] [PubMed] [Google Scholar]
  • 7.Olah A, Pardavi G, Belagyi T, Nagy A, Issekutz A, Mohamed GE. Early nasojejunal feeding in acute pancreatitis is associated with a lower complication rate. Nutrition. 2002;18:259–262. doi: 10.1016/s0899-9007(01)00755-9. [DOI] [PubMed] [Google Scholar]
  • 8.Gupta R, Patel K, Calder PC, Yaqoob P, Primrose JN, Johnson CD. A randomised clinical trial to assess the effect of total enteral and total parenteral nutritional support on metabolic, inflammatory and oxidative markers in patients with predicted severe acute pancreatitis (APACHE II >or =6) Pancreatology. 2003;3:406–413. doi: 10.1159/000073657. [DOI] [PubMed] [Google Scholar]
  • 9.Wang X, Li W, Li N, Li J. Omega-3 fatty acids–supplemented parenteral nutrition decreases hyperinflammatory response and attenuates systemic disease sequelae in severe acute pancreatitis: a randomized and controlled study. JPEN J Parenter Enteral Nutr. 2008;32:236–241. doi: 10.1177/0148607108316189. [DOI] [PubMed] [Google Scholar]
  • 10.Smyrniotis VE, Kostopanagiotou GG, Arkadopoulos NF, et al. Long-chain versus medium-chain lipids in acute pancreatitis complicated by acute respiratory distress syndrome: effects on pulmonary hemodynamics and gas exchange. Clin Nutr. 2001;20:139–143. doi: 10.1054/clnu.2000.0370. [DOI] [PubMed] [Google Scholar]
  • 11.Niederau C, Sonnenberg A, Erckenbrecht J. Effects of intravenous infusion of amino acids, fat, or glucose on unstimulated pancreatic secretion in healthy humans. Dig Dis Sci. 1985;30:445–455. doi: 10.1007/BF01318177. [DOI] [PubMed] [Google Scholar]
  • 12.Stabile BE, Borzatta M, Stubbs RS. Pancreatic secretory responses to intravenous hyperalimentation and intraduodenal elemental and full liquid diets. JPEN J Parenter Enteral Nutr. 1984;8:377–380. doi: 10.1177/0148607184008004377. [DOI] [PubMed] [Google Scholar]
  • 13.Mashako MN, Cezard JP, Boige N, Chayvialle JA, Bernard C, Navarro J. The effect of artificial feeding on cholestasis, gallbladder sludge and lithiasis in infants: correlation with plasma cholecystokinin levels. Clin Nutr. 1991;10:320–327. doi: 10.1016/0261-5614(91)90061-g. [DOI] [PubMed] [Google Scholar]
  • 14.Mashako MN, Bernard C, Cezard JP, Chayvialle JA, Navarro J. Effect of total parenteral nutrition, constant rate enteral nutrition, and discontinuous oral feeding on plasma cholecystokinin immunoreactivity in children. J Pediatr Gastroenterol Nutr. 1987;6:948–952. doi: 10.1097/00005176-198711000-00022. [DOI] [PubMed] [Google Scholar]
  • 15.Singer MV, Niebergall-Roth E. Secretion from acinar cells of the exocrine pancreas: role of enteropancreatic reflexes and cholecystokinin. Cell Biol Int. 2009;33:1–9. doi: 10.1016/j.cellbi.2008.09.008. [DOI] [PubMed] [Google Scholar]
  • 16.Niebergall-Roth E, Singer MV. Enteropancreatic reflexes mediating the pancreatic enzyme response to nutrients. Auton Neurosc. 2006;125:62–69. doi: 10.1016/j.autneu.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 17.de Boer SY, Masclee AA, Jebbink MC, Schipper J, Jansen JB, Lamers CB. Effect of intravenous fat on cholecystokinin secretion and gallbladder motility in man. JPEN J Parenter Enteral Nutr. 1992;16:16–19. doi: 10.1177/014860719201600116. [DOI] [PubMed] [Google Scholar]
  • 18.Quan H, Wang X, Guo C. A meta-analysis of enteral nutrition and total parenteral nutrition in patients with acute pancreatitis. Gastroenterol Res Pract. 2011;2011:698248. doi: 10.1155/2011/698248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yi F, Ge L, Zhao J, et al. Meta-analysis: total parenteral nutrition versus total enteral nutrition in predicted severe acute pancreatitis. Intern Med. 2012;51:523–530. doi: 10.2169/internalmedicine.51.6685. [DOI] [PubMed] [Google Scholar]
  • 20.Casas M, Mora J, Fort E, et al. Total enteral nutrition vs. total parenteral nutrition in patients with severe acute pancreatitis [in Spanish] Rev Esp Enferm Dig. 2007;99:264–269. doi: 10.4321/s1130-01082007000500004. [DOI] [PubMed] [Google Scholar]
  • 21.IAP/APA evidence-based guidelines for the management of acute pancreatitis. Pancreatology. 2013;13:e1–e15. doi: 10.1016/j.pan.2013.07.063. [DOI] [PubMed] [Google Scholar]
  • 22.Gianotti L, Meier R, Lobo DN, et al. ESPEN Guidelines on Parenteral Nutrition: pancreas. Clin Nutr. 2009;28:428–435. doi: 10.1016/j.clnu.2009.04.003. [DOI] [PubMed] [Google Scholar]
  • 23.Xu CF, Huang XX, Shen YZ, Wang XP, Gong L, Wang YD. The effects of enteral nutrition versus total parenteral nutrition on gut barrier function in severe acute pancreatitis [in Chinese] Zhonghua Nei Ke Za Zhi. 2011;50:370–373. [PubMed] [Google Scholar]
  • 24.Eckerwall GE, Axelsson JB, Andersson RG. Early nasogastric feeding in predicted severe acute pancreatitis: a clinical, randomized study. Ann Surg. 2006;244:959–967. doi: 10.1097/01.sla.0000246866.01930.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Al-Omran M, Albalawi ZH, Tashkandi MF, Al-Ansary LA. Enteral versus parenteral nutrition for acute pancreatitis. Cochrane Database Syst Rev. 2010;(1):CD002837. doi: 10.1002/14651858.CD002837.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vanek VW, Seidner DL, Allen P, et al. A.S.P.E.N. position paper: clinical role for alternative intravenous fat emulsions. Nutr Clin Pract. 2012;27:150–192. doi: 10.1177/0884533612439896. [DOI] [PubMed] [Google Scholar]
  • 27.Wall R, Ross RP, Fitzgerald GF, Stanton C. Fatty acids from fish: the anti-inflammatory potential of long-chain omega-3 fatty acids. Nutr Rev. 2010;68:280–289. doi: 10.1111/j.1753-4887.2010.00287.x. [DOI] [PubMed] [Google Scholar]
  • 28.Navina S, Acharya C, DeLany JP, et al. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci Trans Med. 2011;3:107ra110. doi: 10.1126/scitranslmed.3002573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Targarona Modena J, Barreda Cevasco L, Arroyo Basto C, Orellana Vicuna A, Portanova Ramirez M. Total enteral nutrition as prophylactic therapy for pancreatic necrosis infection in severe acute pancreatitis. Pancreatology. 2006;6:58–64. doi: 10.1159/000090024. [DOI] [PubMed] [Google Scholar]
  • 30.Wu BU, Johannes RS, Kurtz S, Banks PA. The impact of hospital-acquired infection on outcome in acute pancreatitis. Gastroenterology. 2008;135:816–820. doi: 10.1053/j.gastro.2008.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kimura T, Toung JK, Margolis S, Permutt S, Cameron JL. Respiratory failure in acute pancreatitis: a possible role for triglycerides. Ann Surg. 1979;189:509–514. [PMC free article] [PubMed] [Google Scholar]
  • 32.Buch A, Buch J, Carlsen A, Schmidt A. Hyperlipidemia and pancreatitis. World J Surg. 1980;4:307–314. doi: 10.1007/BF02393387. [DOI] [PubMed] [Google Scholar]
  • 33.Dominguez-Munoz JE, Malfertheiner P, Ditschuneit HH, et al. Hyperlipidemia in acute pancreatitis: relationship with etiology, onset, and severity of the disease. Int J Pancreatol. 1991;10:261–267. [PubMed] [Google Scholar]
  • 34.Warshaw AL, Lesser PB, Rie M, Cullen DJ. The pathogenesis of pulmonary edema in acute pancreatitis. Ann Surg. 1975;182:505–510. doi: 10.1097/00000658-197510000-00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Van Gossum A, Lemoyne M, Greig PD, Jeejeebhoy KN. Lipid-associated total parenteral nutrition in patients with severe acute pancreatitis. JPEN J Parenter Enteral Nutr. 1988;12:250–255. doi: 10.1177/0148607188012003250. [DOI] [PubMed] [Google Scholar]
  • 36.Nordstoga K, Christophersen B, Ytrehus B, et al. Pancreatitis associated with hyperlipoproteinaemia type I in mink (Mustela vison): earliest detectable changes occur in mitochondria of exocrine cells. J Comp Pathol. 2006;134:320–328. doi: 10.1016/j.jcpa.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 37.Deng LH, Xue P, Xia Q, Yang XN, Wan MH. Effect of admission hypertriglyceridemia on the episodes of severe acute pancreatitis. World J Gastroenterol. 2008;14:4558–4561. doi: 10.3748/wjg.14.4558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lloret Linares C, Pelletier AL, Czernichow S, et al. Acute pancreatitis in a cohort of 129 patients referred for severe hypertriglyceridemia. Pancreas. 2008;37:13–22. doi: 10.1097/MPA.0b013e31816074a1. [DOI] [PubMed] [Google Scholar]
  • 39.Mirtallo JM, Forbes A, McClave SA, Jensen GL, Waitzberg DL, Davies AR. International consensus guidelines for nutrition therapy in pancreatitis. JPEN J Parenter Enteral Nutr. 2012;36:284–291. doi: 10.1177/0148607112440823. [DOI] [PubMed] [Google Scholar]
  • 40.Visschers RG, Olde Damink SW, Gehlen JM, Winkens B, Soeters PB, van Gemert WG. Treatment of hypertriglyceridemia in patients receiving parenteral nutrition. JPEN J Parenter Enteral Nutr. 2011;35:610–615. doi: 10.1177/0148607110389616. [DOI] [PubMed] [Google Scholar]
  • 41.Llop J, Sabin P, Garau M, et al. The importance of clinical factors in parenteral nutrition–associated hypertriglyceridemia. Clin Nutr. 2003;22:577–583. doi: 10.1016/s0261-5614(03)00082-7. [DOI] [PubMed] [Google Scholar]
  • 42.Hughan KS, Bonadonna RC, Lee S, Michaliszyn SF, Arslanian SA. Beta-cell lipotoxicity after an overnight intravenous lipid challenge and free fatty acid elevation in African American versus American white overweight/obese adolescents. J Clin Endocrinol Metab. 2013;98:2062–2069. doi: 10.1210/jc.2012-3492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Domschke S, Malfertheiner P, Uhl W, Buchler M, Domschke W. Free fatty acids in serum of patients with acute necrotizing or edematous pancreatitis. Int J Pancreatol. 1993;13:105–110. doi: 10.1007/BF02786078. [DOI] [PubMed] [Google Scholar]
  • 44.Sztefko K, Panek J. Serum free fatty acid concentration in patients with acute pancreatitis. Pancreatology. 2001;1:230–236. doi: 10.1159/000055816. [DOI] [PubMed] [Google Scholar]
  • 45.Panek J, Sztefko K, Drozdz W. Composition of free fatty acid and triglyceride fractions in human necrotic pancreatic tissue. Med Sci Monitor. 2001;7:894–898. [PubMed] [Google Scholar]
  • 46.Mossner J, Bodeker H, Kimura W, Meyer F, Bohm S, Fischbach W. Isolated rat pancreatic acini as a model to study the potential role of lipase in the pathogenesis of acinar cell destruction. Int J Pancreatol. 1992;12:285–296. doi: 10.1007/BF02924368. [DOI] [PubMed] [Google Scholar]
  • 47.Acharya C, Cline RA, Jaligama D, et al. Fibrosis reduces severity of acute-on-chronic pancreatitis in humans. Gastroenterology. 2013;145:466–475. doi: 10.1053/j.gastro.2013.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Willemer S, Elsasser HP, Kern HF, Adler G. Tubular complexes in ceruleinand oleic acid–induced pancreatitis in rats: glycoconjugate pattern, immunocytochemical, and ultrastructural findings. Pancreas. 1987;2:669–675. doi: 10.1097/00006676-198711000-00008. [DOI] [PubMed] [Google Scholar]
  • 49.Bursten SL, Federighi DA, Parsons P, et al. An increase in serum C18 unsaturated free fatty acids as a predictor of the development of acute respiratory distress syndrome. Crit Care Med. 1996;24:1129–1136. doi: 10.1097/00003246-199607000-00011. [DOI] [PubMed] [Google Scholar]
  • 50.Dettelbach MA, Deftos LJ, Stewart AF. Intraperitoneal free fatty acids induce severe hypocalcemia in rats: a model for the hypocalcemia of pancreatitis. J Bone Miner Res. 1990;5:1249–1255. doi: 10.1002/jbmr.5650051210. [DOI] [PubMed] [Google Scholar]
  • 51.Wu RP, Liang XB, Guo H, et al. Protective effect of low potassium dextran solution on acute kidney injury following acute lung injury induced by oleic acid in piglets. Chin Med J. 2012;125:3093–3097. [PubMed] [Google Scholar]
  • 52.Hussain N, Wu F, Zhu L, Thrall RS, Kresch MJ. Neutrophil apoptosis during the development and resolution of oleic acid–induced acute lung injury in the rat. Am J Respir Cell Mol Biol. 1998;19:867–874. doi: 10.1165/ajrcmb.19.6.3118. [DOI] [PubMed] [Google Scholar]
  • 53.Inoue H, Nakagawa Y, Ikemura M, Usugi E, Nata M. Molecular-biological analysis of acute lung injury (ALI) induced by heat exposure and/or intravenous administration of oleic acid. Legal Med. 2012;14:304–308. doi: 10.1016/j.legalmed.2012.06.003. [DOI] [PubMed] [Google Scholar]
  • 54.Lekka ME, Liokatis S, Nathanail C, Galani V, Nakos G. The impact of intravenous fat emulsion administration in acute lung injury. Am J Respir Crit Care Med. 2004;169:638–644. doi: 10.1164/rccm.200305-620OC. [DOI] [PubMed] [Google Scholar]
  • 55.Liu H, Zhang D, Zhao B, Zhao J. Superoxide anion, the main species of ROS in the development of ARDS induced by oleic acid. Free Radical Res. 2004;38:1281–1287. doi: 10.1080/10715760400006940. [DOI] [PubMed] [Google Scholar]
  • 56.Edwards LM, Lawler NG, Nikolic SB, et al. Metabolomics reveals increased isoleukotoxin diol (12,13-DHOME) in human plasma after acute Intralipid infusion. J Lipid Res. 2012;53:1979–1986. doi: 10.1194/jlr.P027706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Quinlan GJ, Lamb NJ, Evans TW, Gutteridge JM. Plasma fatty acid changes and increased lipid peroxidation in patients with adult respiratory distress syndrome. Crit Care Med. 1996;24:241–246. doi: 10.1097/00003246-199602000-00010. [DOI] [PubMed] [Google Scholar]
  • 58.Ishola DA, Jr, Post JA, van Timmeren MM, et al. Albumin-bound fatty acids induce mitochondrial oxidant stress and impair antioxidant responses in proximal tubular cells. Kidney Int. 2006;70:724–731. doi: 10.1038/sj.ki.5001629. [DOI] [PubMed] [Google Scholar]
  • 59.Moran JH, Nowak G, Grant DF. Analysis of the toxic effects of linoleic acid, 12,13-cis-epoxyoctadecenoic acid, and 12,13-dihydroxyoctadecenoic acid in rabbit renal cortical mitochondria. Toxicol Appl Pharmacol. 2001;172:150–161. doi: 10.1006/taap.2001.9149. [DOI] [PubMed] [Google Scholar]
  • 60.Manjuck J, Zein J, Carpati C, Astiz M. Clinical significance of increased lipase levels on admission to the ICU. Chest. 2005;127:246–250. doi: 10.1378/chest.127.1.246. [DOI] [PubMed] [Google Scholar]
  • 61.Sitzmann JV, Steinborn PA, Zinner MJ, Cameron JL. Total parenteral nutrition and alternate energy substrates in treatment of severe acute pancreatitis. Surg Gynecol Obstet. 1989;168:311–317. [PubMed] [Google Scholar]
  • 62.Robin AP, Campbell R, Palani CK, Liu K, Donahue PE, Nyhus LM. Total parenteral nutrition during acute pancreatitis: clinical experience with 156 patients. World J Surg. 1990;14:572–579. doi: 10.1007/BF01658792. [DOI] [PubMed] [Google Scholar]
  • 63.Raasch RH, Hak LJ, Benaim V, Brower L, Levinson SL, Heizer WD. Effect of intravenous fat emulsion on experimental acute pancreatitis. JPEN J Parenter Enteral Nutr. 1983;7:254–256. doi: 10.1177/0148607183007003254. [DOI] [PubMed] [Google Scholar]
  • 64.Paye F, Chariot J, Molas G, Benessiano J, Roze C. Nonesterified fatty acids in acute cerulein-induced pancreatitis in the rat: are they really deleterious in vivo? Dig Dis Sci. 1995;40:540–545. doi: 10.1007/BF02064365. [DOI] [PubMed] [Google Scholar]
  • 65.Koopmann MC, Baumler MD, Boehler CJ, Chang FL, Ney DM, Groblewski GE. Total parenteral nutrition attenuates cerulein-induced pancreatitis in rats. Pancreas. 2010;39:377–384. doi: 10.1097/MPA.0b013e3181bb908e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Pini M, Rhodes DH, Castellanos KJ, Cabay RJ, Grady EF, Fantuzzi G. Rosiglitazone improves survival and hastens recovery from pancreatic inflammation in obese mice. PLoS One. 2012;7:e40944. doi: 10.1371/journal.pone.0040944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wang X, Li W, Zhang F, Pan L, Li N, Li J. Fish oil–supplemented parenteral nutrition in severe acute pancreatitis patients and effects on immune function and infectious risk: a randomized controlled trial. Inflammation. 2009;32:304–309. doi: 10.1007/s10753-009-9136-0. [DOI] [PubMed] [Google Scholar]
  • 68.Rangel-Huerta OD, Aguilera CM, Mesa MD, Gil A. Omega-3 longchain polyunsaturated fatty acids supplementation on inflammatory biomarkers: a systematic review of randomised clinical trials. Br J Nutr. 2012;107(suppl 2):S159–S170. doi: 10.1017/S0007114512001559. [DOI] [PubMed] [Google Scholar]
  • 69.Foitzik T, Eibl G, Schneider P, Wenger FA, Jacobi CA, Buhr HJ. Omega-3 fatty acid supplementation increases anti-inflammatory cytokines and attenuates systemic disease sequelae in experimental pancreatitis. JPEN J Parenter Enteral Nutr. 2002;26:351–356. doi: 10.1177/0148607102026006351. [DOI] [PubMed] [Google Scholar]
  • 70.Weylandt KH, Nadolny A, Kahlke L, et al. 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. doi: 10.1016/j.bbadis.2008.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Al-Azzawi HH, Wade TE, Swartz-Basile DA, Wang S, Pitt HA, Zyromski NJ. Acute pancreatitis in obesity: adipokines and dietary fish oil. Dig Dis Sci. 2011;56:2318–2325. doi: 10.1007/s10620-011-1626-x. [DOI] [PubMed] [Google Scholar]

RESOURCES