全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Newly Identified Mechanisms of Total Parenteral Nutrition Related Liver Injury

DOI: 10.1155/2014/621380

Full-Text   Cite this paper   Add to My Lib

Abstract:

Total parenteral nutrition (TPN), a lifesaving therapy, involves providing nutrition by bypassing the gut. Unfortunately it is associated with significant complications including gut atrophy and parenteral nutrition associated liver disease (PNALD). PNALD includes steatosis, cholestasis, disrupted glucose metabolism, disrupted lipid metabolism, cirrhosis, and liver failure. The etiopathogenesis remains poorly defined; however, an altered enterohepatic circulation, disrupting nuclear receptor signaling, is emerging as a promising mechanism. Rodent models and our piglet TPN model have shown that, during regular feeding, bile acids activate farnesoid X receptor (FXR) in the gut and enhance fibroblast growth factor 19 (FGF19) level. FGF19 regulates bile acid, lipid, and glucose metabolism. We noted reduced FGF19 with TPN use and substantial improvement in FGF19, bilirubin, and metabolic profiles with the FXR agonist chenodeoxycholic acid (CDCA). Additionally, CDCA caused gut growth and enhanced expression of glucagon like peptides (GLPs). GLPs regulate gut trophic effects, insulin, glucose homeostasis, and hepatic steatosis. GLP secretion is regulated by the CDCA activated receptor TGR5. This leads to an important conclusion that, in addition to a disrupted FXR-FGF19 axis, a disrupted TGR5-GLP axis may contribute to TPN related pathologies. Thus modulators of FXR-FGF19 and the TGR5-GLP axis could help bring forward novel treatment strategies. 1. Introduction Parenteral nutrition has been in use since the 1960s [1, 2]. Total parenteral nutrition (TPN) involves providing all of patient’s nutritional needs intravenously and is lifesaving in clinical settings in which adequate enteral based delivery of such nutrition is not possible. It has been one of the most promising modalities of nutrition in neonates, older pediatric patients, and adults with lost or impaired gut function. TPN infusion therapy has grown enormously over the last few decades. There are tens of thousands of patients worldwide permanently dependent on parenteral nutrition (PN) for survival [3]. In addition, there are several fold higher numbers of patients requiring PN for varying duration during hospital stay or home care. Unfortunately, it is also associated with important complications and significant morbidity and mortality, including the well-recognized parenteral nutrition associated liver disease (PNALD) which includes steatosis, cholestasis, disrupted glucose metabolism, disrupted lipid metabolism, cirrhosis, development of portal hypertension, and ultimately liver failure [4, 5]. The

References

[1]  S. J. Dudrick, D. W. Wilmore, H. M. Vars, and J. E. Rhoads, “Long-term total parenteral nutrition with growth, development, and positive nitrogen balance,” Surgery, vol. 64, no. 1, pp. 134–142, 1968.
[2]  S. J. Dudrick, “Early developments and clinical applications of total parenteral nutrition,” Journal of Parenteral and Enteral Nutrition, vol. 27, no. 4, pp. 291–299, 2003.
[3]  L. Howard, M. Ament, C. R. Fleming, M. Shike, and E. Steiger, “Current use and clinical outcome of home parenteral and enteral nutrition therapies in the United States,” Gastroenterology, vol. 109, no. 2, pp. 355–365, 1995.
[4]  D. H. Teitelbaum, “Parenteral nutrition-associated cholestasis,” Current Opinion in Pediatrics, vol. 9, no. 3, pp. 270–275, 1997.
[5]  E. R. Briones and F. L. Iber, “Liver and biliary tract changes and injury associated with total parenteral nutrition: pathogenesis and prevention,” Journal of the American College of Nutrition, vol. 14, no. 3, pp. 219–228, 1995.
[6]  B. A. Carter and R. J. Shulman, “Mechanisms of disease: update on the molecular etiology and fundamentals of parenteral nutrition associated cholestasis,” Nature Clinical Practice Gastroenterology and Hepatology, vol. 4, no. 5, pp. 277–287, 2007.
[7]  V. J. Kumpf, “Parenteral nutrition-associated liver disease in adult and pediatric patients,” Nutrition in Clinical Practice, vol. 21, no. 3, pp. 279–290, 2006.
[8]  A. K. Jain, B. Stoll, D. G. Burrin, J. J. Holst, and D. D. Moore, “Enteral bile acid treatment improves parenteral nutrition-related liver disease and intestinal mucosal atrophy in neonatal pigs,” The American Journal of Physiology: Gastrointestinal and Liver Physiology, vol. 302, no. 2, pp. G218–G224, 2012.
[9]  D. A. Kelly, “Intestinal failure-associated liver disease: what do we know today?” Gastroenterology, vol. 130, no. 2, pp. S70–S77, 2006.
[10]  D. L. Sigalet, “Short bowel syndrome in infants and children: an overview,” Seminars in Pediatric Surgery, vol. 10, no. 2, pp. 49–55, 2001.
[11]  P. J. Javid, S. Collier, D. Richardson et al., “The role of enteral nutrition in the reversal of parenteral nutrition-associated liver dysfunction in infants,” Journal of Pediatric Surgery, vol. 40, no. 6, pp. 1015–1018, 2005.
[12]  R. A. Drongowski and A. G. Coran, “An analysis of factors contributing to the development of total parenteral nutrition-induced cholestasis,” Journal of Parenteral and Enteral Nutrition, vol. 13, no. 6, pp. 586–589, 1989.
[13]  A. Dilsiz, “Total parenteral nutrition-associated cholestasis in surgical neonates,” Turkish Journal of Medical Sciences, vol. 29, pp. 689–692, 1999.
[14]  C. S. Erickson, A. J. Barlow, J. F. Pierre et al., “Colonic enteric nervous system analysis during parenteral nutrition,” Journal of Surgical Research, vol. 184, no. 1, pp. 132–137, 2013.
[15]  A. C. Mosenthal, D. Xu, and E. A. Deitch, “Elemental and intravenous total parenteral nutrition diet-induced gut barrier failure is intestinal site specific and can be prevented by feeding nonfermentable fiber,” Critical Care Medicine, vol. 30, no. 2, pp. 396–402, 2002.
[16]  M. Ekelund, M. Ekelund, S. S. Qader, M. Hallén, and E. Ekblad, “Effects of total parenteral nutrition on rat enteric nervous system, intestinal morphology, and motility,” Journal of Surgical Research, vol. 124, no. 2, pp. 187–193, 2005.
[17]  H. Niinikoski, B. Stoll, X. Guan et al., “Onset of small intestinal atrophy is associated with reduced intestinal blood flow in TPN-fed neonatal piglets,” Journal of Nutrition, vol. 134, no. 6, pp. 1467–1474, 2004.
[18]  M. Miyata, T. Hata, H. Yamakawa, T. Kagawa, K. Yoshinari, and Y. Yamazoe, “Involvement of multiple elements in FXR-mediated transcriptional activation of FGF19,” Journal of Steroid Biochemistry and Molecular Biology, vol. 132, no. 1-2, pp. 41–47, 2012.
[19]  F. J. Gonzalez, “Nuclear receptor control of enterohepatic circulation,” Comprehensive Physiology, vol. 2, no. 4, pp. 2811–2828, 2012.
[20]  M. Makishima, A. Y. Okamoto, J. J. Repa et al., “Identification of a nuclear receptor for bite acids,” Science, vol. 284, no. 5418, pp. 1362–1365, 1999.
[21]  R. G. J. Visschers, M. D. Luyer, F. G. Schaap, S. W. M. Olde Damink, and P. B. Soeters, “The gut-liver axis,” Current Opinion in Clinical Nutrition and Metabolic Care, vol. 16, no. 5, pp. 576–581, 2013.
[22]  T. Claudel, B. Staels, and F. Kuipers, “The Farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 25, no. 10, pp. 2020–2031, 2005.
[23]  S. Modica, M. Petruzzelli, E. Bellafante et al., “Selective activation of nuclear bile acid receptor FXR in the intestine protects mice against cholestasis,” Gastroenterology, vol. 142, no. 2, pp. 355.e4–365.e4, 2012.
[24]  S. J. L. B. Zweers, K. A. C. Booij, M. Komuta et al., “The human gallbladder secretes fibroblast growth factor 19 into bile: towards defining the role of fibroblast growth factor 19 in the enterobiliary tract,” Hepatology, vol. 55, no. 2, pp. 575–583, 2012.
[25]  B. Goodwin, S. A. Jones, R. R. Price et al., “A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis,” Molecular Cell, vol. 6, no. 3, pp. 517–526, 2000.
[26]  T. T. Lu, M. Makishima, J. J. Repa et al., “Molecular basis for feedback regulation of bile acid synthesis by nuclear receptos,” Molecular Cell, vol. 6, no. 3, pp. 507–515, 2000.
[27]  J. A. Holt, G. Luo, A. N. Billin et al., “Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis,” Genes and Development, vol. 17, no. 13, pp. 1581–1591, 2003.
[28]  L. Wang, Y.-K. Lee, D. Bundman et al., “Redundant pathways for negative feedback regulation of bile acid production,” Developmental Cell, vol. 2, no. 6, pp. 721–731, 2002.
[29]  T. Inagaki, M. Choi, A. Moschetta et al., “Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis,” Cell Metabolism, vol. 2, no. 4, pp. 217–225, 2005.
[30]  T. Lund?sen, C. G?lman, B. Angelin, and M. Rudling, “Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man,” Journal of Internal Medicine, vol. 260, no. 6, pp. 530–536, 2006.
[31]  M.-H. Xie, I. Holcomb, B. Deuel et al., “FGF-19, a novel fibroblast growth factor with unique specificity for FGFR4,” Cytokine, vol. 11, no. 10, pp. 729–735, 1999.
[32]  T. Nishimura, Y. Utsunomiya, M. Hoshikawa, H. Ohuchi, and N. Itoh, “Structure and expression of a novel human FGF, FGF-19, expressed in the fetal brain,” Biochimica et Biophysica Acta, vol. 1444, no. 1, pp. 148–151, 1999.
[33]  E. Tomlinson, L. Fu, L. John et al., “Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity,” Endocrinology, vol. 143, no. 5, pp. 1741–1747, 2002.
[34]  I. Kim, S.-H. Ahn, T. Inagaki et al., “Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine,” Journal of Lipid Research, vol. 48, no. 12, pp. 2664–2672, 2007.
[35]  L. Fu, L. M. John, S. H. Adams et al., “Fibroblast growth factor 19 increases metabolic rate and reverses dietary and leptin-deficient diabetes,” Endocrinology, vol. 145, no. 6, pp. 2594–2603, 2004.
[36]  S. E. Hughes, “Differential expression of the fibroblast growth factor receptor (FGFR) multigene family in normal human adult tissues,” Journal of Histochemistry and Cytochemistry, vol. 45, no. 7, pp. 1005–1019, 1997.
[37]  C. Yu, F. Wang, M. Kan et al., “Elevated cholesterol metabolism and bile acid synthesis in mice lacking membrane tyrosine kinase receptor FGFR4,” The Journal of Biological Chemistry, vol. 275, no. 20, pp. 15482–15489, 2000.
[38]  X. Huang, C. Yang, Y. Luo, C. Jin, F. Wang, and W. L. McKeehan, “FGFR4 prevents hyperlipidemia and insulin resistance but underlies high-fat diet-induced fatty liver,” Diabetes, vol. 56, no. 10, pp. 2501–2510, 2007.
[39]  J. E. Heubi, D. A. Wiechmann, V. Creutzinger et al., “Tauroursodeoxycholic acid (TUDCA) in the prevention of total parenteral nutrition-associated liver disease,” Journal of Pediatrics, vol. 141, no. 2, pp. 237–242, 2002.
[40]  E. Reihner, I. Bjorkhem, B. Angelin, S. Ewerth, and K. Einarsson, “Bile acid synthesis in humans: regulation of hepatic microsomal cholesterol 7α-hydroxylase activity,” Gastroenterology, vol. 97, no. 6, pp. 1498–1505, 1989.
[41]  D. J. Parks, S. G. Blanchard, R. K. Bledsoe et al., “Bile acids: natural ligands for an orphan nuclear receptor,” Science, vol. 284, no. 5418, pp. 1365–1368, 1999.
[42]  A. Lucas, S. R. Bloom, and A. Aynsley Green, “Metabolic and endocrine consequences of depriving preterm infants of enteral nutrition,” Acta Paediatrica Scandinavica, vol. 72, no. 2, pp. 245–249, 1983.
[43]  T. Fedorowski, G. Salen, G. S. Tint, and E. Mosbach, “Transformation of chenodeoxycholic acid and ursodeoxycholic acid by human intestinal bacteria,” Gastroenterology, vol. 77, no. 5, pp. 1068–1073, 1979.
[44]  C. R. Cole and T. R. Ziegler, “Small bowel bacterial overgrowth: a negative factor in gut adaptation in pediatric SBS,” Current Gastroenterology Reports, vol. 9, no. 6, pp. 456–462, 2007.
[45]  S. N. Lichtman, R. B. Sartor, J. Keku, and J. H. Schwab, “Hepatic inflammation in rats with experimental small intestinal bacterial overgrowth,” Gastroenterology, vol. 98, no. 2, pp. 414–423, 1990.
[46]  H. R. Freund, M. Muggia-Sullam, R. LaFrance, E. B. Enrione, M. B. Popp, and H. S. Bjornson, “A possible beneficial effect of metronidazole in reducing TPN-associated liver function derangements,” Journal of Surgical Research, vol. 38, no. 4, pp. 356–363, 1985.
[47]  J. F. Whiting, R. M. Green, A. B. Rosenbluth, and J. L. Gollan, “Tumor necrosis factor—alpha decreases hepatocyte bile salt uptake and mediates endotoxin-induced cholestasis,” Hepatology, vol. 22, no. 4, part 1, pp. 1273–1278, 1995.
[48]  A. A. Nanji and F. H. Anderson, “Cholestasis associated with parenteral nutrition develops more commonly with hematologic malignancy than with inflammatory bowel disease,” Journal of Parenteral and Enteral Nutrition, vol. 8, no. 3, p. 325, 1984.
[49]  H. Fouin-Fortunet, L. Le Quernec, S. Erlinger, E. Lerebours, and R. Colin, “Hepatic alterations during total parenteral nutrition in patients with inflammatory bowel disease: a possible consequence of lithocholate toxicity,” Gastroenterology, vol. 82, no. 5, part 1, pp. 932–937, 1982.
[50]  Y. J. Zheng, Y. K. Tam, and R. T. Coutts, “Endotoxin and cytokine released during parenteral nutrition,” Journal of Parenteral and Enteral Nutrition, vol. 28, no. 3, pp. 163–168, 2004.
[51]  E. H. Forrest, K. A. Oien, S. Dickson, D. Galloway, and P. R. Mills, “Improvement in cholestasis associated with total parenteral nutrition after treatment with an antibody against tumour necrosis factor alpha,” Liver, vol. 22, no. 4, pp. 317–320, 2002.
[52]  M. Trauner, M. Arrese, H. Lee, J. L. Boyer, and S. J. Karpen, “Endotoxin downregulates rat hepatic ntcp gene expression via decreased activity of critical transcription factors,” Journal of Clinical Investigation, vol. 101, no. 10, pp. 2092–2100, 1998.
[53]  M. Hojo, N. Sano, and H. Takikawa, “Effects of lipopolysaccharide on the biliary excretion of bile acids and organic anions in rats,” Journal of Gastroenterology and Hepatology, vol. 18, no. 7, pp. 815–821, 2003.
[54]  R. Ghose, T. L. Zimmerman, S. Thevananther, and S. J. Karpen, “Endotoxin leads to rapid subcellular re-localization of hepatic RXRα: a novel mechanism for reduced hepatic gene expression in inflammation,” Nuclear Receptor, vol. 2, article 4, 2004.
[55]  T. Inagaki, A. Moschetta, Y.-K. Lee et al., “Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 10, pp. 3920–3925, 2006.
[56]  R. M. Gadaleta, K. J. Van Erpecum, B. Oldenburg et al., “Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease,” Gut, vol. 60, no. 4, pp. 463–472, 2011.
[57]  B. A. Carter, O. A. Taylor, D. R. Prendergast et al., “Stigmasterol, a soy lipid-derived phytosterol, is an antagonist of the bile acid nuclear receptor FXR,” Pediatric Research, vol. 62, no. 3, pp. 301–306, 2007.
[58]  J. Fuchs, E. M. Fallon, K. M. Gura, and M. Puder, “Use of an omega-3 fatty acid-based emulsion in the treatment of parenteral nutrition-induced cholestasis in patients with microvillous inclusion disease,” Journal of Pediatric Surgery, vol. 46, no. 12, pp. 2376–2382, 2011.
[59]  H. E. Lilja, Y. Finkel, M. Paulsson, and S. Lucas, “Prevention and reversal of intestinal failure-associated liver disease in premature infants with short bowel syndrome using intravenous fish oil in combination with omega-6/9 lipid emulsions,” Journal of Pediatric Surgery, vol. 46, no. 7, pp. 1361–1367, 2011.
[60]  R. S. Venick and K. Calkins, “The impact of intravenous fish oil emulsions on pediatric intestinal failure-associated liver disease,” Current Opinion in Organ Transplantation, vol. 16, no. 3, pp. 306–311, 2011.
[61]  Y. H. Yu, E. C. Lin, S. C. Wu et al., “Docosahexaenoic acid regulates adipogenic genes in myoblasts via porcine peroxisome proliferator-activated receptor {gamma},” Journal of Animal Science, vol. 86, no. 12, pp. 3385–3392, 2008.
[62]  G. L. St?rvold, K. G. Fleten, C. G. Olsen, T. Follestad, H. E. Krokan, and S. A. Sch?nberg, “Docosahexaenoic acid activates some SREBP-2 targets Independent of cholesterol and ER stress in SW620 colon cancer cells,” Lipids, vol. 44, no. 8, pp. 673–683, 2009.
[63]  G. Howell III, X. Deng, C. Yellaturu et al., “N-3 polyunsaturated fatty acids suppress insulin-induced SREBP-1c transcription via reduced trans-activating capacity of LXRα,” Biochimica et Biophysica Acta, vol. 1791, no. 12, pp. 1190–1196, 2009.
[64]  N. Tanaka, X. Zhang, E. Sugiyama et al., “Eicosapentaenoic acid improves hepatic steatosis independent of PPARα activation through inhibition of SREBP-1 maturation in mice,” Biochemical Pharmacology, vol. 80, no. 10, pp. 1601–1612, 2010.
[65]  A. Zhao, J. Yu, J.-L. Lew, L. Huang, S. D. Wright, and J. Cui, “Polyunsaturated fatty acids are FXR ligands and differentially regulate expression of FXR targets,” DNA and Cell Biology, vol. 23, no. 8, pp. 519–526, 2004.
[66]  T. Yoshikawa, H. Shimano, N. Yahagi et al., “Polyunsaturated fatty acids suppress sterol regulatory element-binding protein 1c promoter activity by inhibition of liver X receptor (LXR) binding to LXR response elements,” Journal of Biological Chemistry, vol. 277, no. 3, pp. 1705–1711, 2002.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413