Quick Search
  Home Journal Information Current Issue Past Issues Services Contact Us  
Articles
Total parenteral nutrition-induced liver dysfunction: evidence and pathogenesis 
 
Total parenteral nutrition-induced liver dysfunction: evidence and pathogenesis
  Xiao-Gang Hui, Yutaka Hayashi
 [Abstract] [Full Text] [PDF]   Pageviews: 13937 Times
 

Total parenteral nutrition-induced liver

dysfunction: evidence and pathogenesis

Xiao-Gang Hui, Yutaka Hayashi

Tianjin, China and Sendai, Japan

Author Affiliations: Department of Pediatric Surgery, Tianjin Children's Hospital, China (Hui XG); Department of Pediatric Surgery, Tohoku University School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, 980-8574, Japan (Hayashi Y)

Corresponding Author: Xiao-Gang Hui, MD, Department of Pediatric Surgery, Tianjin Children's Hospital, China (the present address: Department of Pediatric Surgery of Tohoku University Affiliated Hospital in Sendai, Japan; Email: barberahuizhai@yahoo.com.cn).

Background: Total parenteral nutrition (TPN) is known to be life-saving in patients with intestinal failure. But long-term TPN-related complications especially liver dysfunction have been the focus of studies. In this article, we address the evidence of TPN-induced liver lesions and the pathogenesis of these lesions.

Data sources: The articles about the relationship between TPN and liver function were retrieved from PubMed database.

Results: Varied injuries to the liver are induced clinically and experimentally by progressive deterioration after a longer period of TPN infusion. The mechanisms of cholestasis and fibrosis in pathological changes include alteration of trace elements in hepatocytes, metabolic disturbance of fatty acid, calorie overload, lithocholate effect, sepsis, etc. The administration route of TPN is excluded from the pathogenesis of liver disease, and light infusion should be avoided.

Conclusions: TPN may lead to liver dysfunction, but the causes are multifactorial. The authentic factor is unknown although progresses have been made in this field. Since conspicuous lesions are confined in the mesenchyma, further studies should concentrate on the relationship between Ito cells and TPN administration. The significant step toward the gene expression profiling of TPN-supplied liver is to elucidate the real etiologies.

Key words: total parenteral nutrition; liver dysfunction; pathogenesis

World J Pediatr 2007;3(2):104-109


Introduction

In the late 1960s, total parenteral nutrition (TPN) was life-saving for children with chronic bowel obstruction, fistulae, loss of mucosal body surfaces, short bowel syndrome (SBS), and other clinical problems that precluded enteral diet by mouth or tube feeding for a long period of time. TPN as an essential fluid to meet nutritional needs and to avoid progressive starvation-induced malnutrition changed the outcome of patients from dying.[1] Since then, TPN has been an indispensable treatment in clinical practice and a panacea for infants and children who are unable to eat or absorb enteral nutrition.[1-4] As a result, the prognosis for patients with SBS has changed markedly, and the management of infants with congenital gastrointestinal anomalies and gut failure has improved significantly.[5,6] With the superiority of TPN administration in their mind, doctors have recognized the doctrine "a lager volume of intake, a better treatment for condition" until its safety and complications are investigated, especially in patients with SBS who are dependent on a longer duration of TPN support, and have progressive liver dysfunction, even end-stage liver fibrosis.

TPN-associated liver dysfunction

Loff et al[7] analyzed clinical, biochemical and histological data of 10 infants with TPN-induced liver dysfunction, who had been given TPN for at least 8 weeks. They were diagnosed as having necrotizing enterocolitis, gastroschisis, and intrauterine volvulus. Biopsy specimens were taken from each infant at different periods of TPN administration for histological examination of fibrosis, proliferative and inflammatory changes in five portal tracts (Pt) and for evaluation of degenerative changes in hepatocytes. Inflammation was determined by counting inflammatory cells infiltrating the Pt and classifying the cells as neutrophils, eosinophils and monocytes. Proliferative changes were judged by counting bile duct per Pt, and fibrosis was classified into mild proliferation limited to the Pt, moderate fibrosis with enlargement of the Pt, and severe fibrosis with bridging of portal areas. Degenerative changes were differentiated into fatty changes and hydropic degeneration. Biochemically, the levels of bilirubin and aminotransferase increased intermittently and peaked after a minimum of 5 weeks. After 1 and 3 months, TPN induced slight periportal fibrosis, bile duct proliferation, and infiltration. Pronounced intracellular and canalicular cholestasis was observed in addition to extramedullary erythropoiesis and hydropic degeneration. A biopsy after 18 months showed alterations in the intracellular histoarchitecture, mild canalicular cholestasis, and hydropic degeneration. Biopsy specimen taken after 24 months revealed a precirrhotic liver with severe cholestasis and fibrosis with porto-portal bridging and single-cell necrosis.

In an experimental study, 31 rabbits were divided into three groups: 11 were given continuous TPN for 4 weeks, 9 received 20% reduced TPN and had free access to laboratory chow, and 11 enterally-fed animals as controls underwent the same surgical procedure except TPN. The formula of TPN and the indicators to be investigated were identical. In the 3 groups, mild to moderate periportal inflammation and single-cell necrosis occurred to some extent. Cholestasis was not observed in any group. In the rabbits that received infusions of TPN and partial parenteral nutrition (PPN), there was a marked increase in fibrosis and bile duct proliferation between 7 and 28 days shown by biopsy. Severe hydropic degeneration was shown in 90% of animals by the first biopsy after 7 days of TPN administration (Table). An increased level of bilirubin was observed after 4 weeks in the TPN group. Decreased levels of alkaline phosphatase and albumin and a slightly increased level of aminotransferases were noted in all groups. The different biochemical parameters of the 3 groups were not statistically significant. In conclusion, this study revealed that liver damage was caused by TPN solution itself in infants and rabbits. Mok[8] and others successfully produced animal models of TPN-associated liver complications. It has been accepted that TPN can lead to progressive liver dysfunction.


Table. Histological scoring of liver biopsies[7]

Group

Time (n)

Inflammation

Necrotic foci

Bile-duct proliferation

Portal fibrosis

Hydropic degeneration

G0 

t1 (11)

1.2

0.8

1.2

1.3

0.0

G0

t2 (11)

1.2

0.9

1.6

1.7

0.1

G0

t3 (4)

1.3

0.8

1.5

1.7

0.2

G1

t1 (11)

0.9

0.7

1.0

1.2

2.5

G1

t2 (11)

1.3

1.2

2.2

2.5

2.2

G1

t3 (4)

1.3

0.8

2.2

2.4

2.5

G2

t1 (9)

1.2

0.3

1.2

1.3

2.2

G2

t2 (6)

1.5

0.2

1.5

1.3

2.1

G2

t3 (4)

1.5

0.0

1.8

2.0

2.0

Histological grading: 0: absent, 1: mild, 2: moderate, 3: severe; G0: control group, G1: TPN group, G2: PPN group; t1: first liver biopsy on day 7, t2: second liver biopsy on day 21, t3: third liver biopsy on day 28.


Possible mechanisms of TPN-induced liver dysfunction

Alteration of some trace elements in hepatocytes

Since copper is a cofactor for multiple liver enzymes, it is possible to hypothesize the possible role of copper in liver homeostasis in patients on TPN. In other functions, copper serves as a cofactor of antioxidant and free radical, detoxifying enzymes such as superoxide dismutase.[9] A depletion in liver copper concentration may cause a decrease in the activity of enzyme, making hepatocytes more susceptible to oxidative stress and damage. In addition, the lack of copper in hepatocytes of patients with severe liver damage may also be a surrogate marker for a decrease or absence of such trace elements as zinc in patients on TPN. Thus the concentration of zinc in the liver measured by atomic absorption spectrometry is significantly decreased after 10-12 days of TPN administration, which is in parallel with the liver copper concentration in patients supplied with zinc and copper.[10] Zinc is a cofactor of the enzyme known as tissue matrix metalloproteinase (TMMP), which degrades collagen, laminin, enzymatic activity of TMMP secondary to zinc depletion, leading to the accumulation of collagen and extracellular matrix (ECM) in the liver and finally the development of hepatic fibrosis and cirrhosis.[11]

TPN-induced hepatic steatosis and susceptible apoptosis of hepatocytes

In pathological changes of the liver induced by TPN, hepatic steatosis appears to be the earliest and frequent alteration. Two studies[12,13] found that triglyceride accumulation in the liver may occur within a few days after TPN administration in adults, particularly in children. In TPN-fed newborn piglets,[14] hepatic steatosis occurred 1 week after administration of TPN. Thus the affected lipid metabolism such as increased mobilization of depot fat, increased synthesis, impaired transport, and decreased oxidation of fatty acids synergistically resulted in vesicular steatosis of the liver.[15] To assess the effect of steatosis and low viability of hepatocytes in piglets on TPN, markers of apoptosis were investigated in liver tissues. DNA fragmentation and activation of caspase-3 and -7 were seen exclusively in livers of the TPN piglets. Caspase-3 and -7 are recognized as the key executioners of apoptosis and both are partially or totally responsible for the proteolytic cleavage of many key proteins such as nuclear enzyme poly ADP ribose polymerase (PARP).[16] Experimentally, association of steatosis with apoptosis was also confirmed by increased lipid peroxidation and apoptosis of hepatocytes after TPN administration.[17] There are three major pathways, including activation of death receptors (Fas ligand, TNF-), mitochondrial damage, and stress of the endoplasmic reticulum (ER) that culminate in activation of effector caspases, destruction of chromatin, and subsequent death by apoptosis.[18] Increased release of cytochrome C from mitochondria and cleavage of caspase-9 were observed in livers of TPN piglets compared with enteral nutrition (EN) pigs. Caspase-9 mediates apoptotic signals in response to mitochondrial damage and activation of caspase-9 requires cytochrome C.[19] The Bcl-2 family proteins mediate the major mitochondrial-associated apoptotic-signaling pathway. In this family, Bcl-2 is an antiapoptotic member and Bax is one of the proapoptotic members.[20] Proapoptotic proteins of the Bcl-2 family act on mitochondria and facilitate the release of cytochrome C.[21] In this study, downregulation of Bcl-2 and overexpression of Bax took place in TPN livers compared with EN ones. In addition, apoptosis is an active process that requires ATP for its execution and ATP level is a determinant marker of cell apoptosis.[22] In this animal model the adenosine triphosphate (ATP) concentration in livers of TPN piglets was lower than that in EN piglets. Recent studies suggested that TPN could induce apoptosis through the Fas pathway. Fas, a transmembrane receptor protein of the TNF receptor family, contains a death domain that signals via the apoptotic pathway. Caspase-8 is one of the initiator caspases associated with apoptosis involving death receptors. Increased levels of Fas expression and activation of caspase-8 in TPN liver tissues[14] indicated that TPN may cause hepatic apoptosis via both the mitochondrial and death receptor pathways. The death program may be initiated at the cell surface with activation of Fas or TNFR1 (usually involving in receptor trimerization) by their respective ligands, Fas ligand and TNF-, or may result from a primary disturbance of mitochondrial function. In a recent study,[23] the evidence that apoptotic bodies are phagocytosed by stellate cells has suggested the existence of apoptosis in TPN-induced liver dysfunction.

Pathological changes in hepatocytes caused by TPN-associated sepsis and bacterial translocation

In many studies on TPN-induced hepatic dysfunction, inflammatory cell infiltration was regarded as an essential factor in the multifactorial pathogenesis. The impairment of host defence mechanism by TPN solution has been recognized as an important factor in the development of TPN-associated infection. Several studies have revealed an association between TPN and impairment of immune function in both animals and humans.[24-28]

In animal experiments or clinical investigations, liver biopsy specimens harvested after long-term TPN administration showed inflammatory infiltration of different degree at early stage. TPN induced or aggravated sepsis was demonstrated by empirical or clinical studies.[29] TPN-induced hyperglycemia was thought to contribute to dysfunction of neutrophils.[30] Glucose concentrations above 220 mg/dl have been shown to glycosylated immunoglobulins, causing a significant reduction of opsonic activity, which adversely affects immunity. In addition, the mucous membrane of the digestive tract is an important barrier to systemic invasion of bacteria into the blood stream and body tissues. However, bacteria that normally reside within the intestinal tract are able to invade the barrier and associated mesenteric lymphnodes under certain conditions, in a process of "bacterial translocation".[31] The factors facilitating this process include abnormal proliferation of bacteria, attenuation of host immunity, and physical insult to the mucous membrane barrier of the intestinal tract.[32,33] Studying TPN-induced impairment of local immunity of the digestive tract,[34-36] investigators compared the rats on standard TPN and those on free feeding, and found that TPN treatment caused atrophy of intestinal mucosa, a decreased number of Peyer's patches, and a reduction of S-IgA in bile and portal venous blood in addition to hypotrophy and degeneration of the mucous membrane of the digestive tract and a decrease in the number of S-IgA producing plasma cells of the small intestine. The atrophy of Peyer's patches caused a decrease in the number of T-helper and IL-2 producing cells, leading to a suppression of systemic immunity. Besides, the total count of bacteria was significantly higher in the TPN group than in the control group. Translocated bacteria served as pathogens intruding into the liver through the portal vein or bile duct to induce inflammatory changes or other inflammation-related liver injuries via release of cytokines or their endotoxins. The pro-inflammatory cytokine or tumor necrosis factor (TNF) plays a key role in inflammation, proliferation and programmed cell death of hepatocytes. TNF binding to TNF receptor 1 (TNF-R1) leads to the recruitment of TNF-R associated death domain (TRADD), TNF-R associated factor 2 (TRAF2), and receptor interacting protein 1 (R1P1), thus forming complex I.[37] Signaling from the complex I leads to nuclear factor-B (NF-B) activation via activation of the inhibitor of B kinase (IkK) complex. The IkK catalytic subunit of the IkK complex phosphorylates the NF-B-bound IkB, leading to its ubiquitination and subsequent proteasomal degradation. This makes NF-B translocate to the nucleus, where the transcription of genes is induced with an NF-B consensus site in their promoter region. In addition, signaling from the complex I activates the p38 and c-Jun activating (JNK) mitogen-activated protein (MAP) kinase.[38] Recruitment of Fas-associated death domain (FADD) and procaspase-8 results in the formation of the cytosolic complex II, where caspase-8 is activated. Caspase-8 initiates the mitochondrial pathway by cleaving Bid to tBid, which induces mitochondrial permeabilization that results in the release of cytochrome C. Thus an amplification loop results in full-blown caspase activity and subsequent apoptosis.

Effects of oxygen-derived free radical induced lipid peroxidation on hepatic tissues

Oxygen-derived free radicals are highly reactive short-lived chemical species with an unpaired electron. They are produced normally as a result of a number of physiological processes. In the mitochodrial electron transport chain, oxygen is reduced to water via a series of free radical intermediates.[39] Oxidative stress occurs when there is an imbalance between free radical production and antioxidant activity.[40] The double bonds of polyunsaturated fatty acids are particularly vulnerable to oxygen-derived free radical attack, resulting in the process of lipid peroxidation.[41] TPN may result in increased free radical activity by providing (1) substrates (polyunsatured fatty acids), (2) initiators for free radical reactions (carbon centered radicals derived from fatty acids), and (3) catalysts (transition metal ions) for free radical production. These reactive oxygen species (ROSs) can initiate lipid peroxidation by interacting mostly with cellular membrane of hepatocytes having the highest content of polyunsaturated fatty acids in their membranes.[42] The release of ROSs by inflammatory cells and the onset of lipid peroxidation contribute to perpetuation of liver necrosis.[43-45] Previous studies suggested a possible association between lipid peroxidation and liver fibrosis, involving chronic exposure to ethanol,[46] carbon tetrachloride,[47] or iron overload.[48]

Moreover, the concentration of 4-hydroxynonenal (HNE) is able to strongly stimulate the synthesis of procollagen type I by either cultured human[49] or rat liver fat storing cells (FSCs).[50] A similar but less pronounced profibrogenic effect of malondialdehyde (MDA) has been described on rat liver FSC[51] (MDA formed from the breakdown of lipid peroxides and HNE are both sensitive and specific for monitoring oxidative damage). FSCs are recognized as the main source of collagen and other ECM proteins in fibrotic livers.[52] Some study found that lipid peroxidation in the bile duct ligation rat model may stimulate collagen synthesis by proliferation of FSCs.[53] Although the role of lipid peroxidation in TPN-induced liver damage has not been elucidated, the relationship between oxygen-derived free radical induced lipid peroxidation and pathological alterations of the liver may be possible.

Other interpretations for the mechanisms

Bhatia et al[53] noticed that TPN solution is continually exposed to light and that hepatic dysfunction is the most common metabolic aberration associated with TPN. To explore whether light exposure to nutrient mixtures has effect on hepatobiliary responses, they conducted studies to compare the effects of TPN that had been exposed to light (+L) or protected from light (-L) on hepatobiliary function of rats. The results showed that +L rats lost more weight and had lower bile flow, higher taurocholate output in bile, higher biliary osmolarity, and higher inorganic phosphate in bile. Hepatic histology demonstrated scattered foci of necrosis in all +L rats and only one of eight -L rats. These data demonstrated that protection of TPN solutions from light minimizes TPN-associated alterations in hepatobiliary function and histology. They also observed that histological changes in the +L rats were contrary to TPN-induced histological changes reported previously, suggesting a different mechanism.

Moss et al[54] investigated whether TPN administra-tion route is involved in the progressive cholestasis of prepubescent rabbits. No significant structural and functional changes took place in the liver whether TPN was given intravenously or enterally.

To elucidate the mechanism of liver fibrosis caused by hyperalimentation, a typical cell known as Ito cell, has been studied. Ito cell is one of the perisinusoidal-constituent cells that play multiple roles in liver pathophysiology. The function of the cell is to expand from a fat-storing site to a center of extracellular matrix metabolism and mediator production in the liver. The cell can be detected in the perisinusoidal area of the normal livers of human and mammals.[55-58] Ito cell is confirmed primarily to be responsible for the production of liver fibrogenesis in different animal models including extrahepatic bile duct ligation in broiler chickens[59] and congestive liver in rats.[60] The activation, proliferation and transformation of myofibroblastic features of Ito cell are considered to be critical factors involving liver fibrogenesis. Although this mechanism in other animal model with liver injuries has not been duplicated in TPN-induced liver lesion, this investigation would become a hot topic. Since macroarray analysis has been employed to examine the unknown etiologies for some diseases such as biliary atresia,[61] it would be applicable to this unsolved problem.

In conclusion, for patients with SBS or other conditions involved in gut failure, parenteral nutrition support is very important during the whole period of therapy including transplantation. However, total parenteral nutrition-associated liver dysfunction requires for cessation of TPN and restoration of early enteral feeding. The solution to this dilemma seems to become a compulsive duty and a breakthrough in the treatment of these patients. The present multifactorial viewpoint appears to make this problem more complicated, which requires elucidation of the predominant mechanism in subsequent studies.


Funding: None.

Ethical approval: Not needed.

Competing interest: None.

Contributors: HXG wrote this article.


References

1  Kudsk KA. Current aspects of mucosal immunology and its influence by nutrition. Am J Surg 2002;183:390-398.

2  Dudrick SJ. Wilmore DW, Vars HM, Rhoads JE. Long-term total parenteral nutrition with growth, development, and positive nitrogen balance. Surgery 1968;64:134-142.

3  Buchman AL, Moukarzel AA, Bhuta S, Belle M, Ament ME, Eckhert CD, et al. Parenteral nutrition is associated with intestinal morphologic and functional changes in humans. JPEN J Parenter Enteral Nutr 1995;19:453-460

4  Shulman RJ, Phillips S. Parenteral nutrition in infants and children. J Pediatr Gastroenterol Nutr 2003;36:587-607.

5  Chaet MS, Farrell MK, Ziegler MM, Warner BW. Intensive nutritional support and remedial surgical intervention for extreme short bowel syndrome. J Pediatr Gastroenterol Nutr 1994;19:295-298.

6  Suita S, Masumoto K, Yamanouchi T, Nagano M, Nakamura M. Complications in neonates with short bowel syndrome and long-term parenteral nutrition. JPEN J Parenter Enteral Nutr 1999;23(5 Suppl):S106-109.

7  Loff S, Kranzlin B, Moghadam M, Dzakovic A, Wessel L, Back W, et al. Parenteral nutrition-induced hepatobiliary dysfunction in infants and prepubertal rabbits. Pediatr Surg Int 1999;15:479-482.

8  Mok KT. Hepatobiliary complications in healthy, intra-abdominally infected, and high-output fistula rats receiving total parenteral nutrition. JPEN J Parenter Enteral Nutr 1993;17:449-453.

9  Blaszyk H, Wild PJ, Oliveira A, Kelly DG, Burgart LJ. Hepatic copper in patients receiving long-term total parenteral nutrition. J Clin Gastroenterol 2005;39:318-320.

10 Tulikoura I, Vuori E. Effect of total parenteral nutrition on the zinc, copper, and manganese status of patients with catabolic disease. Scand J Gastroenterol 1986;21:421-427.

11 Hamacher S, Matern S, Roeb E. Extracellular matrix〞from basic research to clinical significance. Surgery 2004;129: 1976-1980.

12 Sheldon GF, Peterson SR, Sanders R. Hepatic dysfunction during hyperalimentation. Arch Surg 1978;113:504-508.

13 Cohen C, Olsen MM. Pediatric total parenteral nutrition. Liver histopathology. Arch Pathol Lab Med 1981;105:152-156.

14 Wang H, Khaoustov VI, Krishnan B, Cai W, Stoll B, Burrin DG, et al. Total parenteral nutrition induces liver steatosis and apoptosis in neonatal piglets. J Nutr 2006;136:2547-2552.

15 Zambrano E, El-Hennawy M, Ehrenkranz RA, Zelterman D, Reyes-Mugica M. Total parenteral nutrition induced liver pathology: an autopsy series of 24 newborn cases. Pediatr Dev Pathol 2004;7:425-432.

16 Herceg Z, Wang ZQ. Functions of poly (ADP-ribose) polymerase (PARP) in DNA repair, genomic integrity and cell death. Mutat Res 2001;477:97-110.

17 Tazuke Y, Drongowski RA, Btaiche I, Coran AG, Teitelbaum DH. Effects of lipid administration on liver apoptotic signals in a mouse model of total parenteral nutrition (TPN). Pediatr Surg Int 2004;20:224-248.

18 Ferri KF, Kroemer G. Organelle-specific initiation of cell death pathways. Nat Cell Biol 2001;3:E255-263.

19 Ribeiro PS, Cortez-Pinto H, Sola S, Castro RE, Ramalho RM, Baptista A, et al. Hepatocyte apoptosis, expression of death receptors, and activation of NF-kappaB in the liver of non-alcoholic and alcoholic steatohepatitis patients. Am J Gastroenterol 2004;99:1708-1717.

20 Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309-1312.

21 Zhao Y, Li S, Childs EE, Kuharsky DK, Yin XM. Activation of prodeath Bcl-2 family proteins and mitochondria apoptosis pathway in tumor necrosis factor-alpha-induced liver injury. J Biol Chem 2001;276:27432-27440.

22 Leist M, Single B, Castoldi AF, Kuhnle S, Nicotera P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J Exp Med 1997;185:1481-1486.

23 Cai W, Wu J, Hong L, Xu Y, Tang Q, Shi C. Oxidative injury and hepatocyte apoptosis in total parenteral nutrition-associated liver dysfunction. J Pediatr Surg 2006;41:1663-1668.

24 Shou J, Lappin J, Minnard EA. Total parenteral nutrition, bacterial translocation, and host immune function. Am J Surg 1994;167:145-150.

25 Sedman PC, Somers SS, Ramsden CW, Brennan TG, Guillou PJ. Effects of different lipid emulsions on lymphocyte function during total parenteral nutrition. Br J Surg 1991;78:1396-1399.

26 Fischer GW, Hunter KW, Wilson SR, Mease AD. Diminished bacterial defences with intralipid. Lancet 1980;2:819-820.

27 Kudsk KA, Minard G, Croce MA, Brown RO, Lowrey TS, Pritchard FE, et al. A randomized trial of isonitrogenous enteral diets after severe trauma. An immune-enhancing diet reduces septic complications. Ann Surg 1996;224:531-540.

28 Shou J, Lappin J, Minnard EA, Daly JM. Total parenteral nutrition, bacterial translocation, and host immune function. Am J Surg 1994;167:145-150.

29 Pierro A, van Saene HK, Ponnell SC, Hughes J, Ewan C, Nunn AJ, et al. Microbial translocation in neonates and infants receiving long-term parenteral nutrition. Arch Surg 1996;131:176-179.

30 Okada, Klein NJ, van-Saene HK, Webb G, Holzel H, Pierro A. Bactericidal activity against coagulase-negative staphylococci is impaired in infants receiving long-term parenteral nutrition. Ann Surg 2000;231:276-281.

31 Gore DC, Chinkes D, Heggers J, Herndon DN, Wolf SE, Desai M. Association of hyperglycemia with increased mortality after severe burn injury. J Trauma 2001;51:540-544.

32 Barber AE, Jones WG 2nd, Miner JP, Fahey TJ 3rd, Lowry SF, Shires GT. Bacterial overgrowth and intestinal atrophy in the etiology of gut barrier failure in the rat. Am J Surg 1991;161:300-304

33 Inoue S, Epstein MD, Alexander JW, Trocki O, Jacobs P, Gura P. Prevention of yeast translocation across the gut by a single enteral feeding after burn injury. JPEN J Parenter Enteral Nutr 1989;13:565-571.

34 Alverdy J, Chi HS, Sheldon GF. The effect of parenteral nutrition on gastrointestinal immunity. The importance of enteral stimulation. Ann Surg 1985;202:681-684.

35 Alverdy JC, Aoys E, Moss GS. Total parenteral nutrition promotes bacterial translocation from the gut. Surgery 1988;104:185-190.

36 Illig KA, Ryan CK, Hardy DJ, Rhodes J, Locke W, Sax HC. Total parenteral nutrition-induced changes in gut mucosal function: atrophy alone is not the issue. Surgery 1992;112:631-637.

37 Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 2003;114:181-190.

38 Beyaert R, Van Loo G, Heyninck K, Vandenabeele P. Signaling to gene activation and cell death by tumor necrosis factor receptors and Fas. Int Rev Cytol 2002;214:225-272.

39 Lunec J. Free radicals: their involvement in disease processes. Ann Clin Biochem 1990;27(Pt 3):173-182.

40 Freeman BA, Crapo JD. Biology of disease: free radicals and tissue injury. Lab Invest 1982;47:412-426.

41 Halliwell B, Chirico S. Lipid peroxidation: its mechanism, measurement, and significance. Am J Clin Nutr 1993;57(5 Suppl):715-724.

42 Gabellec MM, Steffan AM, Dodeur M, Durand G, Kirn A, Rebel G. Membrane lipids of hepatocytes, Kupffer cells and endothelial cells. Biochem Biophys Res Commun 1983;113:845-853.

43 Mathews WR, Guido DM, Fisher MA, Jaeschke H. Lipid peroxidation as molecular mechanism of liver cell injury during reperfusion after ischemia. Free Radic Bilo Med 1994;16:763-770.

44 Jaeschke H. Mechanisms of oxidant stress-induced acute tissue injury. Proc Soc Exp Bilo Med 1995;209:104-111.

45 Gressner AM. Liver fibrosis: perspectives in pathobiochemical research and clinical outlook. Eur J Clin Chem Clin Biochem 1991;29:293-311.

46 Dianzani MU. Lipid peroxidation in ethanol poisoning: a critical reconsideration. Alcohol Alcohol 1985;20:161-173.

47 Parola M, Leonarduzzi G, Biasi F, Albano E, Biocca ME, Poli G, et al. Vitamin E dietary supplementation protects against carbon tetrachloride-induced chronic liver damage and cirrhosis. Hepatology 1992;16:1014-1021.

48 Bacon BR, Britton RS. The pathology of hepatic iron overload: a free radical-mediated process? Hepatology 1990;11:127-137.

49 Parola M, Pinzani M, Casini A, Albano E, Poli G, Gentilini A, et al. Stimulation of lipid peroxidation or 4-hydroxynonenal treatment increases procollagen alpha 1 (I) gene expression in human liver fat-storing cells. Biochem Biophys Res Commun 1993;194:1044-1050.

50 Deleeuw AM, Mccarthy SP, Geerts A, Knook DL. Purified rat liver fat-storing cells in culture divide and contain collagen. Hepatology 1984;4:392-403.

51 Combettes L, Dumont M, Berthon B, Erlinger S, Claret M. Release of calcium from the endoplasmic reticulum by bile acids in rat liver cells. J Biol Chem 1988;263:2299-2303.

52 Parola M, Leonarduzzi G, Robino G, Albano E, Poli G, Dianzani MU. On the role of lipid peroxidation in the pathogenesis of liver damage induced by long-standing cholestasis. Free Radic Biol Med 1996;20:351-359.

53 Bhatia J, Moslen MT, Haque AK, McCleery R, Rassin DK. Total parenteral nutrition-associated alterations in hepatobiliary function and histology in rats: is light exposure a clue? Pediatr Res 1993;33:487-492

54 Moss RL, Das JB, Ansari G, Raffensperger JG. Hepatobiliary dysfunction during total parenteral nutrition is caused by infusate, not the route of administration. J Pediatr Surg 1993;28:391-396.

55 Hines JE, Johnson SJ, Burt AD. In vivo responses of macrophages and perisinusoidal cells to cholestatic liver injury. Am J Pathol 1993;142:511-518.

56 Neubauer K, Knittel T, Aurisch S, Fellmer P, Ramadori G. Glial fibrillary acidic protein〞a cell type specific marker for Ito cells in vivo and in vitro. J Hepatol 1996;24:719-730.

57 Trim N, Morgan S, Evans M, Issa R, Fine D, Afford S, et al. Hepatic stellate cells express the low affinity nerve growth factor receptor p75 and undergo apoptosis in response to nerve growth factor stimulation. Am J Pathol 2000;156:1235-1243.

58 Wake K. "Sternzellen" in the liver: perisinusoidal cells with special reference to storage of vitamin A. Am J Anat 1971;132:429-462.

59 Handharyani E, Ochiai K, Iwata N, Umemura T. Immunohistochemical and ultrastructural study of ito cells (fat-storing cells) in response to extrahepatic bile duct ligation in broiler chickens. J Vet Med Sci 2001;63:547-552.

60 Akiyoshi H, Terada T. Centrilobular and perisinusoidal fibrosis in experimental congestive liver in the rat. J Hepatol 1999;30:433-439.

61 Choe BH, Kim KM, Kwon S, Lee KS, Koo JH, Lee HM, et al. The pattern of differentially expressed genes in biliary atresia. J Korean Med Sci 2003;18:392-396.

Received July 11, 2006; Accepted after revision November 26, 2006

 

 
  [Articles Comment]

  title Author The End Revert Time Revert / Count

  Username:
  Comment Title: 
 
   

 

     
 
     
World Journal of Pediatric Surgery

roger vivier bags 美女 美女

Home  |  Journal Information  |  Current Issue  |  Past Issues  |  Journal Information  |  Contact Us
Children's Hospital, Zhejiang University School of Medicine, China
Copyright 2007  www.wjpch.com  All Rights Reserved Designed by eb