Experimental models of hepatic encephalopathy: ISHEN guidelines

Share Embed


Descripción

Liver International ISSN 1478-3223

6R E V I E W A R T I C L E

Experimental models of hepatic encephalopathy: ISHEN guidelines Roger F. Butterworth1, Michael D. Norenberg2, Vicente Felipo3, Peter Ferenci4, Jan Albrecht5 and Andres T. Blei6 [Members of the ISHEN Commission on Experimental Models of HE] 1 Neuroscience Research Unit, St-Luc Hospital (CHUM), University of Montreal, Montreal, QC, Canada 2 Department of Pathology, University of Miami School of Medicine, Miami, FL, USA ´ Pr´ıncipe Felipe, Valencia, Spain 3 Laboratory of Neurobiology, Centro de Investigacion 4 Department of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria 5 Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland 6 Department of Medicine, Northwestern University, Evanston, IL, USA

Keywords acute liver failure – ammonia – animal models – astrocytes – azoxymethane – bile duct ligation – cirrhosis – galactosamine – hepatic encephalopathy – portacaval anastomosis – thioacetamide

Correspondence Dr Roger F. Butterworth, Neuroscience Research Unit, Hoˆpital St-Luc (CHUM), University of Montreal, 1058, St-Denis, Montreal, QC, Canada H2X 3J4 Tel: +1 514 890 8000 ext. 35740 Fax: +1 514 412 7253 e-mail: [email protected] Received 10 December 2008 Accepted 4 February 2009 DOI:10.1111/j.1478-3231.2009.02034.x

Abstract Objectives of the International Society for Hepatic Encephalopathy and Nitrogen Metabolism Commission were to identify well-characterized animal models of hepatic encephalopathy (HE) and to highlight areas of animal modelling of the disorder that are in need of development. Features essential to HE modelling were identified. The best-characterized animal models of HE in acute liver failure, the so-called Type A HE, were found to be the hepatic devascularized rat and the rat with thioacetamide-induced toxic liver injury. In case of chronic liver failure, surgical models in the rat involving end-to-side portacaval anastomosis or bile duct ligation were considered to best model minimal/mild (Type B) HE. Unfortunately, at this time, there are no satisfactory animal models of Type C HE resulting from end-stage alcoholic liver disease or viral hepatitis, the most common aetiologies encountered in patients. The commission highlighted the urgent need for such models and of improved models of HE in chronic liver failure in general as well as a need for models of post-transplant neuropsychiatric disorders. Studies of HE pathophysiology at the cellular and molecular level continue to benefit from in vitro and or ex vivo models involving brain slices or exposure of cultured cells (principally cultured astrocytes) to toxins such as ammonia, manganese and pro-inflammatory cytokines. More attention could be paid in the future to in vitro models involving the neurovascular unit, microglia and neuronal co-cultures in relation to HE pathogenesis.

Members of the commission were invited by the International Society for Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN) President (Dr Piero Amodio, Padua, Italy) to develop recommendations for animal models of hepatic encephalopathy (HE) with a view to standardization of procedures and a move towards a consensus on the best-characterized models available at the time of writing this document and to identify areas of animal modelling that are in need of development. An early decision was taken to include only models of Type A, B and C HE as defined by the Vienna consensus document on classification of HEs (Table 1). Other liver disorders such as Wilson’s disease, urea cycle enzymopa-

These guidelines were prepared by the Commission on Experimental Models of Hepatic Encephalopathy appointed by the International Society for Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN). The content was discussed and approved in the 13th ISHEN Symposium, Padova, Italy, 28 April to 1 May 2008.

Liver International (2009) 2009 John Wiley & Sons A/S

c

thies and Reye syndrome that frequently result in cerebral dysfunction were not included. A first draft of the consensus document was prepared based on an update of review articles on animal models of HE by Blei et al. (1) and Chamuleau (2) and the document was circulated to commission members by email for comments and modifications. Dr Norenberg was requested to provide a concise assessment of the importance of cell culture models. Dr Albrecht was requested to cover models based on brain slice preparations. An iterative procedure followed whereby commission members were invited to make further corrections. A revised document was presented by Dr Butterworth at the ISHEN meeting in Padua in April 2008 where all participants were invited to submit further corrections/ comments leading to the present document. Hepatic encephalopathy is a neuropsychiatric disorder that results from impaired liver function. In humans, HE occurs in one of the three forms that have recently been reclassified (Table 1).

783

Experimental models of hepatic encephalopathy

Table 1. Classification of hepatic encephalopathy syndromes Type A

Type B Type C

Hepatic encephalopathy (HE) associated with acute liver failure (ALF) resulting from severe inflammatory and/or necrotic liver disease of rapid onset. HE resulting from portacaval shunting in the absence of parenchymal liver disease. HE that accompanies chronic liver failure (cirrhosis). Aetiology of cirrhosis resulting from alcohol, viral infection, biliary obstruction, drugs or toxins. Portal hypertension arises as a result of cirrhosis; the high portal pressure stimulates the opening of embryonic venous channels. Portal-systemic shunts then permit toxins of intestinal origin (examples include ammonia, manganese, cytokines) to bypass the liver into the systemic circulation.

Studies of the pathogenesis of a wide range of human neuropsychiatric disorders including neurodegenerative diseases (e.g. Alzheimer disease, Parkinson disease), stroke, epilepsy and psychiatric disease (e.g. psychosis, depression) have been significantly enhanced by the use of animal models. Pathophysiological mechanisms involved in metabolic encephalopathies such as vitamin deficiencies and HE have also benefited from the use of animal models. However, HE has proven difficult to model since liver diseases in humans have many varied aetiologies (alcoholic, viral, toxic, autoimmune, ischaemic, genetic), involve varying degrees of portal-systemic shunting, damage to other end-organs and in the majority of cases are associated with precipitating factors such as a gastrointestinal bleed, infection, protein loading, hypoglycaemia, hyponatraemia, use of sedatives, etc. It is not possible to reproduce all (or even many) of these aspects of HE. It is therefore essential to choose an animal model that is appropriate to the research objectives and the measurements to be undertaken. There are currently no satisfactory animal models of HE resulting from alcoholic cirrhosis, viral hepatitis or acetaminophen hepatotoxicity, the three major aetiologies encountered in human HE. Choice of animal species

A wide variety of animal species have been used for the study of HE including large animals (dogs, goats, pigs, rabbits) and rodents (rats, mice). Large animals have the advantages of facilitating repeated sampling of body fluids, biopsies, the use of techniques such as visual/ auditory evoked potentials and electroencephalography (EEG). More material is available from large animals and assessment of neuropsychiatric status may be facilitated. Specific research questions (e.g. evaluation of liver support systems for HE) benefit from the use of large animals. In spite of these multiple advantages, relatively few investigators now use large animals. Reasons for this include the costs of animals, animal housing, materials and salaries for personnel. Heightened ethical concerns may also be an issue.

784

Butterworth et al.

Table 2. Animal models of Type A hepatic encephalopathy Species used Anhepatic models Hepatic devascularization Hepatectomy Portacaval anastomosis1ammonia Hepatotoxin models Galactosamine Acetaminophen Thioacetamide Azoxymethane

Rat, rabbit, pig Rat, pig Rat Rat, rabbit, guinea pig Rat, dog, pig Rat Mouse

A review of the recent literature reveals that the laboratory rat and, increasingly, the mouse are the most common species currently used for the studies in HE. Reasons for this include the following: (i) Availability of databases (molecular studies), atlases (anatomical studies) and a wide literature on neurobehavioral, pathological, biochemical methods and findings. (ii) The mouse and rat genomes are now fully cloned and characterized. There is an urgent need for more mouse models of HE for molecular genetic studies. (iii) Antibodies and molecular probes are often commercially available. (iv) Relatively low costs of animals and animal housing. Animal models of hepatic encephalopathy in acute liver failure (Type A hepatic encephalopathy)

Features proposed as essential for an animal model of HE in acute liver failure (ALF) include the following: (i) Reproducible clinical picture to facilitate staging of encephalopathy. (ii) Progression of symptoms to include brain oedema and its complications (intracranial hypertension, brain herniation). (iii) Potential for reversibility. (iv) Hyperammonaemia/increased brain ammonia, glutamine. (v) Well-characterized hepatic and brain pathology. (vi) Minimal hazard to personnel from toxins and infectious agents. Few models satisfy all these criteria. All animal models of ALF lead to hypothermia, hypoglycaemia and other systemic complications. It is therefore of paramount importance to control temperature and glycaemia routinely and effectively and to monitor other organ systems and provide supportive care. In view of the absence of animal models of ALF related to viral hepatitis, animal models of ALF have been developed principally along two lines, namely exclusion of the liver (anhepatic models) from the circulation or administration of a hepatotoxin (Table 2). Anhepatic models of Type A hepatic encephalopathy

In hepatic devascularization, the liver is left in situ and its afferent circulation diverted (portacaval anastomosis)

c

Liver International (2009) 2009 John Wiley & Sons A/S

Butterworth et al.

and interrupted (hepatic artery ligation). In total hepatectomy, the liver is removed generally as a two-stage operation and the splanchnic circulation is rerouted. The course of HE after either hepatic devascularization or hepatectomy is relatively rapid. Neither procedure manifests a potential for recovery. Swelling of astrocyte foot processes has been described in both devascularized (3) and hepatectomized (4) rats. A progressive increase in intracranial pressure has been noted in hepatic devascularized rats where EEG changes are similar to those described in ALF in humans. A model of ammonia-precipitated brain oedema in the context of liver failure uses end-to-side portacaval anastomosis followed 24 h later by ammonia infusions (5). Hepatic devascularized rats manifest a reproducible progression of encephalopathy and brain oedema and have been used extensively for studies on the pathogenesis and treatment of ALF. Animals are hyperammonaemic with brain ammonia in the 1–5 mM range (6). The model has been applied to studies of brain metabolism (7), neurotransmission abnormalities, gene expression and brain inflammation in ALF. Brain oedema and HE in hepatic devascularized animals respond to hypothermia, ammonia-lowering agents and anti-inflammatory drugs.

Experimental models of hepatic encephalopathy

rabbit. Severity of HE and mortality are variable (13). Dixit and Chang (14) described cerebral cortical and cerebellar oedema together with swelling of pericapillary astrocytes. Brain tissue necrosis and breakdown of the blood–brain barrier have also been reported (15), features that are not routinely observed in human ALF. The thioacetamide-treated rat

Thioacetamide treatment results in hepatocellular necrosis, bridging necrosis and lymphocytic infiltrate without cholestasis (9). Rats develop renal failure, hypoglycaemia, hypotension and weight loss if supportive fluids, glucose and external heating are withheld (16). The blood–brain barrier is intact in this model. The thioacetamide model has been used to delineate changes in neuromodulatory functions of astrocytes in relation to progression of HE (17). The azoxymethane-treated mouse

This relatively new model has been characterized from the liver and brain standpoints (18), and used for studies of HE mechanisms involving gene manipulations. Mice rapidly develop encephalopathy and brain oedema.

Toxin models of Type A hepatic encephalopathy

A range of hepatotoxic substances has been used to create ALF in experimental animals including galactosamine, acetaminophen, thioacetamide and azoxymethane. Other toxins known to lead to ALF include phosphorus, nitrosamines and CCl4 but HE resulting from these latter toxins has not been well characterized. The CCl4 model is useful in demonstrating astrocytic response at the level of RNA synthesis (8). Galactosamine is directly hepatotoxic whereas acetaminophen and thioacetamide must be converted to toxic metabolites in the liver via the microsomol P450 system. The pathological nature of the hepatitis produced by each of these toxins varies; bridging necrosis and a lymphocytic inflammatory infiltrate have been described in the thioacetamide rat (9) but not in the galactosamine rabbit (10). Massive centrolobular necrosis is the pathological hallmark of acetaminophen-induced liver injury. The galactosamine-treated rabbit

Traber et al. (11) reported marked swelling of the pericapillary astrocyte foot processes and swelling of processes surrounding axons and dendrites. Cerebral oedema was restricted to grey matter. Neurons appeared normal; the blood–brain barrier remained grossly intact but permeability to a-aminoisobutyric acid (normally impermeable) was increased in basal ganglia (12). This model has been used for evoked potential studies. The galactosamine-treated rat

This model is not as well characterized from the hepatic and brain standpoints as the galactosamine-treated Liver International (2009) 2009 John Wiley & Sons A/S

c

Acetaminophen-treated animals

Acetaminophen administration to dogs has been used as a model for evaluation of artificial liver support systems (19). The rat model of ALF because of acetaminophen results in variable HE and brain oedema. Neuropathology has not been performed in this model. Animal models of hepatic encephalopathy in chronic liver impairment

Features considered essential for an animal model of HE in chronic liver impairment (Type B and C HE) include the following: (i) Chronic liver disease with portal-systemic shunting. (ii) Range of symptoms of encephalopathy from minimal hepatic encephalopathy (MHE) to coma. (iii) Alzheimer Type II astrocytosis at coma stages of encephalopathy. (iv) Hyperammonaemia, increased brain ammonia/ glutamine. (v) Precipitating factor. (vi) Clinical response to established treatments. Animal models of hepatic encephalopathy based on portal-systemic shunting (Type B hepatic encephalopathy)

Portacaval anastomosis has been performed in rats, rabbits, dogs and pigs (Table 3). Plasma ammonia correlates with EEG changes and neurological status in dogs with portacaval anastomoses (20). Attempts to

785

Experimental models of hepatic encephalopathy

Table 3. Animal models of Type B (and C) hepatic encephalopathy Species used Portacaval anastomosis Congenital portacaval shunts Graded portal vein stenosis Portacaval anastomosis1ammonia Bile duct ligation

Rat, dog, rabbit, pig Dogs, cats Rat Rat Rat

Manifests some aspects of Type C hepatic encephalopathy [see ‘Animal

models of hepatic encephalopathy in acute liver failure (Type A hepatic encephalopathy)’].

perform portacaval anastomosis in rabbits may lead to high mortality (21). Dogs and cats with congenital portacaval shunts offer naturally occurring models (Table 3). Dogs develope psychomotor dysfunction, motor signs and hyperammonaemia, reduced hepatic function (22) and are susceptible to high protein diets. Graded portal-vein stenosis affords a model of MHE in the rat. This procedure is easier to perform than the end-to-side portacaval anastomosis and leads to loss of activity, altered day–night rhythms, hyperammonaemia and increased brain ammonia/glutamine. Side-to-side portacaval anastomosis in the rat, although challenging from a surgical standpoint, offers a potential for reversibility (23). Hepatic encephalopathy resulting from portacaval anastomosis in the rat

End-to-side portacaval anastomosis in the rat affords a popular model of Type B HE. Liver dysfunction is limited to liver atrophy and loss of perivenous hepatocytes. This model is widely used to study the effects of portacaval shunting and reduced hepatic capacity on brain function. Stenosis may occur leading to variable results; liver/body weight ratio and testicular weight may be used as a surrogate index of degree of stenosis. Pair feeding of control animals may be necessary with studies of longterm shunted animals to control for anorexia and decreased food intake (24). Shunted animals manifest hyperammonaemia and increased brain ammonia/glutamine, alterations of day–night and circadian rhythms, hypokinesia, reduced memory and learning ability, altered reflexes and testicular atrophy. Altered aromatic/ branched chain amino acid ratios, decreased brain glucose utilization, oxidative/nitrosative stress and altered multiple neurotransmitter function similar to MHE in human cirrhotic patients have been reported. Portacaval shunted rats are sensitive to ammonia administration, which leads to severe encephalopathy (coma) (25, 26). Animal models of hepatic encephalopathy resulting from decompensated liver cirrhosis (Type C hepatic encephalopathy)

An optimal animal model of Type C HE does not exist at the present time. Alcohol administration does not

786

Butterworth et al.

produce micronodular cirrhosis in the small laboratory animal. A common procedure used to produce cirrhosis in experimental animals makes use of the hepatotoxin CCl4. Monitoring makes use of a daily body weight method. Problems include inconsistent lesions animal to animal. The presence of ascites may limit neurobehavioral assessment. Obstruction of the common bile duct induces a reproducible model of biliary cirrhosis in rats (Table 3). In contrast to portacaval anastomosis, bile duct-ligated (BDL) animals have liver failure, developing jaundice, portal hypertension (27), portal-systemic shunting (28), bacterial translocation and immune system dysfunction (29). BDL rats are hyperammonaemic (30) but show only low-grade encephalopathy (decreased locomotor activities) (31). However, BDL rats are infected, they do not eat and lose weight and pair feeding of control groups may be necessary in order to avoid confounds owing to diminished food intake (32). Feeding ammonium salts to BDL rats provides a model of acute-on-chronic HE, which reproduces the human neuropathology (Alzheimer Type II astrocytosis) and alterations of brain osmolytes that are characteristic of human Type C HE as well as low-grade brain oedema, inflammation and deficits of motor coordination (32). Animal models of pure acute/chronic hyperammonia

These models, generally limited to rats and mice, are designed to study the effects of hyperammonia per se (in the absence of liver dysfunction) on brain function. Methods include the feeding of high-ammonia diets or urease. These models have been successfully used to demonstrate alterations of multiple neurotransmitter systems in brain (e.g. glutamate receptor-mediated signal transduction) (33, 34). Hyperammonaemic animals manifest disturbances in learning and memory. Experiments are inexpensive and simple to perform. Use of cultured neural cells for the study of hepatic encephalopathy mechanisms

These studies are valuable in the study of the effects of exposure to particular neural cells to toxic substances that are known to be increased in brain in acute or chronic liver failure. Toxins studied so far include ammonia, manganese, octanoic acid and cytokines. The majority of studies to date have made use of primary cultures of rat cortical astrocytes (35). Some studies use C-6 glioma cells. Few studies so far have made use of cultures of other neural cells (neurons, cerebrovascular endothelial cells, microglia) or cocultures. Studies using these latter systems are greatly encouraged. Cultured astrocytes exposed to ammonia manifest many of the pathological, metabolic and neurochemical changes observed in human HE (35). Cultured neurons exposed to ammonia reproduce alterations in signal

c

Liver International (2009) 2009 John Wiley & Sons A/S

Butterworth et al.

transduction observed in human HE (36). Advantages of the cultured cell approach are its low cost, excellent control of the cellular environment and reproducibility. The use of cell cultures offers a unique opportunity for unravelling complex pathogenetic pathways. Disadvantages include potential differences in cellular/molecular properties between cultured cells and their in situ counterparts, particularly regarding expression of certain proteins. Extrapolation of findings to the in vivo situation should be done with caution. There is a need for cell culture models of the neurovascular unit for the study of its role in HE mechanisms. Use of brain slices for the study of hepatic encephalopathy mechanisms

Brain slices have classically been used to study mechanisms in a wide range of experimental neurological disorders. Brain slices have a relatively well-preserved cytoarchitecture permitting the study of cell–cell interactions. In vitro experiments make use of brain slices from normal animals exposed to toxins implicated in HE (e.g. ammonia). Ex vivo experiments make use of brain slices from animals with HE resulting from experimental acute or chronic liver failure. Disadvantages of brain slices are their relatively short-lived viability (l–2 h). Several investigators have used brain slices to study brain oedema mechanisms (37), astrocyte neuropathology (38), longterm potentiation (39), nerve cell damage (40) and cell signalling mechanisms (41) in relation to toxin exposure in vitro and ex vivo in slices from animal models of HE. Recommendations of the commission

It is important to bear in mind that the recommendations of the commission are based on available information reported in the biomedical literature at the time of the Padua ISHEN meeting in 2008 and that, by their very nature, these recommendations are subject to revision as further models of HE become characterized and existing models undergo modifications. As stated in this review, there are no satisfactory animal models of HE resulting from alcoholic cirrhosis, viral hepatitis or acetaminophen hepatotoxicity, the three most common aetiologies encountered in patients. With the above provisos, the commission makes the following recommendations: 1. Well-characterized animal models of HE in ALF (Type A HE) that meet the majority of criteria are: (a) The hepatic devascularized rat. (b) The rat with thioacetamide-induced toxic liver injury. 2. Well-characterized animal models of HE in chronic liver impairment that meet the majority of criteria are: (a) The rat with end-to-side portacaval anastomosis (Type B HE) (MHE). (b) The BDL rat (some aspects of Type C HE). 3. There is a need for improved animal models of Type C HE in rodents. Liver International (2009) 2009 John Wiley & Sons A/S

c

Experimental models of hepatic encephalopathy

4. There is a need for improved mouse models of all types of HE in the mouse to facilitate studies of the molecular genetics of HE. 5. There is a need for models of post-orthotopic liver transplantation neurological/psychiatric disorders. 6. Studies of molecular and cellular mechanisms related to HE may be supplemented by studies in: (a) Hyperammonaemic animals (with intact liver function) in order to assess the role of ammonia in vivo (per se). (b) Cultured neural cells (particularly glial cells) exposed to pathophysiologically relevant concentrations of toxins (e.g. ammonia, manganese, cytokines) known to accumulate in brain in liver failure. (c) Brain slice preparations from normal animals exposed in vitro to toxins or ex vivo slice preparations from HE animal models. 7. There is a need for studies in cultured cerebrovascular endothelial cells, microglia, neurons and mixed cultures in order to study cell–cell interactions and the role of the neurovascular unit in HE. 8. It is proposed that efforts be made to standardize procedures in order to limit variability of the preparations. This could be achieved, for example, by exchange of research trainees between laboratories. References 1. Blei AT, Omary R, Butterworth RF. Animal models of hepatic encephalopathy. In: Boulton AA, Baker GB, Butterworth RF, eds. Neuromethods, Vol. 22. Totowa, NJ, USA: Humana Press, 1992; 183–222. 2. Chamuleau RA. Animal models of hepatic encephalopathy. Semin Liver Dis 1996; 16: 265–70. 3. Traber P, Dal Canto M, Ganger D, Blei AT. Effect of body temperature on brain edema and encephalopathy in the rat after hepatic devascularization. Gastroenterology 1989; 96: 885–91. 4. Potvin M, Morrison H, Finlayson E, et al. Cerebral abnormalities in hepatectomized rats with acute hepatic coma. Lab Invest 1984; 50: 560–4. 5. Chung C, Gottstein J, Blei AT. Indomethacin prevents the development of experimental ammonia-induced brain edema in rats after portacaval anastomosis. Hepatology 2001; 34: 249–54. 6. Swain M, Butterworth RF, Blei AT. Ammonia and related amino acids in the pathogenesis of brain edema in acute ischemic liver failure in rats. Hepatology 1992; 15: 449–53. 7. Chatauret N, Rose C, Therrien G, Butterworth RF. Mild hypothermia prevents cerebral edema and CSF lactate accumulation in acute liver failure. Metab Brain Dis 2001; 16: 95–102. 8. Albrecht J. Cerebral RNA synthesis in experimental hepatogenic encephalopathy. J Neurosci Res 1981; 6: 553–8. 9. Zimmerman C, Ferenci P, Pifl C, et al. Hepatic encephalopathy in thioacetamide-induced acute liver failure in rats: characterization of an improved model and study of

787

Experimental models of hepatic encephalopathy

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21. 22.

23.

24.

788

amino acid-ergic neurotransmission. Hepatology 1989; 9: 594–601. Keppler D, Lesch R, Reutter W, Decker K. Experimental hepatitis induced by D-galactosamine. Exp Mol Pathol 1968; 9: 279–90. Traber PG, Dal Canto M, Ganger D, Blei AT. Electron microscopic evaluation of brain edema in rabbits with galactosamine-induced fulminant hepatic failure. Hepatology 1987; 7: 1272–7. Horowitz ME, Schafer DF, Molnar P. Increased blood–brain transfer in a rabbit model of acute liver failure. Gastroenterology 1983; 84: 1003–11. Mullen KD, Schafer DF, Cuchi P, et al. Evaluation of the suitability of galactosamine-induced fulminant hepatic failure as a model of hepatic encephalopathy in the rat and the rabbit. In: Soeters PB, Wilson JHP, Meijer AJ, Holm E, eds. Advances in Ammonia Metabolism and Hepatic Encephalopathy. Amsterdam, the Netherlands: Elsevier, 1988; 205–11. Dixit V, Chang TMS. Brain edema and the blood brain barrier in galactosamine-induced fulminant hepatic failure rats. ASAIO Trans 1990; 36: 21–7. Zaki AEO, Ede RJ, Davis M, Williams R. Experimental studies of blood brain barrier permeability in acute hepatic failure. Hepatology 1984; 4: 359–63. Geller D, Gammal SH, Mullen KD, Jones EA. An improved rat model of hepatic encephalopathy due to fulminant hepatic failure: the importance of supportive therapy. In: Soeters PB, Wilson JHP, Meijer AJ, Holm E, eds. Advances in Ammonia Metabolism and Hepatic Encephalopathy. Amsterdam, the Netherlands: Elsevier, 1988; 213–7. Albrecht J, Hilgier W, Łazarewicz JW, et al. Astrocytes in acute hepatic encephalopathy: metabolic properties and transport functions. In: Norenberg MD, et al., eds. Biochemical Pathology of Astrocytes. New York, NY, USA: R. Liss, 1988; 465–76. Belanger M, Coˆt´e J, Butterworth RF. Neurobiological characterization of an azoxymethane mouse model of acute liver failure. Neurochem Int 2006; 48: 434–40. Francavilla A, Makowka L, Polimeno L, et al. A dog model for acetaminophen-induced fulminant hepatic failure. Gastroenterology 1989; 96: 470–8. Okamoto H, Fujimura T, Yashiro K. Correlation between electroencephalogram, hepatic encephalopathy grade and biochemical indices in beagles with portacaval anastomosis. J Parent Ent Nutr 1985; 9: 326–33. van Niekerk JLM, Koopman JP. Portacaval shunt in the rabbit, a model for encephalopathy. Z Versuchstierkd 1986; 28: 7–9. Maddison JE, Dodd PR, Morrison M, et al. Plasma GABA, GABA-like activity and the brain GABA–benzodiazepine receptor complex in rats with chronic hepatic encephalopathy. Hepatology 1987; 7: 621–8. Hawkins PA, Dejoseph MR, Hawkins RA. Eliminating metabolic abnormalities of portacaval shunting by restoring normal liver blood flow. Am J Physiol 1996; 270: E1037–42. Dejong CH, Deutz NE, Soeters PB. Cerebral cortex ammonia and glutamine metabolism in two rat models of chronic liver insufficiency-induced hyperammonemia: influence of pair-feeding. J Neurochem 1993; 60: 1047–57.

Butterworth et al.

25. Hindfelt B, Plum F, Duffy TE. Effects of acute ammonia intoxication on cerebral metabolism in rats with portacaval shunts. J Clin Invest 1977; 59: 386–96. 26. Butterworth RF, Girard G, Gigue`re JF. Regional differences in the capacity for ammonia removal by brain following portacaval anastomosis. J Neurochem 1988; 51: 486–90. 27. Kountouras J, Billing BH, Scheuer PJ. Prolonged bile duct obstruction: a new experimental model for cirrhosis in the rat. Br J Exp Pathol 1984; 65: 305–11. 28. Dunn CW, Horton JW, Megison SM, Vuitch MF. Contribution of portal systemic shunt to Kupffer cell dysfunction in obstructive jaundice. J Surg Res 1991; 50: 234–9. 29. Greve JW, Gouma DJ, Soeters PB, Buurman WA. Suppression of cellular immunity in obstructive jaundice is caused by endotoxins: a study with germ-free rats. Gastroenterology 1990; 98: 478–85. 30. Rodrigo R, Jover R, Candela A, et al. Bile duct ligation plus hyperammonemia in rats reproduces the alterations in the modulation of soluble guanylate cyclase by nitric oxide in brain of cirrhotic patients. Neuroscience 2005; 130: 435–43. 31. Chan CY, Huang SW, Wang TF, et al. Lack of detrimental effects of nitric oxide inhibition in bile duct-ligated rats with hepatic encephalopathy. Eur J Clin Invest 2004; 34: 122–8. 32. Jover R, Rodrigo R, Felipo V, et al. Brain edema and inflammatory activation in bile duct ligated rats and dietinduced hyperammonemia: a model of hepatic encephalopathy in cirrhosis. Hepatology 2006; 43: 1257–66. 33. Felipo V, Butterworth RF. Neurobiology of ammonia. Prog Neurobiol 2002; 67: 259–79. 34. Llansola M, Rodrigo R, Monfort P, et al. NMDA receptors in hyperammonemia and hepatic encephalopathy. Metab Brain Dis 2007; 22: 321–35. 35. Norenberg MD. Role of astrocytes in hepatic encephalopathy. Neurochem Pathol 1987; 6: 13–33. 36. Rodrigo R, Erceg S, Felipo V. Neurons exposed to ammonia reproduce the differential alteration in nitric oxide modulation of guanylate cyclase in cerebellum and cortex of patients with liver cirrhosis. Neurobiol Dis 2005; 19: 150–61. 37. Ganz R, Swain M, Traber P, et al. Ammonia-induced swelling of rat cerebral cortical slices: implications for the pathogenesis of brain edema in acute hepatic failure. Metab Brain Dis 1989; 4: 213–23. 38. Zieli˜nska M, Law RO, Albrecht J. Excitotoxic mechanism of cell swelling in rat cerebral cortical slices treated acutely with ammonia. Neurochem Int 2003; 43: 299–303. 39. Monfort P, Erceg S, Piedrafita B, et al. Chronic liver failure in rats impairs glutamatergic synaptic transmission and long-term potentiation in hippocampus and learning ability. Eur J Neurosci 2007; 25: 2103–11. 40. Pidoplichko V, Dani J. Acid-sensitive ionic channels in midbrain dopamine neurons are sensitive to ammonium, which may contribute to hyperammonemia damage. Proc Natl Acad Sci USA 2006; 103: 11376–80. 41. Rodrigo R, Erceg S, Rodriguez-Diaz J, et al. Glutamateinduced activation of nitric oxide synthase is impaired in cerebral cortex in vivo in rats with chronic liver failure. J Neurochem 2007; 102: 51–64.

c

Liver International (2009) 2009 John Wiley & Sons A/S

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.