Skip to main content
Top
Published in: Journal of Inherited Metabolic Disease 4/2013

01-07-2013 | Review

Ammonia toxicity to the brain

Authors: Olivier Braissant, Valérie A. McLin, Cristina Cudalbu

Published in: Journal of Inherited Metabolic Disease | Issue 4/2013

Login to get access

Abstract

Hyperammonemia can be caused by various acquired or inherited disorders such as urea cycle defects. The brain is much more susceptible to the deleterious effects of ammonium in childhood than in adulthood. Hyperammonemia provokes irreversible damage to the developing central nervous system: cortical atrophy, ventricular enlargement and demyelination lead to cognitive impairment, seizures and cerebral palsy. The mechanisms leading to these severe brain lesions are still not well understood, but recent studies show that ammonium exposure alters several amino acid pathways and neurotransmitter systems, cerebral energy metabolism, nitric oxide synthesis, oxidative stress and signal transduction pathways. All in all, at the cellular level, these are associated with alterations in neuronal differentiation and patterns of cell death. Recent advances in imaging techniques are increasing our understanding of these processes through detailed in vivo longitudinal analysis of neurobiochemical changes associated with hyperammonemia. Further, several potential neuroprotective strategies have been put forward recently, including the use of NMDA receptor antagonists, nitric oxide inhibitors, creatine, acetyl-L-carnitine, CNTF or inhibitors of MAPKs and glutamine synthetase. Magnetic resonance imaging and spectroscopy will ultimately be a powerful tool to measure the effects of these neuroprotective approaches.
Literature
go back to reference Aguilar MA, Minarro J, Felipo V (2000) Chronic moderate hyperammonemia impairs active and passive avoidance behavior and conditional discrimination learning in rats. Exp Neurol 161:704–713PubMedCrossRef Aguilar MA, Minarro J, Felipo V (2000) Chronic moderate hyperammonemia impairs active and passive avoidance behavior and conditional discrimination learning in rats. Exp Neurol 161:704–713PubMedCrossRef
go back to reference Agusti A, Cauli O, Rodrigo R, Llansola M, Hernandez-Rabaza V, Felipo V (2011) p38 MAP kinase is a therapeutic target for hepatic encephalopathy in rats with portacaval shunts. Gut 60:1572–1579PubMedCrossRef Agusti A, Cauli O, Rodrigo R, Llansola M, Hernandez-Rabaza V, Felipo V (2011) p38 MAP kinase is a therapeutic target for hepatic encephalopathy in rats with portacaval shunts. Gut 60:1572–1579PubMedCrossRef
go back to reference Albrecht J, Norenberg MD (2006) Glutamine: a Trojan horse in ammonia neurotoxicity. Hepatology 44:788–794PubMedCrossRef Albrecht J, Norenberg MD (2006) Glutamine: a Trojan horse in ammonia neurotoxicity. Hepatology 44:788–794PubMedCrossRef
go back to reference Albrecht J, Zielinska M, Norenberg MD (2010) Glutamine as a mediator of ammonia neurotoxicity: a critical appraisal. Biochem Pharmacol 80:1303–1308PubMedCrossRef Albrecht J, Zielinska M, Norenberg MD (2010) Glutamine as a mediator of ammonia neurotoxicity: a critical appraisal. Biochem Pharmacol 80:1303–1308PubMedCrossRef
go back to reference Als-Nielsen B, Gluud LL, Gluud C (2004) Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. Br Med J 328:1046CrossRef Als-Nielsen B, Gluud LL, Gluud C (2004) Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. Br Med J 328:1046CrossRef
go back to reference Alvarez VM, Rama Rao KV, Brahmbhatt M, Norenberg MD (2011) Interaction between cytokines and ammonia in the mitochondrial permeability transition in cultured astrocytes. J Neurosci Res 89:2028–2040PubMedCrossRef Alvarez VM, Rama Rao KV, Brahmbhatt M, Norenberg MD (2011) Interaction between cytokines and ammonia in the mitochondrial permeability transition in cultured astrocytes. J Neurosci Res 89:2028–2040PubMedCrossRef
go back to reference Azorin I, Minana MD, Felipo V, Grisolia S (1989) A simple animal model of hyperammonemia. Hepatology 10:311–314PubMedCrossRef Azorin I, Minana MD, Felipo V, Grisolia S (1989) A simple animal model of hyperammonemia. Hepatology 10:311–314PubMedCrossRef
go back to reference Bachmann C (2003) Outcome and survival of 88 patients with urea cycle disorders: a retrospective evaluation. Eur J Pediatr 162:410–416PubMedCrossRef Bachmann C (2003) Outcome and survival of 88 patients with urea cycle disorders: a retrospective evaluation. Eur J Pediatr 162:410–416PubMedCrossRef
go back to reference Bachmann C, Colombo JP (1984) Increase of tryptophan and 5-hydroxyindole acetic acid in the brain of ornithine carbamoyltransferase deficient sparse-fur mice. Pediatr Res 18:372–375PubMedCrossRef Bachmann C, Colombo JP (1984) Increase of tryptophan and 5-hydroxyindole acetic acid in the brain of ornithine carbamoyltransferase deficient sparse-fur mice. Pediatr Res 18:372–375PubMedCrossRef
go back to reference Bachmann C, Braissant O, Villard AM, Boulat O, Henry H (2004) Ammonia toxicity to the brain and creatine. Mol Genet Metab 81(Suppl 1):S52–S57PubMedCrossRef Bachmann C, Braissant O, Villard AM, Boulat O, Henry H (2004) Ammonia toxicity to the brain and creatine. Mol Genet Metab 81(Suppl 1):S52–S57PubMedCrossRef
go back to reference Bai G, Rama Rao KV, Murthy CR, Panickar KS, Jayakumar AR, Norenberg MD (2001) Ammonia induces the mitochondrial permeability transition in primary cultures of rat astrocytes. J Neurosci Res 66:981–991PubMedCrossRef Bai G, Rama Rao KV, Murthy CR, Panickar KS, Jayakumar AR, Norenberg MD (2001) Ammonia induces the mitochondrial permeability transition in primary cultures of rat astrocytes. J Neurosci Res 66:981–991PubMedCrossRef
go back to reference Bajaj JS, Cordoba J, Mullen KD et al (2011) Review article: the design of clinical trials in hepatic encephalopathy–an International Society for Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN) consensus statement. Aliment Pharmacol Ther 33:739–747PubMedCrossRef Bajaj JS, Cordoba J, Mullen KD et al (2011) Review article: the design of clinical trials in hepatic encephalopathy–an International Society for Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN) consensus statement. Aliment Pharmacol Ther 33:739–747PubMedCrossRef
go back to reference Bajaj JS, Pinkerton SD, Sanyal AJ, Heuman DM (2012) Diagnosis and treatment of minimal hepatic encephalopathy to prevent motor vehicle accidents: a cost-effectiveness analysis. Hepatology 55:1164–1171PubMedCrossRef Bajaj JS, Pinkerton SD, Sanyal AJ, Heuman DM (2012) Diagnosis and treatment of minimal hepatic encephalopathy to prevent motor vehicle accidents: a cost-effectiveness analysis. Hepatology 55:1164–1171PubMedCrossRef
go back to reference Bass NM, Mullen KD, Sanyal A et al (2010) Rifaximin treatment in hepatic encephalopathy. N Engl J Med 362:1071–1081PubMedCrossRef Bass NM, Mullen KD, Sanyal A et al (2010) Rifaximin treatment in hepatic encephalopathy. N Engl J Med 362:1071–1081PubMedCrossRef
go back to reference Batshaw ML, MacArthur RB, Tuchman M (2001) Alternative pathway therapy for urea cycle disorders: twenty years later. J Pediatr 138:S46–S54PubMedCrossRef Batshaw ML, MacArthur RB, Tuchman M (2001) Alternative pathway therapy for urea cycle disorders: twenty years later. J Pediatr 138:S46–S54PubMedCrossRef
go back to reference Béard E, Braissant O (2010) Synthesis and transport of creatine in the CNS: importance for cerebral functions. J Neurochem 115:297–313PubMedCrossRef Béard E, Braissant O (2010) Synthesis and transport of creatine in the CNS: importance for cerebral functions. J Neurochem 115:297–313PubMedCrossRef
go back to reference Bélanger M, Asashima T, Ohtsuki S, Yamaguchi H, Ito S, Terasaki T (2007) Hyperammonemia induces transport of taurine and creatine and suppresses claudin-12 gene expression in brain capillary endothelial cells in vitro. Neurochem Int 50:95–101PubMedCrossRef Bélanger M, Asashima T, Ohtsuki S, Yamaguchi H, Ito S, Terasaki T (2007) Hyperammonemia induces transport of taurine and creatine and suppresses claudin-12 gene expression in brain capillary endothelial cells in vitro. Neurochem Int 50:95–101PubMedCrossRef
go back to reference Berl S, Takagaki G, Clarke DD, Waelsch H (1962) Metabolic compartments in vivo. Ammonia and glutamic acid metabolism in brain and liver. J Biol Chem 237:2562–2569PubMed Berl S, Takagaki G, Clarke DD, Waelsch H (1962) Metabolic compartments in vivo. Ammonia and glutamic acid metabolism in brain and liver. J Biol Chem 237:2562–2569PubMed
go back to reference Berry GT, Steiner RD (2001) Long-term management of patients with urea cycle disorders. J Pediatr 138:S56–S60PubMedCrossRef Berry GT, Steiner RD (2001) Long-term management of patients with urea cycle disorders. J Pediatr 138:S56–S60PubMedCrossRef
go back to reference Bosoi CR, Yang X, Huynh J et al (2012) Systemic oxidative stress is implicated in the pathogenesis of brain edema in rats with chronic liver failure. Free Radic Biol Med 52:1228–1235PubMedCrossRef Bosoi CR, Yang X, Huynh J et al (2012) Systemic oxidative stress is implicated in the pathogenesis of brain edema in rats with chronic liver failure. Free Radic Biol Med 52:1228–1235PubMedCrossRef
go back to reference Braissant O (2010a) Current concepts in the pathogenesis of urea cycle disorders. Mol Gen Metab 100(Suppl 1):S3–S12CrossRef Braissant O (2010a) Current concepts in the pathogenesis of urea cycle disorders. Mol Gen Metab 100(Suppl 1):S3–S12CrossRef
go back to reference Braissant O (2010b) Ammonia toxicity to the brain: effects on creatine metabolism and transport and protective roles of creatine. Mol Genet Metab 100(Suppl 1):S53–S58PubMedCrossRef Braissant O (2010b) Ammonia toxicity to the brain: effects on creatine metabolism and transport and protective roles of creatine. Mol Genet Metab 100(Suppl 1):S53–S58PubMedCrossRef
go back to reference Braissant O (2012) Creatine and guanidinoacetate transport at blood–brain and blood-cerebrospinal fluid barriers. J Inherit Metab Dis 35:655–664PubMedCrossRef Braissant O (2012) Creatine and guanidinoacetate transport at blood–brain and blood-cerebrospinal fluid barriers. J Inherit Metab Dis 35:655–664PubMedCrossRef
go back to reference Braissant O, Gotoh T, Loup M, Mori M, Bachmann C (1999a) L-arginine uptake, the citrulline-NO cycle and arginase II in the rat brain: an in situ hybridization study. Mol Brain Res 70:231–241PubMedCrossRef Braissant O, Gotoh T, Loup M, Mori M, Bachmann C (1999a) L-arginine uptake, the citrulline-NO cycle and arginase II in the rat brain: an in situ hybridization study. Mol Brain Res 70:231–241PubMedCrossRef
go back to reference Braissant O, Honegger P, Loup M, Iwase K, Takiguchi M, Bachmann C (1999b) Hyperammonemia: regulation of argininosuccinate synthetase and argininosuccinate lyase genes in aggregating cell cultures of fetal rat brain. Neurosci Lett 266:89–92PubMedCrossRef Braissant O, Honegger P, Loup M, Iwase K, Takiguchi M, Bachmann C (1999b) Hyperammonemia: regulation of argininosuccinate synthetase and argininosuccinate lyase genes in aggregating cell cultures of fetal rat brain. Neurosci Lett 266:89–92PubMedCrossRef
go back to reference Braissant O, Gotoh T, Loup M, Mori M, Bachmann C (2001) Differential expression of the cationic amino acid transporter 2(B) in the adult rat brain. Mol Brain Res 91:189–195PubMedCrossRef Braissant O, Gotoh T, Loup M, Mori M, Bachmann C (2001) Differential expression of the cationic amino acid transporter 2(B) in the adult rat brain. Mol Brain Res 91:189–195PubMedCrossRef
go back to reference Braissant O, Henry H, Villard AM et al (2002) Ammonium-induced impairment of axonal growth is prevented through glial creatine. J Neurosci 22:9810–9820PubMed Braissant O, Henry H, Villard AM et al (2002) Ammonium-induced impairment of axonal growth is prevented through glial creatine. J Neurosci 22:9810–9820PubMed
go back to reference Braissant O, Cagnon L, Monnet-Tschudi F et al (2008) Ammonium alters creatine transport and synthesis in a 3D-culture of developing brain cells, resulting in secondary cerebral creatine deficiency. Eur J Neurosci 27:1673–1685PubMedCrossRef Braissant O, Cagnon L, Monnet-Tschudi F et al (2008) Ammonium alters creatine transport and synthesis in a 3D-culture of developing brain cells, resulting in secondary cerebral creatine deficiency. Eur J Neurosci 27:1673–1685PubMedCrossRef
go back to reference Braissant O, Henry H, Beard E, Uldry J (2011) Creatine deficiency syndromes and the importance of creatine synthesis in the brain. Amino Acids 40:1315–1324PubMedCrossRef Braissant O, Henry H, Beard E, Uldry J (2011) Creatine deficiency syndromes and the importance of creatine synthesis in the brain. Amino Acids 40:1315–1324PubMedCrossRef
go back to reference Brosnan JT, Brosnan ME (2010) Creatine metabolism and the urea cycle. Mol Genet Metab 100(Suppl 1):S49–S52PubMedCrossRef Brosnan JT, Brosnan ME (2010) Creatine metabolism and the urea cycle. Mol Genet Metab 100(Suppl 1):S49–S52PubMedCrossRef
go back to reference Brosnan ME, Edison EE, da Silva R, Brosnan JT (2007) New insights into creatine function and synthesis. Adv Enzym Regul 47:252–260CrossRef Brosnan ME, Edison EE, da Silva R, Brosnan JT (2007) New insights into creatine function and synthesis. Adv Enzym Regul 47:252–260CrossRef
go back to reference Brusilow SW, Maestri NE (1996) Urea cycle disorders: diagnosis, pathophysiology, and therapy. Adv Pediatr 43:127–170PubMed Brusilow SW, Maestri NE (1996) Urea cycle disorders: diagnosis, pathophysiology, and therapy. Adv Pediatr 43:127–170PubMed
go back to reference Brusilow SW, Valle DL, Batshaw M (1979) New pathways of nitrogen excretion in inborn errors of urea synthesis. Lancet 2:452–454PubMedCrossRef Brusilow SW, Valle DL, Batshaw M (1979) New pathways of nitrogen excretion in inborn errors of urea synthesis. Lancet 2:452–454PubMedCrossRef
go back to reference Butterworth RF (1998) Effects of hyperammonaemia on brain function. J Inherit Metab Dis 21(Suppl 1):6–20PubMedCrossRef Butterworth RF (1998) Effects of hyperammonaemia on brain function. J Inherit Metab Dis 21(Suppl 1):6–20PubMedCrossRef
go back to reference Butterworth RF (2003) Hepatic encephalopathy. Alcohol Res Health 27:240–246PubMed Butterworth RF (2003) Hepatic encephalopathy. Alcohol Res Health 27:240–246PubMed
go back to reference Butterworth RF (2012) Brain edema and encephalopathy in acute liver failure: a primary neurogliopathy? Neurochem Int 60:661PubMedCrossRef Butterworth RF (2012) Brain edema and encephalopathy in acute liver failure: a primary neurogliopathy? Neurochem Int 60:661PubMedCrossRef
go back to reference Butterworth RF, Norenberg MD, Felipo V et al (2009) Experimental models of hepatic encephalopathy: ISHEN guidelines. Liver Int 29:783–788PubMedCrossRef Butterworth RF, Norenberg MD, Felipo V et al (2009) Experimental models of hepatic encephalopathy: ISHEN guidelines. Liver Int 29:783–788PubMedCrossRef
go back to reference Cagnon L, Braissant O (2007) Hyperammonemia-induced toxicity for the developing central nervous system. Brain Res Rev 56:183–197PubMedCrossRef Cagnon L, Braissant O (2007) Hyperammonemia-induced toxicity for the developing central nervous system. Brain Res Rev 56:183–197PubMedCrossRef
go back to reference Cagnon L, Braissant O (2008) Role of caspases, calpain and cdk5 in ammonia-induced cell death in developing brain cells. Neurobiol Dis 32:281–292PubMedCrossRef Cagnon L, Braissant O (2008) Role of caspases, calpain and cdk5 in ammonia-induced cell death in developing brain cells. Neurobiol Dis 32:281–292PubMedCrossRef
go back to reference Cagnon L, Braissant O (2009) CNTF protects oligodendrocytes from ammonia toxicity: intracellular signaling pathways involved. Neurobiol Dis 33:133–142PubMedCrossRef Cagnon L, Braissant O (2009) CNTF protects oligodendrocytes from ammonia toxicity: intracellular signaling pathways involved. Neurobiol Dis 33:133–142PubMedCrossRef
go back to reference Call G, Seay AR, Sherry R, Qureshi IA (1984) Clinical features of carbamyl phosphate synthetase-I deficiency in an adult. Ann Neurol 16:90–93PubMedCrossRef Call G, Seay AR, Sherry R, Qureshi IA (1984) Clinical features of carbamyl phosphate synthetase-I deficiency in an adult. Ann Neurol 16:90–93PubMedCrossRef
go back to reference Caudle SE, Katzenstein JM, Karpen SJ, McLin VA (2010) Language and motor skills are impaired in infants with biliary atresia before transplantation. J Pediatr 156:936–940PubMedCrossRef Caudle SE, Katzenstein JM, Karpen SJ, McLin VA (2010) Language and motor skills are impaired in infants with biliary atresia before transplantation. J Pediatr 156:936–940PubMedCrossRef
go back to reference Caudle SE, Katzenstein JM, Karpen S, McLin V (2012) Developmental assessment of infants with biliary atresia: Differences between males and females. J Pediatr Gastroenterol Nutr 55(4):384-389 Caudle SE, Katzenstein JM, Karpen S, McLin V (2012) Developmental assessment of infants with biliary atresia: Differences between males and females. J Pediatr Gastroenterol Nutr 55(4):384-389
go back to reference Cauli O, Lopez-Larrubia P, Rodrigues TB, Cerdan S, Felipo V (2007) Magnetic resonance analysis of the effects of acute ammonia intoxication on rat brain. Role of NMDA receptors. J Neurochem 103:1334–1343PubMedCrossRef Cauli O, Lopez-Larrubia P, Rodrigues TB, Cerdan S, Felipo V (2007) Magnetic resonance analysis of the effects of acute ammonia intoxication on rat brain. Role of NMDA receptors. J Neurochem 103:1334–1343PubMedCrossRef
go back to reference Cauli O, Rodrigo R, Llansola M et al (2009) Glutamatergic and gabaergic neurotransmission and neuronal circuits in hepatic encephalopathy. Metab Brain Dis 24:69–80PubMedCrossRef Cauli O, Rodrigo R, Llansola M et al (2009) Glutamatergic and gabaergic neurotransmission and neuronal circuits in hepatic encephalopathy. Metab Brain Dis 24:69–80PubMedCrossRef
go back to reference Cauli O, Lopez-Larrubia P, Rodrigo R et al (2011) Brain region-selective mechanisms contribute to the progression of cerebral alterations in acute liver failure in rats. Gastroenterology 140:638–645PubMedCrossRef Cauli O, Lopez-Larrubia P, Rodrigo R et al (2011) Brain region-selective mechanisms contribute to the progression of cerebral alterations in acute liver failure in rats. Gastroenterology 140:638–645PubMedCrossRef
go back to reference Cederbaum S, Lemons C, Batshaw ML (2010) Alternative pathway or diversion therapy for urea cycle disorders now and in the future. Mol Genet Metab 100:219–220PubMedCrossRef Cederbaum S, Lemons C, Batshaw ML (2010) Alternative pathway or diversion therapy for urea cycle disorders now and in the future. Mol Genet Metab 100:219–220PubMedCrossRef
go back to reference Chan H, Hazell AS, Desjardins P, Butterworth RF (2000) Effects of ammonia on glutamate transporter (GLAST) protein and mRNA in cultured rat cortical astrocytes. Neurochem Int 37:243–248PubMedCrossRef Chan H, Hazell AS, Desjardins P, Butterworth RF (2000) Effects of ammonia on glutamate transporter (GLAST) protein and mRNA in cultured rat cortical astrocytes. Neurochem Int 37:243–248PubMedCrossRef
go back to reference Chatauret N, Zwingmann C, Rose C, Leibfritz D, Butterworth RF (2003) Effects of hypothermia on brain glucose metabolism in acute liver failure: a 1H/13C-nuclear magnetic resonance study. Gastroenterology 125:815–824PubMedCrossRef Chatauret N, Zwingmann C, Rose C, Leibfritz D, Butterworth RF (2003) Effects of hypothermia on brain glucose metabolism in acute liver failure: a 1H/13C-nuclear magnetic resonance study. Gastroenterology 125:815–824PubMedCrossRef
go back to reference Chavarria L, Oria M, Romero-Gimenez J, Alonso J, Lope-Piedrafita S, Cordoba J (2010) Diffusion tensor imaging supports the cytotoxic origin of brain edema in a rat model of acute liver failure. Gastroenterology 138:1566–1573PubMedCrossRef Chavarria L, Oria M, Romero-Gimenez J, Alonso J, Lope-Piedrafita S, Cordoba J (2010) Diffusion tensor imaging supports the cytotoxic origin of brain edema in a rat model of acute liver failure. Gastroenterology 138:1566–1573PubMedCrossRef
go back to reference Chavarria L, Alonso J, Rovira A, Cordoba J (2011) Neuroimaging in acute liver failure. Neurochem Int 59:1175–1180PubMedCrossRef Chavarria L, Alonso J, Rovira A, Cordoba J (2011) Neuroimaging in acute liver failure. Neurochem Int 59:1175–1180PubMedCrossRef
go back to reference Chepkova AN, Sergeeva OA, Haas HL (2006) Taurine rescues hippocampal long-term potentiation from ammonia-induced impairment. Neurobiol Dis 23:512–521PubMedCrossRef Chepkova AN, Sergeeva OA, Haas HL (2006) Taurine rescues hippocampal long-term potentiation from ammonia-induced impairment. Neurobiol Dis 23:512–521PubMedCrossRef
go back to reference Choi JH, Kim H, Yoo HW (2006) Two cases of citrullinaemia presenting with stroke. J Inherit Metab Dis 29:182–183PubMedCrossRef Choi JH, Kim H, Yoo HW (2006) Two cases of citrullinaemia presenting with stroke. J Inherit Metab Dis 29:182–183PubMedCrossRef
go back to reference Connelly A, Cross JH, Gadian DG, Hunter JV, Kirkham FJ, Leonard JV (1993) Magnetic resonance spectroscopy shows increased brain glutamine in ornithine carbamoyl transferase deficiency. Pediatr Res 33:77–81PubMedCrossRef Connelly A, Cross JH, Gadian DG, Hunter JV, Kirkham FJ, Leonard JV (1993) Magnetic resonance spectroscopy shows increased brain glutamine in ornithine carbamoyl transferase deficiency. Pediatr Res 33:77–81PubMedCrossRef
go back to reference Cordoba J (1996) Glutamine, myo-inositol, and brain edema in acute liver failure. Hepatology 23:1291–1292PubMedCrossRef Cordoba J (1996) Glutamine, myo-inositol, and brain edema in acute liver failure. Hepatology 23:1291–1292PubMedCrossRef
go back to reference Cudalbu C, Mlynárik V, Lanz B, Frenkel H, Costers N, Gruetter R (2010) Imaging glutamine synthesis rates in the hyperammonemic rat brain. Proc Intl Soc Magn Reson Med 18:3324 Cudalbu C, Mlynárik V, Lanz B, Frenkel H, Costers N, Gruetter R (2010) Imaging glutamine synthesis rates in the hyperammonemic rat brain. Proc Intl Soc Magn Reson Med 18:3324
go back to reference Cudalbu C, Mlynárik V, Gruetter R (2012a) Handling macromolecule signals in the quantification of the neurochemical profile. J Alzh Dis 31:S101-115 Cudalbu C, Mlynárik V, Gruetter R (2012a) Handling macromolecule signals in the quantification of the neurochemical profile. J Alzh Dis 31:S101-115
go back to reference Cudalbu C, Lanz B, Duarte JM et al (2012b) Cerebral glutamine metabolism under hyperammonemia determined in vivo by localized 1H and 15N NMR spectroscopy. J Cereb Blood Flow Metab 32:696–708PubMedCrossRef Cudalbu C, Lanz B, Duarte JM et al (2012b) Cerebral glutamine metabolism under hyperammonemia determined in vivo by localized 1H and 15N NMR spectroscopy. J Cereb Blood Flow Metab 32:696–708PubMedCrossRef
go back to reference D’Hooge R, Marescau B, Qureshi IA, De Deyn PP (2000) Impaired cognitive performance in ornithine transcarbamylase-deficient mice on arginine-free diet. Brain Res 876:1–9PubMedCrossRef D’Hooge R, Marescau B, Qureshi IA, De Deyn PP (2000) Impaired cognitive performance in ornithine transcarbamylase-deficient mice on arginine-free diet. Brain Res 876:1–9PubMedCrossRef
go back to reference Dadsetan S, Bak LK, Sørensen M et al (2011) Inhibition of glutamine synthesis induces glutamate dehydrogenase-dependent ammonia fixation into alanine in co-cultures of astrocytes and neurons. Neurochem Int 59:482–488PubMedCrossRef Dadsetan S, Bak LK, Sørensen M et al (2011) Inhibition of glutamine synthesis induces glutamate dehydrogenase-dependent ammonia fixation into alanine in co-cultures of astrocytes and neurons. Neurochem Int 59:482–488PubMedCrossRef
go back to reference de Grauw TJ, Smit LM, Brockstedt M, Meijer Y, van der Klei-van Moorsel JM, Jakobs C (1990) Acute hemiparesis as the presenting sign in a heterozygote for ornithine transcarbamylase deficiency. Neuropediatrics 21:133–135PubMedCrossRef de Grauw TJ, Smit LM, Brockstedt M, Meijer Y, van der Klei-van Moorsel JM, Jakobs C (1990) Acute hemiparesis as the presenting sign in a heterozygote for ornithine transcarbamylase deficiency. Neuropediatrics 21:133–135PubMedCrossRef
go back to reference de Knegt RJ, Schalm SW, van der Rijt CC, Fekkes D, Dalm E, Hekking-Weyma I (1994) Extracellular brain glutamate during acute liver failure and during acute hyperammonemia simulating acute liver failure: an experimental study based on in vivo brain dialysis. J Hepatol 20:19–26PubMedCrossRef de Knegt RJ, Schalm SW, van der Rijt CC, Fekkes D, Dalm E, Hekking-Weyma I (1994) Extracellular brain glutamate during acute liver failure and during acute hyperammonemia simulating acute liver failure: an experimental study based on in vivo brain dialysis. J Hepatol 20:19–26PubMedCrossRef
go back to reference Deignan JL, Cederbaum SD, Grody WW (2008) Contrasting features of urea cycle disorders in human patients and knockout mouse models. Mol Genet Metab 93:7–14PubMedCrossRef Deignan JL, Cederbaum SD, Grody WW (2008) Contrasting features of urea cycle disorders in human patients and knockout mouse models. Mol Genet Metab 93:7–14PubMedCrossRef
go back to reference Desjardins P, Du T, Jiang W, Peng L, Butterworth RF (2012) Pathogenesis of hepatic encephalopathy and brain edema in acute liver failure: role of glutamine redefined. Neurochem Int 60:690–696PubMedCrossRef Desjardins P, Du T, Jiang W, Peng L, Butterworth RF (2012) Pathogenesis of hepatic encephalopathy and brain edema in acute liver failure: role of glutamine redefined. Neurochem Int 60:690–696PubMedCrossRef
go back to reference Dolder M, Walzel B, Speer O, Schlattner U, Wallimann T (2003) Inhibition of the mitochondrial permeability transition by creatine kinase substrates. Requirement for microcompartmentation. J Biol Chem 278:17760–17766PubMedCrossRef Dolder M, Walzel B, Speer O, Schlattner U, Wallimann T (2003) Inhibition of the mitochondrial permeability transition by creatine kinase substrates. Requirement for microcompartmentation. J Biol Chem 278:17760–17766PubMedCrossRef
go back to reference Dolman CL, Clasen RA, Dorovini-Zis K (1988) Severe cerebral damage in ornithine transcarbamylase deficiency. Clin Neuropathol 7:10–15PubMed Dolman CL, Clasen RA, Dorovini-Zis K (1988) Severe cerebral damage in ornithine transcarbamylase deficiency. Clin Neuropathol 7:10–15PubMed
go back to reference Eltawil KM, Laryea M, Peltekian K, Molinari M (2012) Rifaximin vs. conventional oral therapy for hepatic encephalopathy: a meta-analysis. World J Gastroenterol 18:767–777PubMedCrossRef Eltawil KM, Laryea M, Peltekian K, Molinari M (2012) Rifaximin vs. conventional oral therapy for hepatic encephalopathy: a meta-analysis. World J Gastroenterol 18:767–777PubMedCrossRef
go back to reference Enns GM (2008) Neurologic damage and neurocognitive dysfunction in urea cycle disorders. Semin Pediatr Neurol 15:132–139PubMedCrossRef Enns GM (2008) Neurologic damage and neurocognitive dysfunction in urea cycle disorders. Semin Pediatr Neurol 15:132–139PubMedCrossRef
go back to reference Enns GM, Berry SA, Berry GT, Rhead WJ, Brusilow SW, Hamosh A (2007) Survival after treatment with phenylacetate and benzoate for urea-cycle disorders. N Engl J Med 356:2282–2292PubMedCrossRef Enns GM, Berry SA, Berry GT, Rhead WJ, Brusilow SW, Hamosh A (2007) Survival after treatment with phenylacetate and benzoate for urea-cycle disorders. N Engl J Med 356:2282–2292PubMedCrossRef
go back to reference Felipo V, Grau E, Minana MD, Grisolia S (1993) Hyperammonemia decreases protein-kinase-C-dependent phosphorylation of microtubule-associated protein 2 and increases its binding to tubulin. Eur J Biochem 214:243–249PubMedCrossRef Felipo V, Grau E, Minana MD, Grisolia S (1993) Hyperammonemia decreases protein-kinase-C-dependent phosphorylation of microtubule-associated protein 2 and increases its binding to tubulin. Eur J Biochem 214:243–249PubMedCrossRef
go back to reference Felipo V, Ordono JF, Urios A et al (2012) Patients with minimal hepatic encephalopathy show impaired mismatch negativity correlating with reduced performance in attention tests. Hepatology 55:530–539PubMedCrossRef Felipo V, Ordono JF, Urios A et al (2012) Patients with minimal hepatic encephalopathy show impaired mismatch negativity correlating with reduced performance in attention tests. Hepatology 55:530–539PubMedCrossRef
go back to reference Filloux F, Townsend JJ, Leonard C (1986) Ornithine transcarbamylase deficiency: neuropathologic changes acquired in utero. J Pediatr 108:942–945PubMedCrossRef Filloux F, Townsend JJ, Leonard C (1986) Ornithine transcarbamylase deficiency: neuropathologic changes acquired in utero. J Pediatr 108:942–945PubMedCrossRef
go back to reference Fitzpatrick SM, Hetherington HP, Behar KL, Shulman RG (1989) Effects of acute hyperammonemia on cerebral amino acid metabolism and pHi in vivo, measured by 1H and 31P nuclear magnetic resonance. J Neurochem 52:741–749PubMedCrossRef Fitzpatrick SM, Hetherington HP, Behar KL, Shulman RG (1989) Effects of acute hyperammonemia on cerebral amino acid metabolism and pHi in vivo, measured by 1H and 31P nuclear magnetic resonance. J Neurochem 52:741–749PubMedCrossRef
go back to reference Flint Beal M, Martin JB (1998) Major complications of cirrhosis. In: Fauci AS, Braunwald E, Isselbacher KJ, Wilson JD, Martin JB, Kasper DL, Hauser SL, Longo DL (eds) Harrison’s principles of internal medicine, 14th edn. McGraw-Hill, New-York, pp 2451–2457 Flint Beal M, Martin JB (1998) Major complications of cirrhosis. In: Fauci AS, Braunwald E, Isselbacher KJ, Wilson JD, Martin JB, Kasper DL, Hauser SL, Longo DL (eds) Harrison’s principles of internal medicine, 14th edn. McGraw-Hill, New-York, pp 2451–2457
go back to reference Foerster BR, Conklin LS, Petrou M, Barker PB, Schwarz KB (2009) Minimal hepatic encephalopathy in children: evaluation with proton MR spectroscopy. Am J Neuroradiol 30:1610–1613PubMedCrossRef Foerster BR, Conklin LS, Petrou M, Barker PB, Schwarz KB (2009) Minimal hepatic encephalopathy in children: evaluation with proton MR spectroscopy. Am J Neuroradiol 30:1610–1613PubMedCrossRef
go back to reference Garcia-Ayllon MS, Cauli O, Silveyra MX et al (2008) Brain cholinergic impairment in liver failure. Brain 131:2946–2956PubMedCrossRef Garcia-Ayllon MS, Cauli O, Silveyra MX et al (2008) Brain cholinergic impairment in liver failure. Brain 131:2946–2956PubMedCrossRef
go back to reference Gropman AL, Summar M, Leonard JV (2007) Neurological implications of urea cycle disorders. J Inherit Metab Dis 30:865–879PubMedCrossRef Gropman AL, Summar M, Leonard JV (2007) Neurological implications of urea cycle disorders. J Inherit Metab Dis 30:865–879PubMedCrossRef
go back to reference Gropman AL, Gertz B, Shattuck K et al (2010) Diffusion tensor imaging detects areas of abnormal white matter microstructure in patients with partial ornithine transcarbamylase deficiency. Am J Neuroradiol 31:1719–1723PubMedCrossRef Gropman AL, Gertz B, Shattuck K et al (2010) Diffusion tensor imaging detects areas of abnormal white matter microstructure in patients with partial ornithine transcarbamylase deficiency. Am J Neuroradiol 31:1719–1723PubMedCrossRef
go back to reference Grover VP, Dresner MA, Forton DM et al (2006) Current and future applications of magnetic resonance imaging and spectroscopy of the brain in hepatic encephalopathy. World J Gastroenterol 12:2969–2978PubMed Grover VP, Dresner MA, Forton DM et al (2006) Current and future applications of magnetic resonance imaging and spectroscopy of the brain in hepatic encephalopathy. World J Gastroenterol 12:2969–2978PubMed
go back to reference Gruetter R (2002) In vivo 13C NMR studies of compartmentalized cerebral carbohydrate metabolism. Neurochem Int 41:143–154PubMedCrossRef Gruetter R (2002) In vivo 13C NMR studies of compartmentalized cerebral carbohydrate metabolism. Neurochem Int 41:143–154PubMedCrossRef
go back to reference Harada E, Nishiyori A, Tokunaga Y et al (2006) Late-onset ornithine transcarbamylase deficiency in male patients: prognostic factors and characteristics of plasma amino acid profile. Pediatr Int 48:105–111PubMedCrossRef Harada E, Nishiyori A, Tokunaga Y et al (2006) Late-onset ornithine transcarbamylase deficiency in male patients: prognostic factors and characteristics of plasma amino acid profile. Pediatr Int 48:105–111PubMedCrossRef
go back to reference Harding BN, Leonard JV, Erdohazi M (1984) Ornithine carbamoyl transferase deficiency: a neuropathological study. Eur J Pediatr 141:215–220PubMedCrossRef Harding BN, Leonard JV, Erdohazi M (1984) Ornithine carbamoyl transferase deficiency: a neuropathological study. Eur J Pediatr 141:215–220PubMedCrossRef
go back to reference Harding BN, Leonard JV, Erdohazi M (1991) Propionic acidaemia: a neuropathological study of two patients presenting in infancy. Neuropathol Appl Neurobiol 17:133–138PubMedCrossRef Harding BN, Leonard JV, Erdohazi M (1991) Propionic acidaemia: a neuropathological study of two patients presenting in infancy. Neuropathol Appl Neurobiol 17:133–138PubMedCrossRef
go back to reference Hertz L, Kala G (2007) Energy metabolism in brain cells: effects of elevated ammonia concentrations. Metab Brain Dis 22:199–218PubMedCrossRef Hertz L, Kala G (2007) Energy metabolism in brain cells: effects of elevated ammonia concentrations. Metab Brain Dis 22:199–218PubMedCrossRef
go back to reference Holm LM, Jahn TP, Møller AL et al (2005) NH3 and NH4 + permeability in aquaporin-expressing Xenopus oocytes. Pflugers Arch 450:415–428PubMedCrossRef Holm LM, Jahn TP, Møller AL et al (2005) NH3 and NH4 + permeability in aquaporin-expressing Xenopus oocytes. Pflugers Arch 450:415–428PubMedCrossRef
go back to reference Honegger P, Monnet-Tschudi F (2001) Aggregating neural cell culture. In: Fedoroff S, Richardson A (eds) Protocols for neural cell culture, 3rd edn. Humana Press, Totowa, pp 199–218CrossRef Honegger P, Monnet-Tschudi F (2001) Aggregating neural cell culture. In: Fedoroff S, Richardson A (eds) Protocols for neural cell culture, 3rd edn. Humana Press, Totowa, pp 199–218CrossRef
go back to reference Hopkins KJ, McKean J, Mervis RF, Oster-Granite ML (1998) Dendritic alterations in cortical pyramidal cells in the sparse fur mouse. Brain Res 797:167–172PubMedCrossRef Hopkins KJ, McKean J, Mervis RF, Oster-Granite ML (1998) Dendritic alterations in cortical pyramidal cells in the sparse fur mouse. Brain Res 797:167–172PubMedCrossRef
go back to reference Hyman SL, Coyle JT, Parke JC et al (1986) Anorexia and altered serotonin metabolism in a patient with argininosuccinic aciduria. J Pediatr 108:705–709PubMedCrossRef Hyman SL, Coyle JT, Parke JC et al (1986) Anorexia and altered serotonin metabolism in a patient with argininosuccinic aciduria. J Pediatr 108:705–709PubMedCrossRef
go back to reference Hyman SL, Porter CA, Page TJ, Iwata BA, Kissel R, Batshaw ML (1987) Behavior management of feeding disturbances in urea cycle and organic acid disorders. J Pediatr 111:558–562PubMedCrossRef Hyman SL, Porter CA, Page TJ, Iwata BA, Kissel R, Batshaw ML (1987) Behavior management of feeding disturbances in urea cycle and organic acid disorders. J Pediatr 111:558–562PubMedCrossRef
go back to reference Inoue I, Gushiken T, Kobayashi K, Saheki T (1987) Accumulation of large neutral amino acids in the brain of sparse-fur mice at hyperammonemic state. Biochem Med Metab Biol 38:378–386PubMedCrossRef Inoue I, Gushiken T, Kobayashi K, Saheki T (1987) Accumulation of large neutral amino acids in the brain of sparse-fur mice at hyperammonemic state. Biochem Med Metab Biol 38:378–386PubMedCrossRef
go back to reference Izumi Y, Izumi M, Matsukawa M, Funatsu M, Zorumski CF (2005) Ammonia-mediated LTP inhibition: effects of NMDA receptor antagonists and L-carnitine. Neurobiol Dis 20:615–624PubMedCrossRef Izumi Y, Izumi M, Matsukawa M, Funatsu M, Zorumski CF (2005) Ammonia-mediated LTP inhibition: effects of NMDA receptor antagonists and L-carnitine. Neurobiol Dis 20:615–624PubMedCrossRef
go back to reference Jalan R, Wright G, Davies NA, Hodges SJ (2007) L-Ornithine phenylacetate (OP): a novel treatment for hyperammonemia and hepatic encephalopathy. Med Hypotheses 69:1064–1069PubMedCrossRef Jalan R, Wright G, Davies NA, Hodges SJ (2007) L-Ornithine phenylacetate (OP): a novel treatment for hyperammonemia and hepatic encephalopathy. Med Hypotheses 69:1064–1069PubMedCrossRef
go back to reference Jambekar AA, Palma E, Nicolosi L et al (2011) A glutamine synthetase inhibitor increases survival and decreases cytokine response in a mouse model of acute liver failure. Liver Int 31:1209–1221PubMedCrossRef Jambekar AA, Palma E, Nicolosi L et al (2011) A glutamine synthetase inhibitor increases survival and decreases cytokine response in a mouse model of acute liver failure. Liver Int 31:1209–1221PubMedCrossRef
go back to reference Jayakumar AR, Norenberg MD (2010) The Na-K-Cl Co-transporter in astrocyte swelling. Metab Brain Dis 25:31–38PubMedCrossRef Jayakumar AR, Norenberg MD (2010) The Na-K-Cl Co-transporter in astrocyte swelling. Metab Brain Dis 25:31–38PubMedCrossRef
go back to reference Jayakumar AR, Panickar KS, Murthy C, Norenberg MD (2006) Oxidative stress and mitogen-activated protein kinase phosphorylation mediate ammonia-induced cell swelling and glutamate uptake inhibition in cultured astrocytes. J Neurosci 26:4774–4784PubMedCrossRef Jayakumar AR, Panickar KS, Murthy C, Norenberg MD (2006) Oxidative stress and mitogen-activated protein kinase phosphorylation mediate ammonia-induced cell swelling and glutamate uptake inhibition in cultured astrocytes. J Neurosci 26:4774–4784PubMedCrossRef
go back to reference Jayakumar AR, Valdes V, Norenberg MD (2011) The Na-K-Cl cotransporter in the brain edema of acute liver failure. J Hepatol 54:272–278PubMedCrossRef Jayakumar AR, Valdes V, Norenberg MD (2011) The Na-K-Cl cotransporter in the brain edema of acute liver failure. J Hepatol 54:272–278PubMedCrossRef
go back to reference Jayakumar AR, Tong XY, Ospel J, Norenberg MD (2012) Role of cerebral endothelial cells in the astrocyte swelling and brain edema associated with acute hepatic encephalopathy. Neuroscience 218:305–316PubMedCrossRef Jayakumar AR, Tong XY, Ospel J, Norenberg MD (2012) Role of cerebral endothelial cells in the astrocyte swelling and brain edema associated with acute hepatic encephalopathy. Neuroscience 218:305–316PubMedCrossRef
go back to reference Kale RA, Gupta RK, Saraswat VA et al (2006) Demonstration of interstitial cerebral edema with diffusion tensor MR imaging in type C hepatic encephalopathy. Hepatology 43:698–706PubMedCrossRef Kale RA, Gupta RK, Saraswat VA et al (2006) Demonstration of interstitial cerebral edema with diffusion tensor MR imaging in type C hepatic encephalopathy. Hepatology 43:698–706PubMedCrossRef
go back to reference Kanamori K, Ross BD (1995) Steady-state in vivo glutamate dehydrogenase activity in rat brain measured by 15N NMR. J Biol Chem 270:24805–24809PubMedCrossRef Kanamori K, Ross BD (1995) Steady-state in vivo glutamate dehydrogenase activity in rat brain measured by 15N NMR. J Biol Chem 270:24805–24809PubMedCrossRef
go back to reference Kanamori K, Parivar F, Ross BD (1993) A 15N NMR study of in vivo cerebral glutamine synthesis in hyperammonemic rats. NMR Biomed 6:21–26PubMedCrossRef Kanamori K, Parivar F, Ross BD (1993) A 15N NMR study of in vivo cerebral glutamine synthesis in hyperammonemic rats. NMR Biomed 6:21–26PubMedCrossRef
go back to reference Kim IS, Ki CS, Kim JW, Lee M, Jin DK, Lee SY (2006) Characterization of late-onset citrullinemia 1 in a Korean patient: confirmation by argininosuccinate synthetase gene mutation analysis. J Biochem Mol Biol 39:400–405PubMedCrossRef Kim IS, Ki CS, Kim JW, Lee M, Jin DK, Lee SY (2006) Characterization of late-onset citrullinemia 1 in a Korean patient: confirmation by argininosuccinate synthetase gene mutation analysis. J Biochem Mol Biol 39:400–405PubMedCrossRef
go back to reference Kim MJ, Ko JS, Seo JK et al (2012) Clinical features of congenital portosystemic shunt in children. Eur J Pediatr 171:395–400PubMedCrossRef Kim MJ, Ko JS, Seo JK et al (2012) Clinical features of congenital portosystemic shunt in children. Eur J Pediatr 171:395–400PubMedCrossRef
go back to reference Kircheis G, Nilius R, Held C et al (1997) Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double-blind study. Hepatology 25:1351–1360PubMedCrossRef Kircheis G, Nilius R, Held C et al (1997) Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double-blind study. Hepatology 25:1351–1360PubMedCrossRef
go back to reference Klejman A, Wegrzynowicz M, Szatmari EM, Mioduszewska B, Hetman M, Albrecht J (2005) Mechanisms of ammonia-induced cell death in rat cortical neurons: roles of NMDA receptors and glutathione. Neurochem Int 47:51–57PubMedCrossRef Klejman A, Wegrzynowicz M, Szatmari EM, Mioduszewska B, Hetman M, Albrecht J (2005) Mechanisms of ammonia-induced cell death in rat cortical neurons: roles of NMDA receptors and glutathione. Neurochem Int 47:51–57PubMedCrossRef
go back to reference Kosenko E, Kaminsky Y, Kaminsky A et al (1997) Superoxide production and antioxidant enzymes in ammonia intoxication in rats. Free Radic Res 27:637–644PubMedCrossRef Kosenko E, Kaminsky Y, Kaminsky A et al (1997) Superoxide production and antioxidant enzymes in ammonia intoxication in rats. Free Radic Res 27:637–644PubMedCrossRef
go back to reference Kosenko E, Kaminsky Y, Lopata O et al (1998) Nitroarginine, an inhibitor of nitric oxide synthase, prevents changes in superoxide radical and antioxidant enzymes induced by ammonia intoxication. Metab Brain Dis 13:29–41PubMedCrossRef Kosenko E, Kaminsky Y, Lopata O et al (1998) Nitroarginine, an inhibitor of nitric oxide synthase, prevents changes in superoxide radical and antioxidant enzymes induced by ammonia intoxication. Metab Brain Dis 13:29–41PubMedCrossRef
go back to reference Kosenko E, Kaminski Y, Lopata O, Muravyov N, Felipo V (1999) Blocking NMDA receptors prevents the oxidative stress induced by acute ammonia intoxication. Free Radic Biol Med 26:1369–1374PubMedCrossRef Kosenko E, Kaminski Y, Lopata O, Muravyov N, Felipo V (1999) Blocking NMDA receptors prevents the oxidative stress induced by acute ammonia intoxication. Free Radic Biol Med 26:1369–1374PubMedCrossRef
go back to reference Krivitzky L, Babikian T, Lee HS, Thomas NH, Burk-Paull KL, Batshaw ML (2009) Intellectual, adaptive, and behavioral functioning in children with urea cycle disorders. Pediatr Res 66:96–101PubMedCrossRef Krivitzky L, Babikian T, Lee HS, Thomas NH, Burk-Paull KL, Batshaw ML (2009) Intellectual, adaptive, and behavioral functioning in children with urea cycle disorders. Pediatr Res 66:96–101PubMedCrossRef
go back to reference Kurihara A, Takanashi J, Tomita M et al (2003) Magnetic resonance imaging in late-onset ornithine transcarbamylase deficiency. Brain Dev 25:40–44PubMedCrossRef Kurihara A, Takanashi J, Tomita M et al (2003) Magnetic resonance imaging in late-onset ornithine transcarbamylase deficiency. Brain Dev 25:40–44PubMedCrossRef
go back to reference Lai JC, Cooper AJ (1986) Brain alpha-ketoglutarate dehydrogenase complex: kinetic properties, regional distribution, and effects of inhibitors. J Neurochem 47:1376–1386PubMedCrossRef Lai JC, Cooper AJ (1986) Brain alpha-ketoglutarate dehydrogenase complex: kinetic properties, regional distribution, and effects of inhibitors. J Neurochem 47:1376–1386PubMedCrossRef
go back to reference Lanz B, Cudalbu C, Duarte JM, Gruetter R (2011) Direct assessment of increased pyruvate carboxylase in the hyperammonemic brain using 13C MRS. Proc Intl Soc Magn Reson Med 19:2256 Lanz B, Cudalbu C, Duarte JM, Gruetter R (2011) Direct assessment of increased pyruvate carboxylase in the hyperammonemic brain using 13C MRS. Proc Intl Soc Magn Reson Med 19:2256
go back to reference Le Bihan D (ed) (1995) Diffusion and perfusion magnetic resonance imaging: Applications to fonctional MRI. Raven, New York Le Bihan D (ed) (1995) Diffusion and perfusion magnetic resonance imaging: Applications to fonctional MRI. Raven, New York
go back to reference Leonard JV, Ward Platt MP, Morris AA (2008) Hypothesis: proposals for the management of a neonate at risk of hyperammonaemia due to a urea cycle disorder. Eur J Pediatr 167:305–309PubMedCrossRef Leonard JV, Ward Platt MP, Morris AA (2008) Hypothesis: proposals for the management of a neonate at risk of hyperammonaemia due to a urea cycle disorder. Eur J Pediatr 167:305–309PubMedCrossRef
go back to reference Lichter-Konecki U, Mangin JM, Gordish-Dressman H, Hoffman EP, Gallo V (2008) Gene expression profiling of astrocytes from hyperammonemic mice reveals altered pathways for water and potassium homeostasis in vivo. Glia 56:365–377PubMedCrossRef Lichter-Konecki U, Mangin JM, Gordish-Dressman H, Hoffman EP, Gallo V (2008) Gene expression profiling of astrocytes from hyperammonemic mice reveals altered pathways for water and potassium homeostasis in vivo. Glia 56:365–377PubMedCrossRef
go back to reference Majoie CB, Mourmans JM, Akkerman EM, Duran M, Poll-The BT (2004) Neonatal citrullinemia: comparison of conventional MR, diffusion-weighted, and diffusion tensor findings. Am J Neuroradiol 25:32–35PubMed Majoie CB, Mourmans JM, Akkerman EM, Duran M, Poll-The BT (2004) Neonatal citrullinemia: comparison of conventional MR, diffusion-weighted, and diffusion tensor findings. Am J Neuroradiol 25:32–35PubMed
go back to reference Marcaida G, Minana MD, Grisolia S, Felipo V (1995) Lack of correlation between glutamate-induced depletion of ATP and neuronal death in primary cultures of cerebellum. Brain Res 695:146–150PubMedCrossRef Marcaida G, Minana MD, Grisolia S, Felipo V (1995) Lack of correlation between glutamate-induced depletion of ATP and neuronal death in primary cultures of cerebellum. Brain Res 695:146–150PubMedCrossRef
go back to reference McLin VA, Braissant O, van de Looij Y, Kunz N, Gruetter R, Cudalbu C (2012) Assessment of cerebral osmotic regulation in a rat model of biliary cirrhosis using MR spectroscopy. J Hepatol 56(Suppl 2):S231–S232CrossRef McLin VA, Braissant O, van de Looij Y, Kunz N, Gruetter R, Cudalbu C (2012) Assessment of cerebral osmotic regulation in a rat model of biliary cirrhosis using MR spectroscopy. J Hepatol 56(Suppl 2):S231–S232CrossRef
go back to reference Mekle R, Mlynarik V, Gambarota G, Hergt M, Krueger G, Gruetter R (2009) MR spectroscopy of the human brain with enhanced signal intensity at ultrashort echo times on a clinical platform at 3T and 7T. Magn Reson Med 61:1279–1285PubMedCrossRef Mekle R, Mlynarik V, Gambarota G, Hergt M, Krueger G, Gruetter R (2009) MR spectroscopy of the human brain with enhanced signal intensity at ultrashort echo times on a clinical platform at 3T and 7T. Magn Reson Med 61:1279–1285PubMedCrossRef
go back to reference Meyburg J, Hoffmann GF (2010) Liver, liver cell and stem cell transplantation for the treatment of urea cycle defects. Mol Genet Metab 100(Suppl 1):S77–S83PubMedCrossRef Meyburg J, Hoffmann GF (2010) Liver, liver cell and stem cell transplantation for the treatment of urea cycle defects. Mol Genet Metab 100(Suppl 1):S77–S83PubMedCrossRef
go back to reference Michalak A, Butterworth RF (1997) Ornithine transcarbamylase deficiency: pathogenesis of the cerebral disorder and new prospects for therapy. Metab Brain Dis 12:171–182PubMedCrossRef Michalak A, Butterworth RF (1997) Ornithine transcarbamylase deficiency: pathogenesis of the cerebral disorder and new prospects for therapy. Metab Brain Dis 12:171–182PubMedCrossRef
go back to reference Mlynarik V, Cudalbu C, Xin L, Gruetter R (2008a) 1H NMR spectroscopy of rat brain in vivo at 14.1Tesla: improvements in quantification of the neurochemical profile. J Magn Reson 194:163–168PubMedCrossRef Mlynarik V, Cudalbu C, Xin L, Gruetter R (2008a) 1H NMR spectroscopy of rat brain in vivo at 14.1Tesla: improvements in quantification of the neurochemical profile. J Magn Reson 194:163–168PubMedCrossRef
go back to reference Mlynarik V, Kohler I, Gambarota G, Vaslin A, Clarke PG, Gruetter R (2008b) Quantitative proton spectroscopic imaging of the neurochemical profile in rat brain with microliter resolution at ultra-short echo times. Magn Reson Med 59:52–58PubMedCrossRef Mlynarik V, Kohler I, Gambarota G, Vaslin A, Clarke PG, Gruetter R (2008b) Quantitative proton spectroscopic imaging of the neurochemical profile in rat brain with microliter resolution at ultra-short echo times. Magn Reson Med 59:52–58PubMedCrossRef
go back to reference Montoliu C, Gonzalez-Escamilla G, Atienza M et al (2012) Focal cortical damage parallels cognitive impairment in minimal hepatic encephalopathy. NeuroImage 61:1165–1175PubMedCrossRef Montoliu C, Gonzalez-Escamilla G, Atienza M et al (2012) Focal cortical damage parallels cognitive impairment in minimal hepatic encephalopathy. NeuroImage 61:1165–1175PubMedCrossRef
go back to reference Moriyama M, Jayakumar AR, Tong XY, Norenberg MD (2010) Role of mitogen-activated protein kinases in the mechanism of oxidant-induced cell swelling in cultured astrocytes. J Neurosci Res 88:2450–2458PubMed Moriyama M, Jayakumar AR, Tong XY, Norenberg MD (2010) Role of mitogen-activated protein kinases in the mechanism of oxidant-induced cell swelling in cultured astrocytes. J Neurosci Res 88:2450–2458PubMed
go back to reference Msall M, Batshaw ML, Suss R, Brusilow SW, Mellits ED (1984) Neurologic outcome in children with inborn errors of urea synthesis. Outcome of urea-cycle enzymopathies. N Engl J Med 310:1500–1505PubMedCrossRef Msall M, Batshaw ML, Suss R, Brusilow SW, Mellits ED (1984) Neurologic outcome in children with inborn errors of urea synthesis. Outcome of urea-cycle enzymopathies. N Engl J Med 310:1500–1505PubMedCrossRef
go back to reference Munoz MD, Monfort P, Gaztelu JM, Felipo V (2000) Hyperammonemia impairs NMDA receptor-dependent long-term potentiation in the CA1 of rat hippocampus in vitro. Neurochem Res 25:437–441PubMedCrossRef Munoz MD, Monfort P, Gaztelu JM, Felipo V (2000) Hyperammonemia impairs NMDA receptor-dependent long-term potentiation in the CA1 of rat hippocampus in vitro. Neurochem Res 25:437–441PubMedCrossRef
go back to reference Murthy CR, Rama Rao KV, Bai G, Norenberg MD (2001) Ammonia-induced production of free radicals in primary cultures of rat astrocytes. J Neurosci Res 66:282–288PubMedCrossRef Murthy CR, Rama Rao KV, Bai G, Norenberg MD (2001) Ammonia-induced production of free radicals in primary cultures of rat astrocytes. J Neurosci Res 66:282–288PubMedCrossRef
go back to reference Nagasaka H, Komatsu H, Ohura T et al (2004) Nitric oxide synthesis in ornithine transcarbamylase deficiency: possible involvement of low NO synthesis in clinical manifestations of urea cycle defect. J Pediatr 145:259–262PubMedCrossRef Nagasaka H, Komatsu H, Ohura T et al (2004) Nitric oxide synthesis in ornithine transcarbamylase deficiency: possible involvement of low NO synthesis in clinical manifestations of urea cycle defect. J Pediatr 145:259–262PubMedCrossRef
go back to reference Norenberg MD, Rao KV, Jayakumar AR (2005) Mechanisms of ammonia-induced astrocyte swelling. Metab Brain Dis 20:303–318PubMedCrossRef Norenberg MD, Rao KV, Jayakumar AR (2005) Mechanisms of ammonia-induced astrocyte swelling. Metab Brain Dis 20:303–318PubMedCrossRef
go back to reference Nyberg SL, Cerra FB, Gruetter R (1998) Brain lactate by magnetic resonance spectroscopy during fulminant hepatic failure in the dog. Liver Transp Surg 4:158–165CrossRef Nyberg SL, Cerra FB, Gruetter R (1998) Brain lactate by magnetic resonance spectroscopy during fulminant hepatic failure in the dog. Liver Transp Surg 4:158–165CrossRef
go back to reference Obara-Michlewska M, Pannicke T, Karl A et al (2011) Down-regulation of Kir4.1 in the cerebral cortex of rats with liver failure and in cultured astrocytes treated with glutamine: Implications for astrocytic dysfunction in hepatic encephalopathy. J Neurosci Res 89:2018–2027PubMedCrossRef Obara-Michlewska M, Pannicke T, Karl A et al (2011) Down-regulation of Kir4.1 in the cerebral cortex of rats with liver failure and in cultured astrocytes treated with glutamine: Implications for astrocytic dysfunction in hepatic encephalopathy. J Neurosci Res 89:2018–2027PubMedCrossRef
go back to reference Olde Damink SW, Deutz NE, Dejong CH, Soeters PB, Jalan R (2002) Interorgan ammonia metabolism in liver failure. Neurochem Int 41:177–188PubMedCrossRef Olde Damink SW, Deutz NE, Dejong CH, Soeters PB, Jalan R (2002) Interorgan ammonia metabolism in liver failure. Neurochem Int 41:177–188PubMedCrossRef
go back to reference Oldham MS, vanMeter JW, Shattuck KF, Cederbaum SD, Gropman AL (2010) Diffusion tensor imaging in arginase deficiency reveals damage to corticospinal tracts. Pediatr Neurol 42:49–52PubMedCrossRef Oldham MS, vanMeter JW, Shattuck KF, Cederbaum SD, Gropman AL (2010) Diffusion tensor imaging in arginase deficiency reveals damage to corticospinal tracts. Pediatr Neurol 42:49–52PubMedCrossRef
go back to reference Oria M, Romero-Giménez J, Arranz JA, Riudor E, Raquer N, Córdoba J (2012) Ornithine phenylacetate prevents disturbances of motor-evoked potentials induced by intestinal blood in rats with portacaval anastomosis. J Hepatol 56:109–114PubMedCrossRef Oria M, Romero-Giménez J, Arranz JA, Riudor E, Raquer N, Córdoba J (2012) Ornithine phenylacetate prevents disturbances of motor-evoked potentials induced by intestinal blood in rats with portacaval anastomosis. J Hepatol 56:109–114PubMedCrossRef
go back to reference Panickar KS, Jayakumar AR, Rao KV, Norenberg MD (2009) Ammonia-induced activation of p53 in cultured astrocytes: role in cell swelling and glutamate uptake. Neurochem Int 55:98–105PubMedCrossRef Panickar KS, Jayakumar AR, Rao KV, Norenberg MD (2009) Ammonia-induced activation of p53 in cultured astrocytes: role in cell swelling and glutamate uptake. Neurochem Int 55:98–105PubMedCrossRef
go back to reference Patil DH, Westaby D, Mahida YR et al (1987) Comparative modes of action of lactitol and lactulose in the treatment of hepatic encephalopathy. Gut 28:255–259PubMedCrossRef Patil DH, Westaby D, Mahida YR et al (1987) Comparative modes of action of lactitol and lactulose in the treatment of hepatic encephalopathy. Gut 28:255–259PubMedCrossRef
go back to reference Pfeuffer J, Tkác I, Provencher SW, Gruetter R (1999) Toward an in vivo neurochemical profile: quantification of 18 metabolites in short-echo-time 1H NMR spectra of the rat brain. J Magn Reson 141:104–120PubMedCrossRef Pfeuffer J, Tkác I, Provencher SW, Gruetter R (1999) Toward an in vivo neurochemical profile: quantification of 18 metabolites in short-echo-time 1H NMR spectra of the rat brain. J Magn Reson 141:104–120PubMedCrossRef
go back to reference Pietrobattista A, Luciani M, Abraldes JG et al (2010) Extrahepatic portal vein thrombosis in children and adolescents: influence of genetic Thrombophilic disorders. World J Gastroenterol 16:6123–6127PubMedCrossRef Pietrobattista A, Luciani M, Abraldes JG et al (2010) Extrahepatic portal vein thrombosis in children and adolescents: influence of genetic Thrombophilic disorders. World J Gastroenterol 16:6123–6127PubMedCrossRef
go back to reference Qureshi IA, Rao KV (1997) Sparse-fur (spf) mouse as a model of hyperammonemia: alterations in the neurotransmitter systems. Adv Exp Med Biol 420:143–158PubMedCrossRef Qureshi IA, Rao KV (1997) Sparse-fur (spf) mouse as a model of hyperammonemia: alterations in the neurotransmitter systems. Adv Exp Med Biol 420:143–158PubMedCrossRef
go back to reference Rama Rao KV, Jayakumar AR, Tong X, Curtis KM, Norenberg MD (2010) Brain aquaporin-4 in experimental acute liver failure. J Neuropathol Exp Neurol 69:869–879PubMedCrossRef Rama Rao KV, Jayakumar AR, Tong X, Curtis KM, Norenberg MD (2010) Brain aquaporin-4 in experimental acute liver failure. J Neuropathol Exp Neurol 69:869–879PubMedCrossRef
go back to reference Rama Rao KV, Jayakumar AR, Norenberg MD (2012) Glutamine in the pathogenesis of acute hepatic encephalopathy. Neurochem Int 61:575–580PubMedCrossRef Rama Rao KV, Jayakumar AR, Norenberg MD (2012) Glutamine in the pathogenesis of acute hepatic encephalopathy. Neurochem Int 61:575–580PubMedCrossRef
go back to reference Ranjan P, Mishra AM, Kale R, Saraswat VA, Gupta RK (2005) Cytotoxic edema is responsible for raised intracranial pressure in fulminant hepatic failure: in vivo demonstration using diffusion-weighted MRI in human subjects. Metab Brain Dis 20:181–192PubMedCrossRef Ranjan P, Mishra AM, Kale R, Saraswat VA, Gupta RK (2005) Cytotoxic edema is responsible for raised intracranial pressure in fulminant hepatic failure: in vivo demonstration using diffusion-weighted MRI in human subjects. Metab Brain Dis 20:181–192PubMedCrossRef
go back to reference Rao VLR (2002) Nitric oxide in hepatic encephalopathy and hyperammonemia. Neurochem Int 41:161–170PubMedCrossRef Rao VLR (2002) Nitric oxide in hepatic encephalopathy and hyperammonemia. Neurochem Int 41:161–170PubMedCrossRef
go back to reference Rao KV, Qureshi IA (1999) Reduction in the MK-801 binding sites of the NMDA sub-type of glutamate receptor in a mouse model of congenital hyperammonemia: prevention by acetyl-L-carnitine. Neuropharmacology 38:383–394PubMedCrossRef Rao KV, Qureshi IA (1999) Reduction in the MK-801 binding sites of the NMDA sub-type of glutamate receptor in a mouse model of congenital hyperammonemia: prevention by acetyl-L-carnitine. Neuropharmacology 38:383–394PubMedCrossRef
go back to reference Rao KV, Mawal YR, Qureshi IA (1997) Progressive decrease of cerebral cytochrome C oxidase activity in sparse-fur mice: role of acetyl-L-carnitine in restoring the ammonia-induced cerebral energy depletion. Neurosci Lett 224:83–86PubMedCrossRef Rao KV, Mawal YR, Qureshi IA (1997) Progressive decrease of cerebral cytochrome C oxidase activity in sparse-fur mice: role of acetyl-L-carnitine in restoring the ammonia-induced cerebral energy depletion. Neurosci Lett 224:83–86PubMedCrossRef
go back to reference Ratnakumari L, Qureshi IA, Butterworth RF (1992) Effects of congenital hyperammonemia on the cerebral and hepatic levels of the intermediates of energy metabolism in spf mice. Biochem Biophys Res Commun 184:746–751PubMedCrossRef Ratnakumari L, Qureshi IA, Butterworth RF (1992) Effects of congenital hyperammonemia on the cerebral and hepatic levels of the intermediates of energy metabolism in spf mice. Biochem Biophys Res Commun 184:746–751PubMedCrossRef
go back to reference Ratnakumari L, Qureshi IA, Butterworth RF (1994a) Regional amino acid neurotransmitter changes in brains of spf/Y mice with congenital ornithine transcarbamylase deficiency. Metab Brain Dis 9:43–51PubMedCrossRef Ratnakumari L, Qureshi IA, Butterworth RF (1994a) Regional amino acid neurotransmitter changes in brains of spf/Y mice with congenital ornithine transcarbamylase deficiency. Metab Brain Dis 9:43–51PubMedCrossRef
go back to reference Ratnakumari L, Qureshi IA, Butterworth RF (1994b) Evidence for cholinergic neuronal loss in brain in congenital ornithine transcarbamylase deficiency. Neurosci Lett 178:63–65PubMedCrossRef Ratnakumari L, Qureshi IA, Butterworth RF (1994b) Evidence for cholinergic neuronal loss in brain in congenital ornithine transcarbamylase deficiency. Neurosci Lett 178:63–65PubMedCrossRef
go back to reference Ratnakumari L, Qureshi IA, Maysinger D, Butterworth RF (1995) Developmental deficiency of the cholinergic system in congenitally hyperammonemic spf mice: effect of acetyl-L-carnitine. J Pharmacol Exp Ther 274:437–443PubMed Ratnakumari L, Qureshi IA, Maysinger D, Butterworth RF (1995) Developmental deficiency of the cholinergic system in congenitally hyperammonemic spf mice: effect of acetyl-L-carnitine. J Pharmacol Exp Ther 274:437–443PubMed
go back to reference Ratnakumari L, Qureshi IA, Butterworth RF (1996a) Central muscarinic cholinergic M1 and M2 receptor changes in congenital ornithine transcarbamylase deficiency. Pediatr Res 40:25–28PubMedCrossRef Ratnakumari L, Qureshi IA, Butterworth RF (1996a) Central muscarinic cholinergic M1 and M2 receptor changes in congenital ornithine transcarbamylase deficiency. Pediatr Res 40:25–28PubMedCrossRef
go back to reference Ratnakumari L, Qureshi IA, Butterworth RF, Marescau B, De Deyn PP (1996b) Arginine-related guanidino compounds and nitric oxide synthase in the brain of ornithine transcarbamylase deficient spf mutant mouse: effect of metabolic arginine deficiency. Neurosci Lett 215:153–156PubMedCrossRef Ratnakumari L, Qureshi IA, Butterworth RF, Marescau B, De Deyn PP (1996b) Arginine-related guanidino compounds and nitric oxide synthase in the brain of ornithine transcarbamylase deficient spf mutant mouse: effect of metabolic arginine deficiency. Neurosci Lett 215:153–156PubMedCrossRef
go back to reference Reinehr R, Gorg B, Becker S et al (2007) Hypoosmotic swelling and ammonia increase oxidative stress by NADPH oxidase in cultured astrocytes and vital brain slices. Glia 55:758–771PubMedCrossRef Reinehr R, Gorg B, Becker S et al (2007) Hypoosmotic swelling and ammonia increase oxidative stress by NADPH oxidase in cultured astrocytes and vital brain slices. Glia 55:758–771PubMedCrossRef
go back to reference Robinson MB, Anegawa NJ, Gorry E et al (1992a) Brain serotonin2 and serotonin1A receptors are altered in the congenitally hyperammonemic sparse fur mouse. J Neurochem 58:1016–1022PubMedCrossRef Robinson MB, Anegawa NJ, Gorry E et al (1992a) Brain serotonin2 and serotonin1A receptors are altered in the congenitally hyperammonemic sparse fur mouse. J Neurochem 58:1016–1022PubMedCrossRef
go back to reference Robinson MB, Heyes MP, Anegawa NJ et al (1992b) Quinolinate in brain and cerebrospinal fluid in rat models of congenital hyperammonemia. Pediatr Res 32:483–488PubMedCrossRef Robinson MB, Heyes MP, Anegawa NJ et al (1992b) Quinolinate in brain and cerebrospinal fluid in rat models of congenital hyperammonemia. Pediatr Res 32:483–488PubMedCrossRef
go back to reference Rodrigo R, Erceg S, Felipo V (2005) Neurons exposed to ammonia reproduce the differential alteration in nitric oxide modulation of guanylate cyclase in the cerebellum and cortex of patients with liver cirrhosis. Neurobiol Dis 19:150–161PubMedCrossRef Rodrigo R, Erceg S, Felipo V (2005) Neurons exposed to ammonia reproduce the differential alteration in nitric oxide modulation of guanylate cyclase in the cerebellum and cortex of patients with liver cirrhosis. Neurobiol Dis 19:150–161PubMedCrossRef
go back to reference Rodrigo R, Cauli O, Boix J, El Mlili N, Agusti A, Felipo V (2009) Role of NMDA receptors in acute liver failure and ammonia toxicity: therapeutical implications. Neurochem Int 55:113–118PubMedCrossRef Rodrigo R, Cauli O, Boix J, El Mlili N, Agusti A, Felipo V (2009) Role of NMDA receptors in acute liver failure and ammonia toxicity: therapeutical implications. Neurochem Int 55:113–118PubMedCrossRef
go back to reference Rose C (2006) Effect of ammonia on astrocytic glutamate uptake/release mechanisms. J Neurochem 97(Suppl 1):11–15PubMedCrossRef Rose C (2006) Effect of ammonia on astrocytic glutamate uptake/release mechanisms. J Neurochem 97(Suppl 1):11–15PubMedCrossRef
go back to reference Rose C, Felipo V (2005) Limited capacity for ammonia removal by brain in chronic liver failure: potential role of nitric oxide. Metab Brain Dis 20:275–283PubMedCrossRef Rose C, Felipo V (2005) Limited capacity for ammonia removal by brain in chronic liver failure: potential role of nitric oxide. Metab Brain Dis 20:275–283PubMedCrossRef
go back to reference Rose C, Michalak A, Pannunzio P et al (1998) L-ornithine-L-aspartate in experimental portal-systemic encephalopathy: therapeutic efficacy and mechanism of action. Metab Brain Dis 13:147–157PubMedCrossRef Rose C, Michalak A, Pannunzio P et al (1998) L-ornithine-L-aspartate in experimental portal-systemic encephalopathy: therapeutic efficacy and mechanism of action. Metab Brain Dis 13:147–157PubMedCrossRef
go back to reference Rose C, Kresse W, Kettenmann H (2005) Acute insult of ammonia leads to calcium-dependent glutamate release from cultured astrocytes, an effect of pH. J Biol Chem 280:20937–20944PubMedCrossRef Rose C, Kresse W, Kettenmann H (2005) Acute insult of ammonia leads to calcium-dependent glutamate release from cultured astrocytes, an effect of pH. J Biol Chem 280:20937–20944PubMedCrossRef
go back to reference Rovira A, Alonso J, Cordoba J (2008) MR imaging findings in hepatic encephalopathy. Am J Neuroradiol 29:1612–1621PubMedCrossRef Rovira A, Alonso J, Cordoba J (2008) MR imaging findings in hepatic encephalopathy. Am J Neuroradiol 29:1612–1621PubMedCrossRef
go back to reference Salvi S, Santorelli FM, Bertini E et al (2001) Clinical and molecular findings in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome. Neurology 57:911–914PubMedCrossRef Salvi S, Santorelli FM, Bertini E et al (2001) Clinical and molecular findings in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome. Neurology 57:911–914PubMedCrossRef
go back to reference Scaglia F (2010) New insights in nutritional management and amino acid supplementation in urea cycle disorders. Mol Genet Metab 100(Suppl 1):S72–S76PubMedCrossRef Scaglia F (2010) New insights in nutritional management and amino acid supplementation in urea cycle disorders. Mol Genet Metab 100(Suppl 1):S72–S76PubMedCrossRef
go back to reference Scaglia F, Lee B (2006) Clinical, biochemical, and molecular spectrum of hyperargininemia due to arginase I deficiency. Am J Med Genet C Semin Med Genet 142:113–120 Scaglia F, Lee B (2006) Clinical, biochemical, and molecular spectrum of hyperargininemia due to arginase I deficiency. Am J Med Genet C Semin Med Genet 142:113–120
go back to reference Scaglia F, Zheng Q, O'Brien WE et al (2002) An integrated approach to the diagnosis and prospective management of partial ornithine transcarbamylase deficiency. Pediatrics 109:150–152PubMedCrossRef Scaglia F, Zheng Q, O'Brien WE et al (2002) An integrated approach to the diagnosis and prospective management of partial ornithine transcarbamylase deficiency. Pediatrics 109:150–152PubMedCrossRef
go back to reference Scaglia F, Brunetti-Pierri N, Kleppe S et al (2004) Clinical consequences of urea cycle enzyme deficiencies and potential links to arginine and nitric oxide metabolism. J Nutr 134:2775S–2782SPubMed Scaglia F, Brunetti-Pierri N, Kleppe S et al (2004) Clinical consequences of urea cycle enzyme deficiencies and potential links to arginine and nitric oxide metabolism. J Nutr 134:2775S–2782SPubMed
go back to reference Schliess F, Gorg B, Fischer R et al (2002) Ammonia induces MK-801-sensitive nitration and phosphorylation of protein tyrosine residues in rat astrocytes. FASEB J 16:739–741PubMed Schliess F, Gorg B, Fischer R et al (2002) Ammonia induces MK-801-sensitive nitration and phosphorylation of protein tyrosine residues in rat astrocytes. FASEB J 16:739–741PubMed
go back to reference Shen J, Sibson NR, Cline G, Behar KL, Rothman DL, Shulman RG (1998) 15N-NMR spectroscopy studies of ammonia transport and glutamine synthesis in the hyperammonemic rat brain. Dev Neurosci 20:434–443PubMedCrossRef Shen J, Sibson NR, Cline G, Behar KL, Rothman DL, Shulman RG (1998) 15N-NMR spectroscopy studies of ammonia transport and glutamine synthesis in the hyperammonemic rat brain. Dev Neurosci 20:434–443PubMedCrossRef
go back to reference Sibson NR, Dhankhar A, Mason GF, Behar KL, Rothman DL, Shulman RG (1997) In vivo 13C NMR measurements of cerebral glutamine synthesis as evidence for glutamate-glutamine cycling. Proc Natl Acad Sci USA 94:2699–2704PubMedCrossRef Sibson NR, Dhankhar A, Mason GF, Behar KL, Rothman DL, Shulman RG (1997) In vivo 13C NMR measurements of cerebral glutamine synthesis as evidence for glutamate-glutamine cycling. Proc Natl Acad Sci USA 94:2699–2704PubMedCrossRef
go back to reference Sibson NR, Mason GF, Shen J et al (2001) In vivo 13C NMR measurement of neurotransmitter glutamate cycling, anaplerosis and TCA cycle flux in rat brain during [2-13C]glucose infusion. J Neurochem 76:975–989PubMedCrossRef Sibson NR, Mason GF, Shen J et al (2001) In vivo 13C NMR measurement of neurotransmitter glutamate cycling, anaplerosis and TCA cycle flux in rat brain during [2-13C]glucose infusion. J Neurochem 76:975–989PubMedCrossRef
go back to reference Skowronska M, Zielinska M, Wojcik-Stanaszek L et al (2012) Ammonia increases paracellular permeability of rat brain endothelial cells by a mechanism encompassing oxidative/nitrosative stress and activation of matrix metalloproteinases. J Neurochem 121:125–134PubMedCrossRef Skowronska M, Zielinska M, Wojcik-Stanaszek L et al (2012) Ammonia increases paracellular permeability of rat brain endothelial cells by a mechanism encompassing oxidative/nitrosative stress and activation of matrix metalloproteinases. J Neurochem 121:125–134PubMedCrossRef
go back to reference Smith W, Kishnani PS, Lee B et al (2005) Urea cycle disorders: clinical presentation outside the newborn period. Crit Care Clin 21:S9–S17PubMedCrossRef Smith W, Kishnani PS, Lee B et al (2005) Urea cycle disorders: clinical presentation outside the newborn period. Crit Care Clin 21:S9–S17PubMedCrossRef
go back to reference Song G, Dhodda VK, Blei AT, Dempsey RJ, Rao VL (2002) GeneChip analysis shows altered mRNA expression of transcripts of neurotransmitter and signal transduction pathways in the cerebral cortex of portacaval shunted rats. J Neurosci Res 68:730–737PubMedCrossRef Song G, Dhodda VK, Blei AT, Dempsey RJ, Rao VL (2002) GeneChip analysis shows altered mRNA expression of transcripts of neurotransmitter and signal transduction pathways in the cerebral cortex of portacaval shunted rats. J Neurosci Res 68:730–737PubMedCrossRef
go back to reference Spahr L, Burkhard PR, Grotzsch H, Hadengue A (2002) Clinical significance of basal ganglia alterations at brain MRI and 1H MRS in cirrhosis and role in the pathogenesis of hepatic encephalopathy. Metab Brain Dis 17:399–413PubMedCrossRef Spahr L, Burkhard PR, Grotzsch H, Hadengue A (2002) Clinical significance of basal ganglia alterations at brain MRI and 1H MRS in cirrhosis and role in the pathogenesis of hepatic encephalopathy. Metab Brain Dis 17:399–413PubMedCrossRef
go back to reference Summar M (2001) Current strategies for the management of neonatal urea cycle disorders. J Pediatr 138:S30–S39PubMedCrossRef Summar M (2001) Current strategies for the management of neonatal urea cycle disorders. J Pediatr 138:S30–S39PubMedCrossRef
go back to reference Takanashi J, Kurihara A, Tomita M et al (2002) Distinctly abnormal brain metabolism in late-onset ornithine transcarbamylase deficiency. Neurology 59:210–214PubMedCrossRef Takanashi J, Kurihara A, Tomita M et al (2002) Distinctly abnormal brain metabolism in late-onset ornithine transcarbamylase deficiency. Neurology 59:210–214PubMedCrossRef
go back to reference Takanashi J, Barkovich AJ, Cheng SF et al (2003) Brain MR imaging in neonatal hyperammonemic encephalopathy resulting from proximal urea cycle disorders. Am J Neuroradiol 24:1184–1187PubMed Takanashi J, Barkovich AJ, Cheng SF et al (2003) Brain MR imaging in neonatal hyperammonemic encephalopathy resulting from proximal urea cycle disorders. Am J Neuroradiol 24:1184–1187PubMed
go back to reference Takeoka M, Soman TB, Shih VE, Caviness VS, Krishnamoorthy KS (2001) Carbamyl phosphate synthetase 1 deficiency: a destructive encephalopathy. Pediatr Neurol 24:193–199PubMedCrossRef Takeoka M, Soman TB, Shih VE, Caviness VS, Krishnamoorthy KS (2001) Carbamyl phosphate synthetase 1 deficiency: a destructive encephalopathy. Pediatr Neurol 24:193–199PubMedCrossRef
go back to reference Tanigami H, Rebel A, Martin LJ et al (2005) Effect of glutamine synthetase inhibition on astrocyte swelling and altered astroglial protein expression during hyperammonemia in rats. Neuroscience 131:437–449PubMedCrossRef Tanigami H, Rebel A, Martin LJ et al (2005) Effect of glutamine synthetase inhibition on astrocyte swelling and altered astroglial protein expression during hyperammonemia in rats. Neuroscience 131:437–449PubMedCrossRef
go back to reference Tkác I, Starcuk Z, Choi IY, Gruetter R (1999) In vivo 1H NMR spectroscopy of rat brain at 1 ms echo time. Magn Reson Med 41:649–656PubMedCrossRef Tkác I, Starcuk Z, Choi IY, Gruetter R (1999) In vivo 1H NMR spectroscopy of rat brain at 1 ms echo time. Magn Reson Med 41:649–656PubMedCrossRef
go back to reference Tkác I, Oz G, Adriany G, Ugurbil K, Gruetter R (2009) In vivo 1H NMR spectroscopy of the human brain at high magnetic fields: metabolite quantification at 4T vs. 7T. Magn Reson Med 62:868–879PubMedCrossRef Tkác I, Oz G, Adriany G, Ugurbil K, Gruetter R (2009) In vivo 1H NMR spectroscopy of the human brain at high magnetic fields: metabolite quantification at 4T vs. 7T. Magn Reson Med 62:868–879PubMedCrossRef
go back to reference Tuchman M, Lee B, Lichter-Konecki U et al (2008) Cross-sectional multicenter study of patients with urea cycle disorders in the United States. Mol Genet Metab 94:397–402PubMedCrossRef Tuchman M, Lee B, Lichter-Konecki U et al (2008) Cross-sectional multicenter study of patients with urea cycle disorders in the United States. Mol Genet Metab 94:397–402PubMedCrossRef
go back to reference Uchino T, Endo F, Matsuda I (1998) Neurodevelopmental outcome of long-term therapy of urea cycle disorders in Japan. J Inherit Metab Dis 21(Suppl 1):151–159PubMedCrossRef Uchino T, Endo F, Matsuda I (1998) Neurodevelopmental outcome of long-term therapy of urea cycle disorders in Japan. J Inherit Metab Dis 21(Suppl 1):151–159PubMedCrossRef
go back to reference Veres G, Gibbs RA, Scherer SE, Caskey CT (1987) The molecular basis of the sparse fur mouse mutation. Science 237:415–417PubMedCrossRef Veres G, Gibbs RA, Scherer SE, Caskey CT (1987) The molecular basis of the sparse fur mouse mutation. Science 237:415–417PubMedCrossRef
go back to reference Walker V (2009) Ammonia toxicity and its prevention in inherited defects of the urea cycle. Diabetes Obes Metab 11:823–835PubMedCrossRef Walker V (2009) Ammonia toxicity and its prevention in inherited defects of the urea cycle. Diabetes Obes Metab 11:823–835PubMedCrossRef
go back to reference Wiesinger H (2001) Arginine metabolism and the synthesis of nitric oxide in the nervous system. Prog Neurobiol 64:365–391PubMedCrossRef Wiesinger H (2001) Arginine metabolism and the synthesis of nitric oxide in the nervous system. Prog Neurobiol 64:365–391PubMedCrossRef
go back to reference Willard-Mack CL, Koehler RC, Hirata T et al (1996) Inhibition of glutamine synthetase reduces ammonia-induced astrocyte swelling in rat. Neuroscience 71:589–599PubMedCrossRef Willard-Mack CL, Koehler RC, Hirata T et al (1996) Inhibition of glutamine synthetase reduces ammonia-induced astrocyte swelling in rat. Neuroscience 71:589–599PubMedCrossRef
go back to reference Wright G, Vairappan B, Stadlbauer V, Mookerjee RP, Davies NA, Jalan R (2012) Reduction in hyperammonaemia by ornithine phenylacetate prevents lipopolysaccharide-induced brain edema and coma in cirrhotic rats. Liver Int 32:410–419PubMed Wright G, Vairappan B, Stadlbauer V, Mookerjee RP, Davies NA, Jalan R (2012) Reduction in hyperammonaemia by ornithine phenylacetate prevents lipopolysaccharide-induced brain edema and coma in cirrhotic rats. Liver Int 32:410–419PubMed
go back to reference Yadav SK, Srivastava A, Thomas MA et al (2010) Encephalopathy assessment in children with extra-hepatic portal vein obstruction with MR, psychometry and critical flicker frequency. J Hepatol 52:348–354PubMedCrossRef Yadav SK, Srivastava A, Thomas MA et al (2010) Encephalopathy assessment in children with extra-hepatic portal vein obstruction with MR, psychometry and critical flicker frequency. J Hepatol 52:348–354PubMedCrossRef
go back to reference Yamanouchi H, Yokoo H, Yuhara Y et al (2002) An autopsy case of ornithine transcarbamylase deficiency. Brain Dev 24:91–94PubMedCrossRef Yamanouchi H, Yokoo H, Yuhara Y et al (2002) An autopsy case of ornithine transcarbamylase deficiency. Brain Dev 24:91–94PubMedCrossRef
go back to reference Zanelli SA, Solenski NJ, Rosenthal RE, Fiskum G (2005) Mechanisms of ischemic neuroprotection by acetyl-L-carnitine. Ann N Y Acad Sci 1053:153–161PubMedCrossRef Zanelli SA, Solenski NJ, Rosenthal RE, Fiskum G (2005) Mechanisms of ischemic neuroprotection by acetyl-L-carnitine. Ann N Y Acad Sci 1053:153–161PubMedCrossRef
go back to reference Zielinska M, Ruszkiewicz J, Hilgier W, Fresko I, Albrecht J (2011) Hyperammonemia increases the expression and activity of the glutamine/arginine transporter y+ LAT2 in rat cerebral cortex: implications for the nitric oxide/cGMP pathway. Neurochem Int 58:190–195PubMedCrossRef Zielinska M, Ruszkiewicz J, Hilgier W, Fresko I, Albrecht J (2011) Hyperammonemia increases the expression and activity of the glutamine/arginine transporter y+ LAT2 in rat cerebral cortex: implications for the nitric oxide/cGMP pathway. Neurochem Int 58:190–195PubMedCrossRef
go back to reference Zielinska M, Skowronska M, Fresko I, Albrecht J (2012) Upregulation of the heteromeric y(+)LAT2 transporter contributes to ammonia-induced increase of arginine uptake in rat cerebral cortical astrocytes. Neurochem Int 61:531–535PubMedCrossRef Zielinska M, Skowronska M, Fresko I, Albrecht J (2012) Upregulation of the heteromeric y(+)LAT2 transporter contributes to ammonia-induced increase of arginine uptake in rat cerebral cortical astrocytes. Neurochem Int 61:531–535PubMedCrossRef
go back to reference Zwingmann C (2007) The anaplerotic flux and ammonia detoxification in hepatic encephalopathy. Metab Brain Dis 22:235–249PubMedCrossRef Zwingmann C (2007) The anaplerotic flux and ammonia detoxification in hepatic encephalopathy. Metab Brain Dis 22:235–249PubMedCrossRef
go back to reference Zwingmann C, Chatauret N, Leibfritz D, Butterworth RF (2003) Selective increase of brain lactate synthesis in experimental acute liver failure: results of a [1H-13C] nuclear magnetic resonance study. Hepatology 37:420–428PubMedCrossRef Zwingmann C, Chatauret N, Leibfritz D, Butterworth RF (2003) Selective increase of brain lactate synthesis in experimental acute liver failure: results of a [1H-13C] nuclear magnetic resonance study. Hepatology 37:420–428PubMedCrossRef
Metadata
Title
Ammonia toxicity to the brain
Authors
Olivier Braissant
Valérie A. McLin
Cristina Cudalbu
Publication date
01-07-2013
Publisher
Springer Netherlands
Published in
Journal of Inherited Metabolic Disease / Issue 4/2013
Print ISSN: 0141-8955
Electronic ISSN: 1573-2665
DOI
https://doi.org/10.1007/s10545-012-9546-2

Other articles of this Issue 4/2013

Journal of Inherited Metabolic Disease 4/2013 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.