Skip to main content
Top
Published in: Journal of Inherited Metabolic Disease 4/2012

01-07-2012 | SSIEM Symposium 2011

Creatine and guanidinoacetate transport at blood-brain and blood-cerebrospinal fluid barriers

Author: Olivier Braissant

Published in: Journal of Inherited Metabolic Disease | Issue 4/2012

Login to get access

Abstract

While it was thought that most of cerebral creatine is of peripheral origin, AGAT and GAMT are well expressed in CNS where brain cells synthesize creatine. While the creatine transporter SLC6A8 is expressed by microcapillary endothelial cells (MCEC) at blood-brain barrier (BBB), it is absent from their surrounding astrocytes. This raised the concept that BBB has a limited permeability for peripheral creatine, and that the brain supplies a part of its creatine by endogenous synthesis. This review brings together the latest data on creatine and guanidinoacetate transport through BBB and blood-CSF barrier (BCSFB) with the clinical evidence of AGAT-, GAMT- and SLC6A8-deficient patients, in order to delineate a clearer view on the roles of BBB and BCSFB in the transport of creatine and guanidinoacetate between periphery and CNS, and on brain synthesis and transport of creatine. It shows that in physiological conditions, creatine is taken up by CNS from periphery through SLC6A8 at BBB, but in limited amounts, and that CNS also needs its own creatine synthesis. No uptake of guanidinoacetate from periphery occurs at BBB except under GAMT deficiency, but a net exit of guanidinoacetate seems to occur from CSF to blood at BCSFB, predominantly through the taurine transporter TauT.
Literature
go back to reference Acosta ML, Kalloniatis M, Christie DL (2005) Creatine transporter localization in developing and adult retina: importance of creatine to retinal function. Am J Physiol Cell Physiol 289:C1015–C1023PubMedCrossRef Acosta ML, Kalloniatis M, Christie DL (2005) Creatine transporter localization in developing and adult retina: importance of creatine to retinal function. Am J Physiol Cell Physiol 289:C1015–C1023PubMedCrossRef
go back to reference Adcock KH, Nedelcu J, Loenneker T, Martin E, Wallimann T, Wagner BP (2002) Neuroprotection of creatine supplementation in neonatal rats with transient cerebral hypoxia-ischemia. Dev Neurosci 24:382–388PubMedCrossRef Adcock KH, Nedelcu J, Loenneker T, Martin E, Wallimann T, Wagner BP (2002) Neuroprotection of creatine supplementation in neonatal rats with transient cerebral hypoxia-ischemia. Dev Neurosci 24:382–388PubMedCrossRef
go back to reference Adriano E, Garbati P, Damonte G, Salis A, Armirotti A, Balestrino M (in press) Searching for a therapy of creatine transporter deficiency: Some effects of creatine ethyl ester in brain slices in vitro. Neuroscience Adriano E, Garbati P, Damonte G, Salis A, Armirotti A, Balestrino M (in press) Searching for a therapy of creatine transporter deficiency: Some effects of creatine ethyl ester in brain slices in vitro. Neuroscience
go back to reference Almeida LS, Verhoeven NM, Roos B et al. (2004) Creatine and guanidinoacetate: diagnostic markers for inborn errors in creatine biosynthesis and transport. Mol Genet Metab 82:214–219PubMedCrossRef Almeida LS, Verhoeven NM, Roos B et al. (2004) Creatine and guanidinoacetate: diagnostic markers for inborn errors in creatine biosynthesis and transport. Mol Genet Metab 82:214–219PubMedCrossRef
go back to reference Almeida LS, Salomons GS, Hogenboom F, Jakobs C, Schoffelmeer AN (2006) Exocytotic release of creatine in rat brain. Synapse 60:118–123PubMedCrossRef Almeida LS, Salomons GS, Hogenboom F, Jakobs C, Schoffelmeer AN (2006) Exocytotic release of creatine in rat brain. Synapse 60:118–123PubMedCrossRef
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 Balestrino M, Lensman M, Parodi M, Perasso L, Rebaudo R, Melani R, Polenov S, Cupello A (2002) Role of creatine and phosphocreatine in neuronal protection from anoxic and ischemic damage. Amino Acids 23:221–229PubMedCrossRef Balestrino M, Lensman M, Parodi M, Perasso L, Rebaudo R, Melani R, Polenov S, Cupello A (2002) Role of creatine and phosphocreatine in neuronal protection from anoxic and ischemic damage. Amino Acids 23:221–229PubMedCrossRef
go back to reference Battini R, Leuzzi V, Carducci C et al. (2002) Creatine depletion in a new case with AGAT deficiency: clinical and genetic study in a large pedigree. Mol Genet Metab 77:326–331PubMedCrossRef Battini R, Leuzzi V, Carducci C et al. (2002) Creatine depletion in a new case with AGAT deficiency: clinical and genetic study in a large pedigree. Mol Genet Metab 77:326–331PubMedCrossRef
go back to reference Battini R, Alessandri MG, Leuzzi V et al. (2006) Arginine:glycine amidinotransferase (AGAT) deficiency in a newborn: early treatment can prevent phenotypic expression of the disease. J Pediatr 148:828–830PubMedCrossRef Battini R, Alessandri MG, Leuzzi V et al. (2006) Arginine:glycine amidinotransferase (AGAT) deficiency in a newborn: early treatment can prevent phenotypic expression of the disease. J Pediatr 148:828–830PubMedCrossRef
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 Bender A, Koch W, Elstner M et al. (2006) Creatine supplementation in Parkinson disease: a placebo-controlled randomized pilot trial. Neurology 67:1262–1264PubMedCrossRef Bender A, Koch W, Elstner M et al. (2006) Creatine supplementation in Parkinson disease: a placebo-controlled randomized pilot trial. Neurology 67:1262–1264PubMedCrossRef
go back to reference Bizzi A, Bugiani M, Salomons GS et al. (2002) X-linked creatine deficiency syndrome: a novel mutation in creatine transporter gene SLC6A8. Ann Neurol 52:227–231PubMedCrossRef Bizzi A, Bugiani M, Salomons GS et al. (2002) X-linked creatine deficiency syndrome: a novel mutation in creatine transporter gene SLC6A8. Ann Neurol 52:227–231PubMedCrossRef
go back to reference Bothwell JH, Styles P, Bhakoo KK (2002) Swelling-activated taurine and creatine effluxes from rat cortical astrocytes are pharmacologically distinct. J Membr Biol 185:157–164PubMedCrossRef Bothwell JH, Styles P, Bhakoo KK (2002) Swelling-activated taurine and creatine effluxes from rat cortical astrocytes are pharmacologically distinct. J Membr Biol 185:157–164PubMedCrossRef
go back to reference Braissant O (2010a) 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 (2010a) 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 (2010b) Current concepts in the pathogenesis of urea cycle disorders. Mol Gen Metab 100(Suppl 1):S3–S12CrossRef Braissant O (2010b) Current concepts in the pathogenesis of urea cycle disorders. Mol Gen Metab 100(Suppl 1):S3–S12CrossRef
go back to reference Braissant O, Henry H (2008) AGAT, GAMT and SLC6A8 distribution in the central nervous system, in relation to creatine deficiency syndromes: a review. J Inher Metab Dis 31:230–239CrossRef Braissant O, Henry H (2008) AGAT, GAMT and SLC6A8 distribution in the central nervous system, in relation to creatine deficiency syndromes: a review. J Inher Metab Dis 31:230–239CrossRef
go back to reference Braissant O, Gotoh T, Loup M, Mori M, Bachmann C (1999) 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 (1999) 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, Gotoh T, Loup M, Mori M, Bachmann C (2001a) 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 (2001a) 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, Loup M, Eilers B, Bachmann C (2001b) Endogenous synthesis and transport of creatine in the rat brain: an in situ hybridization study. Mol Brain Res 86:193–201PubMedCrossRef Braissant O, Henry H, Loup M, Eilers B, Bachmann C (2001b) Endogenous synthesis and transport of creatine in the rat brain: an in situ hybridization study. Mol Brain Res 86:193–201PubMedCrossRef
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, Henry H, Villard AM, Speer O, Wallimann T, Bachmann C (2005) Creatine synthesis and transport during rat embryogenesis: spatiotemporal expression of AGAT, GAMT and CT1. BMC Dev Biol 5:9PubMedCrossRef Braissant O, Henry H, Villard AM, Speer O, Wallimann T, Bachmann C (2005) Creatine synthesis and transport during rat embryogenesis: spatiotemporal expression of AGAT, GAMT and CT1. BMC Dev Biol 5:9PubMedCrossRef
go back to reference Braissant O, Bachmann C, Henry H (2007) Expression and function of AGAT, GAMT and CT1 in the mammalian brain. Subcell Biochem 46:67–81PubMedCrossRef Braissant O, Bachmann C, Henry H (2007) Expression and function of AGAT, GAMT and CT1 in the mammalian brain. Subcell Biochem 46:67–81PubMedCrossRef
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, Béard E, Torrent C, Henry H (2010) Dissociation of AGAT, GAMT and SLC6A8 in CNS: relevance to creatine deficiency syndromes. Neurobiol Dis 37:423–433PubMedCrossRef Braissant O, Béard E, Torrent C, Henry H (2010) Dissociation of AGAT, GAMT and SLC6A8 in CNS: relevance to creatine deficiency syndromes. Neurobiol Dis 37:423–433PubMedCrossRef
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 (2007) Creatine: endogenous metabolite, dietary, and therapeutic supplement. Annu Rev Nutr 27:241–261PubMedCrossRef Brosnan JT, Brosnan ME (2007) Creatine: endogenous metabolite, dietary, and therapeutic supplement. Annu Rev Nutr 27:241–261PubMedCrossRef
go back to reference Burov S, Leko M, Dorosh M, Dobrodumov A, Veselkina O (2011) Creatinyl amino acids-new hybrid compounds with neuroprotective activity. J Pept Sci 17:620–626PubMedCrossRef Burov S, Leko M, Dorosh M, Dobrodumov A, Veselkina O (2011) Creatinyl amino acids-new hybrid compounds with neuroprotective activity. J Pept Sci 17:620–626PubMedCrossRef
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 Cardoso FL, Brites D, Brito MA (2010) Looking at the blood-brain barrier: molecular anatomy and possible investigation approaches. Brain Res Rev 64:328–363PubMedCrossRef Cardoso FL, Brites D, Brito MA (2010) Looking at the blood-brain barrier: molecular anatomy and possible investigation approaches. Brain Res Rev 64:328–363PubMedCrossRef
go back to reference Cecil KM, Salomons GS, Ball WS et al. (2001) Irreversible brain creatine deficiency with elevated serum and urine creatine: a creatine transporter defect? Ann Neurol 49:401–404PubMedCrossRef Cecil KM, Salomons GS, Ball WS et al. (2001) Irreversible brain creatine deficiency with elevated serum and urine creatine: a creatine transporter defect? Ann Neurol 49:401–404PubMedCrossRef
go back to reference Cecil KM, DeGrauw TJ, Salomons GS, Jakobs C, Egelhoff JC, Clark JF (2003) Magnetic resonance spectroscopy in a 9-day-old heterozygous female child with creatine transporter deficiency. J Comput Assist Tomogr 27:44–47PubMedCrossRef Cecil KM, DeGrauw TJ, Salomons GS, Jakobs C, Egelhoff JC, Clark JF (2003) Magnetic resonance spectroscopy in a 9-day-old heterozygous female child with creatine transporter deficiency. J Comput Assist Tomogr 27:44–47PubMedCrossRef
go back to reference Chanutin A (1927) A study on the effect of creatine on growth and its distribution in the tissues of normal rats. J Biol Chem 75:549–557 Chanutin A (1927) A study on the effect of creatine on growth and its distribution in the tissues of normal rats. J Biol Chem 75:549–557
go back to reference Chilosi A, Leuzzi V, Battini R et al. (2008) Treatment with L-arginine improves neuropsychological disorders in a child with creatine transporter defect. Neurocase 14:151–161PubMedCrossRef Chilosi A, Leuzzi V, Battini R et al. (2008) Treatment with L-arginine improves neuropsychological disorders in a child with creatine transporter defect. Neurocase 14:151–161PubMedCrossRef
go back to reference Davis BM, Miller RK, Brent RL, Koszalka TR (1978) Materno-fetal transport of creatine in the rat. Biol Neonate 33:43–54PubMedCrossRef Davis BM, Miller RK, Brent RL, Koszalka TR (1978) Materno-fetal transport of creatine in the rat. Biol Neonate 33:43–54PubMedCrossRef
go back to reference Dechent P, Pouwels PJ, Wilken B, Hanefeld F, Frahm J (1999) Increase of total creatine in human brain after oral supplementation of creatine-monohydrate. Am J Physiol 277:R698–R704PubMed Dechent P, Pouwels PJ, Wilken B, Hanefeld F, Frahm J (1999) Increase of total creatine in human brain after oral supplementation of creatine-monohydrate. Am J Physiol 277:R698–R704PubMed
go back to reference DeGrauw TJ, Salomons GS, Cecil KM et al. (2002) Congenital creatine transporter deficiency. Neuropediatrics 33:232–238PubMedCrossRef DeGrauw TJ, Salomons GS, Cecil KM et al. (2002) Congenital creatine transporter deficiency. Neuropediatrics 33:232–238PubMedCrossRef
go back to reference Dezortova M, Jiru F, Petrasek J, Malinova V, Zeman J, Jirsa M, Hajek M (2008) 1H MR spectroscopy as a diagnostic tool for cerebral creatine deficiency. Magn Reson Mater Phy 21:327–332CrossRef Dezortova M, Jiru F, Petrasek J, Malinova V, Zeman J, Jirsa M, Hajek M (2008) 1H MR spectroscopy as a diagnostic tool for cerebral creatine deficiency. Magn Reson Mater Phy 21:327–332CrossRef
go back to reference Dityatev A, Seidenbecher CI, Schachner M (2010) Compartmentalization from the outside: the extracellular matrix and functional microdomains in the brain. Trends Neurosci 33:503–512PubMedCrossRef Dityatev A, Seidenbecher CI, Schachner M (2010) Compartmentalization from the outside: the extracellular matrix and functional microdomains in the brain. Trends Neurosci 33:503–512PubMedCrossRef
go back to reference Engelhardt B, Sorokin L (2009) The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol 31:497–511PubMedCrossRef Engelhardt B, Sorokin L (2009) The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol 31:497–511PubMedCrossRef
go back to reference Ensenauer R, Thiel T, Schwab KO et al. (2004) Guanidinoacetate methyltransferase deficiency: differences of creatine uptake in human brain and muscle. Mol Genet Metab 82:208–213PubMedCrossRef Ensenauer R, Thiel T, Schwab KO et al. (2004) Guanidinoacetate methyltransferase deficiency: differences of creatine uptake in human brain and muscle. Mol Genet Metab 82:208–213PubMedCrossRef
go back to reference Fons C, Sempere A, Arias A et al. (2008) Arginine supplementation in four patients with X-linked creatine transporter defect. J Inherit Metab Dis 31:724–728PubMedCrossRef Fons C, Sempere A, Arias A et al. (2008) Arginine supplementation in four patients with X-linked creatine transporter defect. J Inherit Metab Dis 31:724–728PubMedCrossRef
go back to reference Fons C, Arias A, Sempere A et al. (2010) Response to creatine analogs in fibroblasts and patients with creatine transporter deficiency. Mol Genet Metab 99:296–299PubMedCrossRef Fons C, Arias A, Sempere A et al. (2010) Response to creatine analogs in fibroblasts and patients with creatine transporter deficiency. Mol Genet Metab 99:296–299PubMedCrossRef
go back to reference Ganesan V, Johnson A, Connelly A, Eckhardt S, Surtees RA (1997) Guanidinoacetate methyltransferase deficiency: new clinical features. Pediatr Neurol 17:155–157PubMedCrossRef Ganesan V, Johnson A, Connelly A, Eckhardt S, Surtees RA (1997) Guanidinoacetate methyltransferase deficiency: new clinical features. Pediatr Neurol 17:155–157PubMedCrossRef
go back to reference Giese MW, Lecher CS (2009) Non-enzymatic cyclization of creatine ethyl ester to creatinine. Biochem Biophys Res Commun 388:252–255PubMedCrossRef Giese MW, Lecher CS (2009) Non-enzymatic cyclization of creatine ethyl ester to creatinine. Biochem Biophys Res Commun 388:252–255PubMedCrossRef
go back to reference Happe HK, Murrin LC (1995) In situ hybridization analysis of CHOT1, a creatine transporter, in the rat central nervous system. J Comp Neurol 351:94–103PubMedCrossRef Happe HK, Murrin LC (1995) In situ hybridization analysis of CHOT1, a creatine transporter, in the rat central nervous system. J Comp Neurol 351:94–103PubMedCrossRef
go back to reference Hasselbalch SG, Knudsen GM, Jakobsen J, Hageman LP, Holm S, Paulson OB (1995) Blood-brain barrier permeability of glucose and ketone bodies during short-term starvation in humans. Am J Physiol 268:E1161–E1166PubMed Hasselbalch SG, Knudsen GM, Jakobsen J, Hageman LP, Holm S, Paulson OB (1995) Blood-brain barrier permeability of glucose and ketone bodies during short-term starvation in humans. Am J Physiol 268:E1161–E1166PubMed
go back to reference Hausmann ON, Fouad K, Wallimann T, Schwab ME (2002) Protective effects of oral creatine supplementation on spinal cord injury in rats. Spinal Cord 40:449–456PubMedCrossRef Hausmann ON, Fouad K, Wallimann T, Schwab ME (2002) Protective effects of oral creatine supplementation on spinal cord injury in rats. Spinal Cord 40:449–456PubMedCrossRef
go back to reference Hersch SM, Gevorkian S, Marder K et al. (2006) Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2'dG. Neurology 66:250–252PubMedCrossRef Hersch SM, Gevorkian S, Marder K et al. (2006) Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2'dG. Neurology 66:250–252PubMedCrossRef
go back to reference Ireland Z, Dickinson H, Snow R, Walker DW (2008) Maternal creatine: does it reach the fetus and improve survival after an acute hypoxic episode in the spiny mouse (Acomys cahirinus)? Am J Obstet Gynecol 198:431–436PubMedCrossRef Ireland Z, Dickinson H, Snow R, Walker DW (2008) Maternal creatine: does it reach the fetus and improve survival after an acute hypoxic episode in the spiny mouse (Acomys cahirinus)? Am J Obstet Gynecol 198:431–436PubMedCrossRef
go back to reference Ireland Z, Russell AP, Wallimann T, Walker DW, Snow R (2009) Developmental changes in the expression of creatine synthesizing enzymes and creatine transporter in a precocial rodent, the spiny mouse. BMC Dev Biol 9:39PubMedCrossRef Ireland Z, Russell AP, Wallimann T, Walker DW, Snow R (2009) Developmental changes in the expression of creatine synthesizing enzymes and creatine transporter in a precocial rodent, the spiny mouse. BMC Dev Biol 9:39PubMedCrossRef
go back to reference Ireland Z, Castillo-Melendez M, Dickinson H, Snow R, Walker DW (2011) A maternal diet supplemented with creatine from mid-pregnancy protects the newborn spiny mouse brain from birth hypoxia. Neuroscience Ireland Z, Castillo-Melendez M, Dickinson H, Snow R, Walker DW (2011) A maternal diet supplemented with creatine from mid-pregnancy protects the newborn spiny mouse brain from birth hypoxia. Neuroscience
go back to reference Item CB, Stöckler-Ipsiroglu S, Stromberger C et al. (2001) Arginine:glycine amidinotransferase deficiency: the third inborn error of creatine metabolism in humans. Am J Hum Genet 69:1127–1133PubMedCrossRef Item CB, Stöckler-Ipsiroglu S, Stromberger C et al. (2001) Arginine:glycine amidinotransferase deficiency: the third inborn error of creatine metabolism in humans. Am J Hum Genet 69:1127–1133PubMedCrossRef
go back to reference Kan HE, Meeuwissen E, van Asten JJ, Veltien A, Isbrandt D, Heerschap A (2007) Creatine uptake in brain and skeletal muscle of mice lacking guanidinoacetate methyltransferase assessed by magnetic resonance spectroscopy. J Appl Physiol 102:2121–2127PubMedCrossRef Kan HE, Meeuwissen E, van Asten JJ, Veltien A, Isbrandt D, Heerschap A (2007) Creatine uptake in brain and skeletal muscle of mice lacking guanidinoacetate methyltransferase assessed by magnetic resonance spectroscopy. J Appl Physiol 102:2121–2127PubMedCrossRef
go back to reference Lensman M, Korzhevskii DE, Mourovets VO, Kostkin VB, Izvarina N, Perasso L, Gandolfo C, Otellin VA, Polenov SA, Balestrino M (2006) Intracerebroventricular administration of creatine protects against damage by global cerebral ischemia in rat. Brain Res 1114:187–194PubMedCrossRef Lensman M, Korzhevskii DE, Mourovets VO, Kostkin VB, Izvarina N, Perasso L, Gandolfo C, Otellin VA, Polenov SA, Balestrino M (2006) Intracerebroventricular administration of creatine protects against damage by global cerebral ischemia in rat. Brain Res 1114:187–194PubMedCrossRef
go back to reference Lunardi G, Parodi A, Perasso L et al. (2006) The creatine transporter mediates the uptake of creatine by brain tissue, but not the uptake of two creatine-derived compounds. Neuroscience 142:991–997PubMedCrossRef Lunardi G, Parodi A, Perasso L et al. (2006) The creatine transporter mediates the uptake of creatine by brain tissue, but not the uptake of two creatine-derived compounds. Neuroscience 142:991–997PubMedCrossRef
go back to reference Mak CS, Waldvogel HJ, Dodd JR et al. (2009) Immunohistochemical localisation of the creatine transporter in the rat brain. Neuroscience 163:571–585PubMedCrossRef Mak CS, Waldvogel HJ, Dodd JR et al. (2009) Immunohistochemical localisation of the creatine transporter in the rat brain. Neuroscience 163:571–585PubMedCrossRef
go back to reference Mancini GM, Catsman-Berrevoets CE et al. (2005) Two novel mutations in SLC6A8 cause creatine transporter defect and distinctive X-linked mental retardation in two unrelated Dutch families. Am J Med Genet A 132:288–295 Mancini GM, Catsman-Berrevoets CE et al. (2005) Two novel mutations in SLC6A8 cause creatine transporter defect and distinctive X-linked mental retardation in two unrelated Dutch families. Am J Med Genet A 132:288–295
go back to reference Nakashima T, Tomi M, Katayama K et al. (2004) Blood-to-retina transport of creatine via creatine transporter (CRT) at the rat inner blood-retinal barrier. J Neurochem 89:1454–1461PubMedCrossRef Nakashima T, Tomi M, Katayama K et al. (2004) Blood-to-retina transport of creatine via creatine transporter (CRT) at the rat inner blood-retinal barrier. J Neurochem 89:1454–1461PubMedCrossRef
go back to reference Nakashima T, Tomi M, Tachikawa M, Watanabe M, Terasaki T, Hosoya K (2005) Evidence for creatine biosynthesis in Müller glia. GLIA 52:47–52PubMedCrossRef Nakashima T, Tomi M, Tachikawa M, Watanabe M, Terasaki T, Hosoya K (2005) Evidence for creatine biosynthesis in Müller glia. GLIA 52:47–52PubMedCrossRef
go back to reference Neuwelt EA, Bauer B, Fahlke C et al. (2011) Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 12:169–182PubMedCrossRef Neuwelt EA, Bauer B, Fahlke C et al. (2011) Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 12:169–182PubMedCrossRef
go back to reference Ohtsuki S, Tachikawa M, Takanaga H et al. (2002) The blood-brain barrier creatine transporter is a major pathway for supplying creatine to the brain. J Cereb Blood Flow Metab 22:1327–1335PubMedCrossRef Ohtsuki S, Tachikawa M, Takanaga H et al. (2002) The blood-brain barrier creatine transporter is a major pathway for supplying creatine to the brain. J Cereb Blood Flow Metab 22:1327–1335PubMedCrossRef
go back to reference Perasso L, Cupello A, Lunardi GL, Principato C, Gandolfo C, Balestrino M (2003) Kinetics of creatine in blood and brain after intraperitoneal injection in the rat. Brain Res 974:37–42PubMedCrossRef Perasso L, Cupello A, Lunardi GL, Principato C, Gandolfo C, Balestrino M (2003) Kinetics of creatine in blood and brain after intraperitoneal injection in the rat. Brain Res 974:37–42PubMedCrossRef
go back to reference Perasso L, Lunardi GL, Risso F et al. (2008) Protective effects of some creatine derivatives in brain tissue anoxia. Neurochem Res 33:765–775PubMedCrossRef Perasso L, Lunardi GL, Risso F et al. (2008) Protective effects of some creatine derivatives in brain tissue anoxia. Neurochem Res 33:765–775PubMedCrossRef
go back to reference Perasso L, Adriano E, Ruggeri P, Burov SV, Gandolfo C, Balestrino M (2009) In vivo neuroprotection by a creatine-derived compound: phosphocreatine-Mg-complex acetate. Brain Res 1285:158–163PubMedCrossRef Perasso L, Adriano E, Ruggeri P, Burov SV, Gandolfo C, Balestrino M (2009) In vivo neuroprotection by a creatine-derived compound: phosphocreatine-Mg-complex acetate. Brain Res 1285:158–163PubMedCrossRef
go back to reference Pisano JJ, Abraham D, Udenfriend S (1963) Biosynthesis and disposition of g-guanidinobutyric acid in mammalian tissues. Arch Biochem Biophys 100:323–329CrossRef Pisano JJ, Abraham D, Udenfriend S (1963) Biosynthesis and disposition of g-guanidinobutyric acid in mammalian tissues. Arch Biochem Biophys 100:323–329CrossRef
go back to reference Póo-Argüelles P, Arias A, Vilaseca MA et al. (2006) X-Linked creatine transporter deficiency in two patients with severe mental retardation and autism. J Inherit Metab Dis 29:220–223PubMedCrossRef Póo-Argüelles P, Arias A, Vilaseca MA et al. (2006) X-Linked creatine transporter deficiency in two patients with severe mental retardation and autism. J Inherit Metab Dis 29:220–223PubMedCrossRef
go back to reference Salomons GS, van Dooren SJ, Verhoeven NM et al. (2001) X-linked creatine-transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome. Am J Hum Genet 68:1497–1500PubMedCrossRef Salomons GS, van Dooren SJ, Verhoeven NM et al. (2001) X-linked creatine-transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome. Am J Hum Genet 68:1497–1500PubMedCrossRef
go back to reference Schloss P, Mayser W, Betz H (1994) The putative rat choline transporter CHOT1 transports creatine and is highly expressed in neural and muscle-rich tissues. Biochem Biophys Res Commun 198:637–645PubMedCrossRef Schloss P, Mayser W, Betz H (1994) The putative rat choline transporter CHOT1 transports creatine and is highly expressed in neural and muscle-rich tissues. Biochem Biophys Res Commun 198:637–645PubMedCrossRef
go back to reference Schmidt A, Marescau B, Boehm EA et al. (2004) Severely altered guanidino compound levels, disturbed body weight homeostasis and impaired fertility in a mouse model of guanidinoacetate N-methyltransferase (GAMT) deficiency. Hum Mol Genet 13:905–921PubMedCrossRef Schmidt A, Marescau B, Boehm EA et al. (2004) Severely altered guanidino compound levels, disturbed body weight homeostasis and impaired fertility in a mouse model of guanidinoacetate N-methyltransferase (GAMT) deficiency. Hum Mol Genet 13:905–921PubMedCrossRef
go back to reference Schulze A (2005) Strategies in the treatment of GAMT deficiency. In Jakobs C, Stöckler-Ipsiroglu S, eds; Verhoeven NM, Salomons GS, co-eds. Clinical and molecular aspects of defects in creatine and polyol metabolism. Heilbronn: SPS Verlagsgesellschaft, 19–33 Schulze A (2005) Strategies in the treatment of GAMT deficiency. In Jakobs C, Stöckler-Ipsiroglu S, eds; Verhoeven NM, Salomons GS, co-eds. Clinical and molecular aspects of defects in creatine and polyol metabolism. Heilbronn: SPS Verlagsgesellschaft, 19–33
go back to reference Schulze A, Battini R (2007) Pre-symptomatic treatment of creatine biosynthesis defects. Subcell Biochem 46:167–181PubMedCrossRef Schulze A, Battini R (2007) Pre-symptomatic treatment of creatine biosynthesis defects. Subcell Biochem 46:167–181PubMedCrossRef
go back to reference Schulze A, Hess T, Wevers R et al. (1997) Creatine deficiency syndrome caused by guanidinoacetate methyltransferase deficiency: diagnostic tools for a new inborn error of metabolism. J Pediatr 131:626–631PubMedCrossRef Schulze A, Hess T, Wevers R et al. (1997) Creatine deficiency syndrome caused by guanidinoacetate methyltransferase deficiency: diagnostic tools for a new inborn error of metabolism. J Pediatr 131:626–631PubMedCrossRef
go back to reference Schulze A, Mayatepek E, Bachert P, Marescau B, De Deyn PP, Rating D (1998) Therapeutic trial of arginine restriction in creatine deficiency syndrome. Eur J Pediatr 157:606–607PubMedCrossRef Schulze A, Mayatepek E, Bachert P, Marescau B, De Deyn PP, Rating D (1998) Therapeutic trial of arginine restriction in creatine deficiency syndrome. Eur J Pediatr 157:606–607PubMedCrossRef
go back to reference Schulze A, Bachert P, Schlemmer H et al. (2003) Lack of creatine in muscle and brain in an adult with GAMT deficiency. Ann Neurol 53:248–251PubMedCrossRef Schulze A, Bachert P, Schlemmer H et al. (2003) Lack of creatine in muscle and brain in an adult with GAMT deficiency. Ann Neurol 53:248–251PubMedCrossRef
go back to reference Schulze A, Hoffmann GF, Bachert P et al. (2006) Presymptomatic treatment of neonatal guanidinoacetate methyltransferase deficiency. Neurology 67:719–721PubMedCrossRef Schulze A, Hoffmann GF, Bachert P et al. (2006) Presymptomatic treatment of neonatal guanidinoacetate methyltransferase deficiency. Neurology 67:719–721PubMedCrossRef
go back to reference Sijens PE, Verbruggen KT, Oudkerk M, van Spronsen FJ, Soorani-Lunsing RJ (2005) 1H MR spectroscopy of the brain in Cr transporter defect. Mol Genet Metab 86:421–422PubMedCrossRef Sijens PE, Verbruggen KT, Oudkerk M, van Spronsen FJ, Soorani-Lunsing RJ (2005) 1H MR spectroscopy of the brain in Cr transporter defect. Mol Genet Metab 86:421–422PubMedCrossRef
go back to reference Spector R (2010) Nature and consequences of mammalian brain and CSF efflux transporters: four decades of progress. J Neurochem 112:13–23PubMedCrossRef Spector R (2010) Nature and consequences of mammalian brain and CSF efflux transporters: four decades of progress. J Neurochem 112:13–23PubMedCrossRef
go back to reference Stöckler S, Holzbach U, Hanefeld F et al. (1994) Creatine deficiency in the brain: a new, treatable inborn error of metabolism. Pediatr Res 36:409–413PubMedCrossRef Stöckler S, Holzbach U, Hanefeld F et al. (1994) Creatine deficiency in the brain: a new, treatable inborn error of metabolism. Pediatr Res 36:409–413PubMedCrossRef
go back to reference Stöckler S, Hanefeld F, Frahm J (1996) Creatine replacement therapy in guanidinoacetate methyltransferase deficiency, a novel inborn error of metabolism. Lancet 348:789–790PubMedCrossRef Stöckler S, Hanefeld F, Frahm J (1996) Creatine replacement therapy in guanidinoacetate methyltransferase deficiency, a novel inborn error of metabolism. Lancet 348:789–790PubMedCrossRef
go back to reference Stöckler S, Schutz PW, Salomons GS (2007) Cerebral creatine deficiency syndromes: Clinical aspects, treatment and pathophysiology. Subcell Biochem 46:149–166PubMedCrossRef Stöckler S, Schutz PW, Salomons GS (2007) Cerebral creatine deficiency syndromes: Clinical aspects, treatment and pathophysiology. Subcell Biochem 46:149–166PubMedCrossRef
go back to reference Sullivan PG, Geiger JD, Mattson MP, Scheff SW (2000) Dietary supplement creatine protects against traumatic brain injury. Ann Neurol 48:723–729PubMedCrossRef Sullivan PG, Geiger JD, Mattson MP, Scheff SW (2000) Dietary supplement creatine protects against traumatic brain injury. Ann Neurol 48:723–729PubMedCrossRef
go back to reference Tachikawa M, Hosoya K (2011) Transport characteristics of guanidino compounds at the blood-brain barrier and blood-cerebrospinal fluid barrier: relevance to neural disorders. Fluids Barriers CNS 8:13PubMedCrossRef Tachikawa M, Hosoya K (2011) Transport characteristics of guanidino compounds at the blood-brain barrier and blood-cerebrospinal fluid barrier: relevance to neural disorders. Fluids Barriers CNS 8:13PubMedCrossRef
go back to reference Tachikawa M, Fukaya M, Terasaki T, Ohtsuki S, Watanabe M (2004) Distinct cellular expressions of creatine synthetic enzyme GAMT and creatine kinases uCK-Mi and CK-B suggest a novel neuron-glial relationship for brain energy homeostasis. Eur J Neurosci 20:144–160PubMedCrossRef Tachikawa M, Fukaya M, Terasaki T, Ohtsuki S, Watanabe M (2004) Distinct cellular expressions of creatine synthetic enzyme GAMT and creatine kinases uCK-Mi and CK-B suggest a novel neuron-glial relationship for brain energy homeostasis. Eur J Neurosci 20:144–160PubMedCrossRef
go back to reference Tachikawa M, Fujinawa J, Takahashi M et al. (2008) Expression and possible role of creatine transporter in the brain and at the blood-cerebrospinal fluid barrier as a transporting protein of guanidinoacetate, an endogenous convulsant. J Neurochem 107:768–778PubMedCrossRef Tachikawa M, Fujinawa J, Takahashi M et al. (2008) Expression and possible role of creatine transporter in the brain and at the blood-cerebrospinal fluid barrier as a transporting protein of guanidinoacetate, an endogenous convulsant. J Neurochem 107:768–778PubMedCrossRef
go back to reference Tachikawa M, Kasai Y, Yokoyama R et al. (2009) The blood-brain barrier transport and cerebral distribution of guanidinoacetate in rats: involvement of creatine and taurine transporters. J Neurochem Tachikawa M, Kasai Y, Yokoyama R et al. (2009) The blood-brain barrier transport and cerebral distribution of guanidinoacetate in rats: involvement of creatine and taurine transporters. J Neurochem
go back to reference Valayannopoulos V, Boddaert N, Chabli A et al. (2011) Treatment by oral creatine, L-arginine and L-glycine in six severely affected patients with creatine transporter defect. J Inherit Metab Dis Valayannopoulos V, Boddaert N, Chabli A et al. (2011) Treatment by oral creatine, L-arginine and L-glycine in six severely affected patients with creatine transporter defect. J Inherit Metab Dis
go back to reference van de Kamp JM, Pouwels PJ, Aarsen FK et al. (2011a) Long-term follow-up and treatment in nine boys with X-linked creatine transporter defect. J Inherit Metab Dis Epub ahead of print van de Kamp JM, Pouwels PJ, Aarsen FK et al. (2011a) Long-term follow-up and treatment in nine boys with X-linked creatine transporter defect. J Inherit Metab Dis Epub ahead of print
go back to reference van de Kamp JM, Mancini GMS, Pouwels PJW, Betsalel OT, van Dooren SJM, de Koning I, Steenweg ME, Jakobs C, van der Knaap MS, Salomons GS (2011b) Clinical features and X-inactivation in females heterozygous for creatine transporter defect. Clin Genet 79:199–299CrossRef van de Kamp JM, Mancini GMS, Pouwels PJW, Betsalel OT, van Dooren SJM, de Koning I, Steenweg ME, Jakobs C, van der Knaap MS, Salomons GS (2011b) Clinical features and X-inactivation in females heterozygous for creatine transporter defect. Clin Genet 79:199–299CrossRef
go back to reference Wang L, Zhang Y, Shao M, Zhang H (2007) Spatiotemporal expression of the creatine metabolism related genes agat, gamt and ct1 during zebrafish embryogenesis. Int J Dev Biol 51:247–253PubMedCrossRef Wang L, Zhang Y, Shao M, Zhang H (2007) Spatiotemporal expression of the creatine metabolism related genes agat, gamt and ct1 during zebrafish embryogenesis. Int J Dev Biol 51:247–253PubMedCrossRef
go back to reference Wilcken B, Fagan E, Sim K, Carpenter KH, Salomons GS (2008) Creatine transporter defect: results of 6 months' treatment. J Inher Metab Dis 31(Suppl):70 Wilcken B, Fagan E, Sim K, Carpenter KH, Salomons GS (2008) Creatine transporter defect: results of 6 months' treatment. J Inher Metab Dis 31(Suppl):70
go back to reference Wyss M, Kaddurah-Daouk R (2000) Creatine and creatinine metabolism. Physiol Rev 80:1107–1213PubMed Wyss M, Kaddurah-Daouk R (2000) Creatine and creatinine metabolism. Physiol Rev 80:1107–1213PubMed
go back to reference Zhang Y, Barres BA (2010) Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr Opin Neurobiol 20:588–594PubMedCrossRef Zhang Y, Barres BA (2010) Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr Opin Neurobiol 20:588–594PubMedCrossRef
Metadata
Title
Creatine and guanidinoacetate transport at blood-brain and blood-cerebrospinal fluid barriers
Author
Olivier Braissant
Publication date
01-07-2012
Publisher
Springer Netherlands
Published in
Journal of Inherited Metabolic Disease / Issue 4/2012
Print ISSN: 0141-8955
Electronic ISSN: 1573-2665
DOI
https://doi.org/10.1007/s10545-011-9433-2

Other articles of this Issue 4/2012

Journal of Inherited Metabolic Disease 4/2012 Go to the issue

Editorial

SSIEM 2011

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
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 discusses last year's major advances in heart failure and cardiomyopathies.