Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neurochem ; 168(2): 69-82, 2024 02.
Article in English | MEDLINE | ID: mdl-38178803

ABSTRACT

N-acetylaspartate (NAA) is an abundant central nervous system amino acid derivative that is tightly coupled to mitochondria and energy metabolism in neurons. A reduced NAA signature is a prominent early pathological biomarker in multiple neurodegenerative diseases and becomes progressively more pronounced as disease advances. Because NAA synthesis requires aspartate drawn directly from mitochondria, we argued that this process is in direct competition with oxidative phosphorylation for substrate and that sustained high levels of NAA synthesis would be incompatible with pathological energy crisis. We show here that over-expression of the rate-limiting NAA synthetic enzyme in the hippocampus of the 5x familial Alzheimer's disease (5xFAD) mouse results in an exaggerated pathological ATP deficit and accelerated cognitive decline. Over-expression of NAA synthase did not increase amyloid burden or result in cell loss but did significantly deplete mitochondrial aspartate and impair the ability of mitochondria to oxidize glutamate for adenosine triphosphate (ATP) synthesis. These results define NAA as a sink for energetic substrate and suggest initial pathological reductions in NAA are part of a response to energetic crisis designed to preserve substrate bioavailability for mitochondrial ATP synthesis.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Aspartic Acid/metabolism , Adenosine Triphosphate/metabolism
2.
Mol Ther Methods Clin Dev ; 20: 520-534, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33614826

ABSTRACT

Recent advances in adeno-associated viral (AAV) capsid variants with novel oligotropism require validation in models of disease in order to be viable candidates for white matter disease gene therapy. We present here an assessment of the biodistribution, tropism, and efficacy of a novel AAV capsid variant (AAV/ Olig001) in a model of Canavan disease. We first define a combination of dose and route of administration of an AAV/Olig001-GFP reporter conducive to widespread CNS oligodendrocyte transduction in acutely symptomatic animals that model the Canavan brain at time of diagnosis. Administration of AAV/Olig001-GFP resulted in >70% oligotropism in all regions of interest except the cerebellum without the need for lineage-specific expression elements. Intracerebroventricular infusion into the cerebrospinal fluid (CSF) was identified as the most appropriate route of administration and employed for delivery of an AAV/Olig001 vector to reconstitute oligodendroglial aspartoacylase (ASPA) in adult Canavan mice, which resulted in a dose-dependent rescue of ASPA activity, motor function, and a near-total reduction in vacuolation. A head-to-head efficacy comparison with astrogliotropic AAV9 highlighted a significant advantage conferred by oligotropic AAV/Olig001 that was independent of overall transduction efficiency. These results support the continued development of AAV/Olig001 for advancement to clinical application to white matter disease.

3.
Neurobiol Dis ; 96: 323-334, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27717881

ABSTRACT

Breakdown of neuro-glial N-acetyl-aspartate (NAA) metabolism results in the failure of developmental myelination, manifest in the congenital pediatric leukodystrophy Canavan disease caused by mutations to the sole NAA catabolizing enzyme aspartoacylase. Canavan disease is a major point of focus for efforts to define NAA function, with available evidence suggesting NAA serves as an acetyl donor for fatty acid synthesis during myelination. Elevated NAA is a diagnostic hallmark of Canavan disease, which contrasts with a broad spectrum of alternative neurodegenerative contexts in which levels of NAA are inversely proportional to pathological progression. Recently generated data in the nur7 mouse model of Canavan disease suggests loss of aspartoacylase function results in compromised energetic integrity prior to oligodendrocyte death, abnormalities in myelin content, spongiform degeneration, and motor deficit. The present study utilized a next-generation "oligotropic" adeno-associated virus vector (AAV-Olig001) to quantitatively assess the impact of aspartoacylase reconstitution on developmental myelination. AAV-Olig001-aspartoacylase promoted normalization of NAA, increased bioavailable acetyl-CoA, and restored energetic balance within a window of postnatal development preceding gross histopathology and deteriorating motor function. Long-term effects included increased oligodendrocyte numbers, a global increase in myelination, reversal of vacuolation, and rescue of motor function. Effects on brain energy observed following AAV-Olig001-aspartoacylase gene therapy are shown to be consistent with a metabolic profile observed in mild cases of Canavan disease, implicating NAA in the maintenance of energetic integrity during myelination via oligodendroglial aspartoacylase.


Subject(s)
Amidohydrolases/metabolism , Aspartic Acid/analogs & derivatives , Brain/enzymology , Canavan Disease/pathology , Myelin Sheath/physiology , Oligodendroglia/enzymology , Amidohydrolases/genetics , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Autophagy-Related Proteins , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/pathology , Canavan Disease/complications , Canavan Disease/diagnostic imaging , Canavan Disease/genetics , Child , Child, Preschool , Dependovirus/genetics , Disease Progression , Energy Metabolism/genetics , Female , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Infant , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Movement Disorders/etiology , Myelin Basic Protein/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics
4.
Mol Cell Neurosci ; 65: 143-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25766789

ABSTRACT

N-acetylaspartate (NAA) provides a non-invasive clinical index of neuronal metabolic integrity across the entire neurodegenerative spectrum. While NAA function is not comprehensively defined, reductions in the brain are associated with compromised mitochondrial metabolism and are tightly linked to ATP. We have undertaken an analysis of abnormalities in NAA during early stage pathology in the 5xFAD mouse model of familial Alzheimer's disease and show here that dysregulated expression of the gene encoding for the rate-limiting NAA synthetic enzyme (Nat8L) is associated with deficits in mitochondrial oxidative phosphorylation in this model system. Downreguation of Nat8L is particularly pronounced in the 5xFAD hippocampus, and is preceded by a significant upregulation of oligodendrocytic aspartoacylase (aspa), which encodes for the sole known NAA-catabolizing enzyme in the brain. Reductions in 5xFAD NAA and Nat8L cannot be accounted for by discrepancies in either neuron content or activity of the substrate-providing malate-aspartate shuttle, thereby implicating transcriptional regulation in a coordinated response to pathological energetic crisis. A central role for ASPA in this response is supported by a parallel developmental analysis showing highly significant increases in Nat8L expression in an ASPA-null mouse model during a period of early postnatal development normally punctuated by the transcriptional upregulation of aspa. These results provide preliminary evidence of a signaling mechanism in Alzheimer's disease that involves cross talk between neurons and oligodendrocytes, and suggest that ASPA acts to negatively regulate Nat8L expression. This mechanism is proposed to be a fundamental means by which the brain conserves available substrate during energy crises.


Subject(s)
Acetyltransferases/metabolism , Alzheimer Disease/metabolism , Neuroglia/metabolism , Neurons/metabolism , Oxidative Phosphorylation , Transcription, Genetic , Acetyltransferases/genetics , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Down-Regulation , Hippocampus/cytology , Hippocampus/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
J Exp Med ; 211(8): 1525-31, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25024137

ABSTRACT

The transcription factor Gata6 regulates proliferation and differentiation of epithelial and endocrine cells and cancers. Among hematopoietic cells, Gata6 is expressed selectively in resident peritoneal macrophages. We thus examined whether the loss of Gata6 in the macrophage compartment affected peritoneal macrophages, using Lyz2-Cre x Gata6(flox/flox) mice to tackle this issue. In Lyz2-Cre x Gata6(flox/flox) mice, the resident peritoneal macrophage compartment, but not macrophages in other organs, was contracted, with only a third the normal number of macrophages remaining. Heightened rates of death explained the marked decrease in peritoneal macrophage observed. The metabolism of the remaining macrophages was skewed to favor oxidative phosphorylation and alternative activation markers were spontaneously and selectively induced in Gata6-deficient macrophages. Gene expression profiling revealed perturbed metabolic regulators, including aspartoacylase (Aspa), which facilitates generation of acetyl CoA. Mutant mice lacking functional Aspa phenocopied the higher propensity to death and led to a contraction of resident peritoneal macrophages. Thus, Gata6 regulates differentiation, metabolism, and survival of resident peritoneal macrophages.


Subject(s)
Amidohydrolases/metabolism , GATA6 Transcription Factor/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/enzymology , Animals , Apoptosis/genetics , Cell Count , Cell Survival , GATA6 Transcription Factor/deficiency , Gene Expression Regulation , Macrophage Activation/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data
6.
J Inherit Metab Dis ; 37(3): 369-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24288037

ABSTRACT

The inherited pediatric leukodystrophy Canavan disease is characterized by dysmyelination and severe spongiform degeneration, and is currently refractory to treatment. A definitive understanding of core disease mechanisms is lacking, but pathology is believed to result at least in part compromised fatty acid synthesis during myelination. Recent evidence generated in an animal model suggests that the breakdown of N-acetylaspartate metabolism in CD results in a heightened coupling of fatty acid synthesis to oligodendrocyte oxidative metabolism during the early stages of myelination, thereby causing acute oxidative stress. We present here the results of a dietary intervention designed to support oxidative integrity during developmental myelination in the nur7 mouse model of Canavan disease. Provision of the odd carbon triglyceride triheptanoin to neonatal nur7 mice reduced oxidative stress, promoted long-term oligodendrocyte survival, and increased myelin in the brain. Improvements in oligodendrocyte survival and myelination were associated with a highly significant reduction in spongiform degeneration and improved motor function in triheptanoin treated mice. Initiation of triheptanoin treatment in older animals resulted in markedly more modest effects on these same pathological indices, indicating a window of therapeutic intervention that corresponds with developmental myelination. These results support the targeting of oxidative integrity at early stages of Canavan disease, and provide a foundation for the clinical development of a non-invasive dietary triheptanoin treatment regimen.


Subject(s)
Amidohydrolases/genetics , Canavan Disease/therapy , Myelin Sheath/physiology , Oligodendroglia/pathology , Triglycerides/administration & dosage , Adenosine Triphosphate/metabolism , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Canavan Disease/pathology , Canavan Disease/physiopathology , Disease Models, Animal , Mice
7.
Sci Transl Med ; 4(165): 165ra163, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23253610

ABSTRACT

Canavan disease is a hereditary leukodystrophy caused by mutations in the aspartoacylase gene (ASPA), leading to loss of enzyme activity and increased concentrations of the substrate N-acetyl-aspartate (NAA) in the brain. Accumulation of NAA results in spongiform degeneration of white matter and severe impairment of psychomotor development. The goal of this prospective cohort study was to assess long-term safety and preliminary efficacy measures after gene therapy with an adeno-associated viral vector carrying the ASPA gene (AAV2-ASPA). Using noninvasive magnetic resonance imaging and standardized clinical rating scales, we observed Canavan disease in 28 patients, with a subset of 13 patients being treated with AAV2-ASPA. Each patient received 9 × 10(11) vector genomes via intraparenchymal delivery at six brain infusion sites. Safety data collected over a minimum 5-year follow-up period showed a lack of long-term adverse events related to the AAV2 vector. Posttreatment effects were analyzed using a generalized linear mixed model, which showed changes in predefined surrogate markers of disease progression and clinical assessment subscores. AAV2-ASPA gene therapy resulted in a decrease in elevated NAA in the brain and slowed progression of brain atrophy, with some improvement in seizure frequency and with stabilization of overall clinical status.


Subject(s)
Canavan Disease/therapy , Genetic Therapy , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/metabolism , Canavan Disease/metabolism , Child , Child, Preschool , Humans , Infant , Prospective Studies
8.
J Cereb Blood Flow Metab ; 32(9): 1725-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22617649

ABSTRACT

The inherited leukodystrophy Canavan disease arises due to a loss of the ability to catabolize N-acetylaspartic acid (NAA) in the brain and constitutes a major point of focus for efforts to define NAA function. Accumulation of noncatabolized NAA is diagnostic for Canavan disease, but contrasts with the abnormally low NAA associated with compromised neuronal integrity in a broad spectrum of other clinical conditions. Experimental evidence for NAA function supports a role in white matter lipid synthesis, but does not explain how both elevated and lowered NAA can be associated with pathology in the brain. We have undertaken a systematic analysis of postnatal development in a mouse model of Canavan disease that delineates development and pathology by identifying markers of oxidative stress preceding oligodendrocyte loss and dysmyelination. These data suggest a role for NAA in the maintenance of metabolic integrity in oligodendrocytes that may be of relevance to the strong association between NAA and neuronal viability. N-acetylaspartic acid is proposed here to support lipid synthesis and energy metabolism via the provision of substrate for both cellular processes during early postnatal development.


Subject(s)
Amidohydrolases/metabolism , Energy Metabolism/physiology , Myelin Sheath/physiology , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Aspartic Acid/pharmacology , Biomarkers , Canavan Disease/enzymology , Canavan Disease/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Dependovirus/genetics , Glucose/deficiency , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mutation/physiology , Oligodendroglia/metabolism , Oxidation-Reduction , Oxidative Stress/physiology
9.
Glia ; 59(10): 1435-46, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21608034

ABSTRACT

Aspartoacylase (ASPA) is an enzyme that functions to catabolize the neuronal amino acid derivative N-acetyl-L-aspartic acid (NAA). Loss of this function results in the failure of developmental myelination. NAA synthesis and catabolism are tightly compartmentalized within neurons and oligodendrocytes, respectively, and there is evidence to suggest the developmental regulation of ASPA activity is transcriptional. NAA has no known direct physiological mode of action, and the identification of a transcriptional regulator of aspa would provide an opportunity to link NAA to relatively more characterized physiological contexts with a view to definitive functional classification. Using transcriptional and immunohistochemical endpoints, we define a window of postnatal development punctuated by increases in aspa within a pre-existing population of oligodendrocytes in the rat brain. Ontological mining of expression data generated in aspa-null rats during this developmental window identifies both neuronal and oligodendroglial transcriptional abnormalities that suggest an association between glutamatergic synaptic activity and ASPA. Glutamate, but not NAA, is shown to activate endogenous aspa expression in vitro, and endogenous aspa expression is upregulated in the brains of animals over expressing vesicular glutamate transporter type-I (vglut1). These results define a discrete period of postnatal development within which glutamate provides a means by which the tightly compartmentalized NAA metabolic cycle can function in sync with normal development and may be a factor in pathological models of dysregulated NAA metabolism.


Subject(s)
Amidohydrolases/metabolism , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Glutamic Acid/metabolism , Oligodendroglia/metabolism , Age Factors , Amidohydrolases/pharmacology , Animals , Animals, Newborn , Brain/cytology , Cell Line, Transformed , Cells, Cultured , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Transfer Techniques , Genetic Vectors , Glutamic Acid/pharmacology , Green Fluorescent Proteins/genetics , Humans , Mice , O Antigens/metabolism , Rats , Transfection/methods , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism
10.
Neurochem Res ; 34(1): 138-48, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18478328

ABSTRACT

The high concentration of N-acetylaspartate (NAA) in neurons of the central nervous system and its growing clinical use as an indicator of neuronal viability has intensified interest in the biological function of this amino acid derivative. The biomedical relevance of such inquiries is highlighted by the myelin-associated pathology of Canavan disease, an inherited childhood disorder resulting from mutation of aspartoacylase (ASPA), the NAA-hydrolyzing enzyme. This enzyme is known to be localized in oligodendrocytes with bimodal distribution in cytosol and the myelin sheath, and to produce acetyl groups utilized in myelin lipid synthesis. Loss of this acetyl source in Canavan disease and rodent models such as the tremor rat are thought to account for the observed myelin deficit. This study was undertaken to further define and quantify the specific lipid abnormalities that occur as a result of ASPA deficit in the tremor rat. Employing mass spectrometry together with high performance thin-layer chromatography, we found that myelin from 28-day-old animals showed major reduction in cerebrosides (CB) and sulfatides (Sulf) with unsubstituted fatty acids, and equal if not greater changes in myelin from 7-month-old tremors. Cerebrosides with 2-hydroxyfatty acids showed little if any change at either age; Sulf with 2-hydroxyfatty acids showed no significant change at 28 days, but surprisingly a major increase at 7 months. Two species of phosphatidylcholine, 32:0 and 34:1, also showed significant increase, but only at 28 days. One form of phosphatidylethanolamine, PE36:1, was reduced a modest amount at both ages, whereas the plasmalogen form did not change. The dysmyelination that results from inactivation of ASPA is thus characterized by selective decreases as well as some increases in specific lipids.


Subject(s)
Canavan Disease/metabolism , Lipids/chemistry , Myelin Sheath/metabolism , Animals , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Disease Models, Animal , Myelin Sheath/chemistry , Rats , Rats, Inbred WKY , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Exp Neurol ; 205(1): 177-89, 2007 May.
Article in English | MEDLINE | ID: mdl-17382931

ABSTRACT

We have investigated the gliogenic potential of cells isolated from a recently described GFP-transgenic rat [Inoue, H., Ohsawa, I., Murakami, T., Kimura, A., Hakamata, Y., Sato, Y., Kaneko, T., Takahashi, M., Okada, T., Ozawa, K., Francis, J., Leone, P., Kobayashi, E., 2005. Development of new inbred transgenic strains of rats with LacZ or GFP. Biochem Biophys Res Commun 329 288-295.] for application to oligodendrocyte replacement in models of white matter insult and disease. These transgenic rats present native GFP fluorescence in oligodendrocytes of the CNS, with no detectable fluorescence in astrocytes or mature neurons. By targeting a highly gliogenic period of postnatal development, we show that sphere-forming cultures of proliferating cells generated from the GFP-transgenic brain give rise to significant numbers of differentiated oligodendrocytes in vitro. Postnatal source tissue was significantly more gliogenic than embryonic source tissue, with greater than 50% of postnatally derived cells differentiating into GFP-positive oligodendrocytes. Differentiated oligodendrocytes exhibited an increased intensity of GFP fluorescence concomitant with the acquisition of mature oligodendrocyte-specific markers in both isolated cultures and in co-culture with primary neurons. Transplantation of postnatally derived GFP-positive sphere-forming cells into ethidium bromide lesioned Kyoto-Wistar rats resulted in the engraftment and survival of GFP-positive oligodendrocytes for at least 6 weeks in the host white matter and cerebral cortex. Our results show that sphere-forming cultures of cells isolated from the early postnatal GFP-Lewis rat brain are a useful tool for oligodendrocyte replacement studies.


Subject(s)
Animals, Genetically Modified , Cell Proliferation , Green Fluorescent Proteins/genetics , Oligodendroglia/cytology , Oligodendroglia/transplantation , Rats, Inbred Lew , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/drug effects , Brain/pathology , Cell Differentiation , Cell Survival , Cells, Cultured , Cellular Senescence , Cerebral Cortex/pathology , Coculture Techniques , Embryo, Mammalian/cytology , Ethidium/pharmacology , Fluorescence , Green Fluorescent Proteins/metabolism , Male , Oligodendroglia/pathology , Oligodendroglia/physiology , Rats , Rats, Inbred WKY , Spheroids, Cellular/cytology , Time Factors
12.
J Neurosci Res ; 84(1): 151-69, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16634055

ABSTRACT

Neuronal growth factors are thought to exert a significant degree of control over postnatal oligodendrogenesis, but mechanisms by which these factors coordinateoligodendrocyte development with the maturation of neural networks are poorly characterized. We present here a developmental analysis of aspartoacylase (Aspa)-null tremor rats and show a potential role for this hydrolytic enzyme in the regulation of a postnatal neurotrophic stimulus that impacts on early stages of oligodendrocyte differentiation. Abnormally high levels of brain-derived neurotrophic factor (BDNF) expression in the Aspa-null Tremor brain are associated with dysregulated oligodendrogenesis at a stage in development normally characterized by high levels of Aspa expression. BDNF promotes the survival of proliferating cells during the early stages of oligodendrocyte maturation in vitro, but seems to compromise the ability of these cells to populate the cortex in vivo. Aspartoacylase activity in oligodendrocytes is shown to provide for the negative regulation of BDNF in neurons, thereby determining the availability of a developmental stimulus via a mechanism that links oligodendroglial differentiation with neuronal maturation.


Subject(s)
Amidohydrolases/physiology , Brain-Derived Neurotrophic Factor/metabolism , Brain/cytology , Gene Expression/physiology , Oligodendroglia/physiology , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Age Factors , Animals , Animals, Genetically Modified , Animals, Newborn , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Fluorescent Antibody Technique/methods , In Situ Hybridization/methods , Myelin Basic Protein/metabolism , Neurons/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Tremor/genetics , Tremor/pathology , Tremor/physiopathology
13.
Clin Biochem ; 38(11): 997-1008, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16139832

ABSTRACT

OBJECTIVES: To set up a novel simple, sensitive, and reliable ion-pairing HPLC method for the synchronous separation of several purines, pyrimidines, N-acetylated amino acids, and dicarboxylic acids for the chemical diagnosis and screening of inborn errors of metabolism (IEM). DESIGN AND METHODS: The separation was set up using a Hypersil C-18, 5-microm particle size, 250 x 4.6 mm column, and a step gradient using two buffers and tetrabutylammonium hydroxide as the pairing reagent. A highly sensitive diode array UV detector was set up at a wavelength between 200 and 300 nm that revealed purines and pyrimidines at 260 nm and other compounds at 206 nm. RESULTS: Compounds were determined in the plasma of 15 healthy adults, in the urine of 50 healthy subjects (1-3 years, 4-6 years, 8-10 years, 12-18 years, 25-35 years), and in 10 non-pathological amniotic fluid samples. To assess the validity of the chemical diagnosis of IEM, plasma and urine samples were analyzed in patients affected by Canavan disease (n = 10; mean age 4.6 +/- 2.3). Low plasma levels of N-acetylaspartate (16.96 +/- 19.57 micromol/L plasma; not detectable in healthy adults) and dramatically high urinary N-acetylaspartate concentrations (1872.03 +/- 631.86 micromol/mmol creatinine; 450 times higher than that which was observed in age-matched controls) were recorded. Neither N-acetylglutamate nor N-acetylaspartylglutamate could be detected in the plasma or urine of controls or patients with Canavan disease. CONCLUSIONS: The results demonstrate the suitability of the present ion-pairing HPLC separation with UV detection of cytosine, cytidine, creatinine, uracil, uridine, beta-pseudouridine, adenine, 3-methyladenine, hypoxanthine, xanthine, xanthosine, inosine, guanosine, ascorbic acid, thymine, thymidine, uric acid, 1-methyluric acid, orotic acid, N-acetylaspartate, N-acetylglutamate, N-acetylaspartylglutamate, malonic acid, methylmalonic acid, GSH, and GSSG as a reliable method for the prenatal and neonatal chemical diagnosis and screening of IEM using biological fluids.


Subject(s)
Amino Acids/isolation & purification , Chromatography, High Pressure Liquid/methods , Dicarboxylic Acids/isolation & purification , Metabolism, Inborn Errors/diagnosis , Purines/isolation & purification , Pyrimidines/isolation & purification , Adolescent , Adult , Amniotic Fluid/chemistry , Aspartic Acid/analogs & derivatives , Aspartic Acid/blood , Aspartic Acid/urine , Canavan Disease/diagnosis , Child , Child, Preschool , Humans , Infant , Mass Screening/methods , Middle Aged , Prenatal Diagnosis/methods , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet
14.
Brain Res Mol Brain Res ; 124(2): 199-203, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15135228

ABSTRACT

ATF-3 is a member of the ATF superfamily of transcription factors and is strongly associated with episodes of cellular stress. We demonstrate an association between increases in ATF-3 protein and resistance to exitotoxic cell death in vivo. Intra-hippocampal injection of kainic acid elicited a robust increase in endogenous ATF-3 within kainate-resistant cells of the dentate gyrus, while overexpression of exogenous ATF-3 was found to protect vulnerable CA3 neurons from the same insult. These results suggest a positive contribution to neuronal survival in the context of stress-induced death, and support an anti-apoptotic role for ATF-3 in the brain.


Subject(s)
Apoptosis/genetics , Hippocampus/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress/genetics , Transcription Factors/biosynthesis , Activating Transcription Factor 3 , Animals , Apoptosis/drug effects , Cytoprotection/genetics , Hippocampus/drug effects , Hippocampus/physiopathology , Immunity, Innate/genetics , Kainic Acid , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Transcription Factors/genetics , Transcription Factors/pharmacology
15.
Nat Med ; 9(9): 1173-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12925848

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a gut peptide that, together with its receptor, GLP-1R, is expressed in the brain. Here we show that intracerebroventricular (i.c.v.) GLP-1 and [Ser(2)]exendin(1-9) (HSEGTFTSD; homologous to a conserved domain in the glucagon/GLP-1 family) enhance associative and spatial learning through GLP-1R. [Ser(2)]exendin(1-9), but not GLP-1, is also active when administered peripherally. GLP-1R-deficient mice have a phenotype characterized by a learning deficit that is restored after hippocampal Glp1r gene transfer. In addition, rats overexpressing GLP-1R in the hippocampus show improved learning and memory. GLP-1R-deficient mice also have enhanced seizure severity and neuronal injury after kainate administration, with an intermediate phenotype in heterozygotes and phenotypic correction after Glp1r gene transfer in hippocampal somatic cells. Systemic administration of [Ser(2)]exendin(1-9) in wild-type animals prevents kainate-induced apoptosis of hippocampal neurons. Brain GLP-1R represents a promising new target for both cognitive-enhancing and neuroprotective agents.


Subject(s)
Glucagon/pharmacology , Hippocampus/physiology , Learning/physiology , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Administration, Intranasal , Amino Acid Sequence , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Cognition/drug effects , Gene Expression , Gene Transfer Techniques , Glucagon/administration & dosage , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Hippocampus/cytology , Hippocampus/drug effects , Kainic Acid/adverse effects , Learning/drug effects , Male , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Peptide Fragments/administration & dosage , Protein Precursors/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/drug effects , Seizures/chemically induced , Seizures/genetics , Sequence Homology, Amino Acid , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...