Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Mol Biol Lett ; 29(1): 44, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553684

ABSTRACT

Aspartate-glutamate carrier isoform 1 (AGC1) is a carrier responsible for the export of mitochondrial aspartate in exchange for cytosolic glutamate and is part of the malate-aspartate shuttle, essential for the balance of reducing equivalents in the cells. In the brain, mutations in SLC25A12 gene, encoding for AGC1, cause an ultra-rare genetic disease, reported as a neurodevelopmental encephalopathy, whose symptoms include global hypomyelination, arrested psychomotor development, hypotonia and seizures. Among the biological components most affected by AGC1 deficiency are oligodendrocytes, glial cells responsible for myelination processes, and their precursors [oligodendrocyte progenitor cells (OPCs)]. The AGC1 silencing in an in vitro model of OPCs was documented to cause defects of proliferation and differentiation, mediated by alterations of histone acetylation/deacetylation. Disrupting AGC1 activity could possibly reduce the availability of acetyl groups, leading to perturbation of many biological pathways, such as histone modifications and fatty acids formation for myelin production. Here, we explore the transcriptome of mouse OPCs partially silenced for AGC1, reporting results of canonical analyses (differential expression) and pathway enrichment analyses, which highlight a disruption in fatty acids synthesis from both a regulatory and enzymatic stand. We further investigate the cellular effects of AGC1 deficiency through the identification of most affected transcriptional networks and altered alternative splicing. Transcriptional data were integrated with differential metabolite abundance analysis, showing downregulation of several amino acids, including glutamine and aspartate. Taken together, our results provide a molecular foundation for the effects of AGC1 deficiency in OPCs, highlighting the molecular mechanisms affected and providing a list of actionable targets to mitigate the effects of this pathology.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Hereditary Central Nervous System Demyelinating Diseases , Mitochondrial Diseases , Oligodendrocyte Precursor Cells , Psychomotor Disorders , Mice , Animals , Down-Regulation/genetics , Oligodendrocyte Precursor Cells/metabolism , Aspartic Acid/metabolism , Protein Isoforms/metabolism , Fatty Acids
2.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008954

ABSTRACT

AGC1/Aralar/Slc25a12 is the mitochondrial carrier of aspartate-glutamate, the regulatory component of the NADH malate-aspartate shuttle (MAS) that transfers cytosolic redox power to neuronal mitochondria. The deficiency in AGC1/Aralar leads to the human rare disease named "early infantile epileptic encephalopathy 39" (EIEE 39, OMIM # 612949) characterized by epilepsy, hypotonia, arrested psychomotor neurodevelopment, hypo myelination and a drastic drop in brain aspartate (Asp) and N-acetylaspartate (NAA). Current evidence suggest that neurons are the main brain cell type expressing Aralar. However, paradoxically, glial functions such as myelin and Glutamine (Gln) synthesis are markedly impaired in AGC1 deficiency. Herein, we discuss the role of the AGC1/Aralar-MAS pathway in neuronal functions such as Asp and NAA synthesis, lactate use, respiration on glucose, glutamate (Glu) oxidation and other neurometabolic aspects. The possible mechanism triggering the pathophysiological findings in AGC1 deficiency, such as epilepsy and postnatal hypomyelination observed in humans and mice, are also included. Many of these mechanisms arise from findings in the aralar-KO mice model that extensively recapitulate the human disease including the astroglial failure to synthesize Gln and the dopamine (DA) mishandling in the nigrostriatal system. Epilepsy and DA mishandling are a direct consequence of the metabolic defect in neurons due to AGC1/Aralar deficiency. However, the deficits in myelin and Gln synthesis may be a consequence of neuronal affectation or a direct effect of AGC1/Aralar deficiency in glial cells. Further research is needed to clarify this question and delineate the transcellular metabolic fluxes that control brain functions. Finally, we discuss therapeutic approaches successfully used in AGC1-deficient patients and mice.


Subject(s)
Aggrecans/genetics , Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Genetic Predisposition to Disease , Hereditary Central Nervous System Demyelinating Diseases/etiology , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Mitochondrial Diseases/etiology , Mitochondrial Diseases/metabolism , Psychomotor Disorders/etiology , Psychomotor Disorders/metabolism , Aggrecans/deficiency , Aggrecans/metabolism , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antiporters/metabolism , Biomarkers , Brain/metabolism , Combined Modality Therapy , Disease Management , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Energy Metabolism , Genetic Association Studies , Glutamic Acid/metabolism , Hereditary Central Nervous System Demyelinating Diseases/diagnosis , Hereditary Central Nervous System Demyelinating Diseases/therapy , Humans , Malates/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/therapy , Myelin Sheath/metabolism , Oxidation-Reduction , Phenotype , Psychomotor Disorders/diagnosis , Psychomotor Disorders/therapy
3.
J Child Neurol ; 36(10): 805-811, 2021 09.
Article in English | MEDLINE | ID: mdl-34514881

ABSTRACT

Mutations in TUBB4A are associated with a spectrum of neurologic disorders categorized as TUBB4A-related leukoencephalopathy. Affected children can present with global developmental delay or normal early development, followed by a variable loss of skills over time. Further research is needed to characterize the factors associated with the divergent developmental trajectories in this rare monogenic disorder because this phenotypic spectrum is not fully explained by genotype alone.To characterize early psychomotor features, developmental milestones and age of disease onset were collected from medical records (n=54 individuals). Three subcohorts were identified: individuals with the common p.Asp249Asn variant vs all other genotypes with either early (<12 months of age) or late onset of presentation. Individuals with the p.Asp249Asn variant or those with non-p.Asp249Asn genotypes with later disease onset attained key milestones, including head control, sitting, and independent walking. Subjects with early-onset, non-p.Asp249Asn-associated disease were less likely to achieve developmental milestones. Next, we defined the developmental severity as the percentage of milestones attained by age 2 years. The mild form was defined as attaining at least 75% of key developmental milestones. Among cohort categorized as mild, individuals with p.Asp249Asn variant were more likely to lose acquired abilities when compared with non-p.Asp249Asn individuals.Our results suggest multiple influences on developmental trajectory, including a strong contribution from genotype and age of onset. Further studies are needed to identify additional factors that influence overall outcomes to better counsel families and to design clinical trials with appropriate clinical endpoints.


Subject(s)
Basal Ganglia/pathology , Cerebellum/pathology , Developmental Disabilities/complications , Developmental Disabilities/genetics , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Adolescent , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/genetics , Antiporters/deficiency , Antiporters/genetics , Atrophy , Child , Child Development , Child, Preschool , Cohort Studies , Developmental Disabilities/pathology , Female , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Infant , Infant, Newborn , Leukoencephalopathies/pathology , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mutation , Psychomotor Disorders/complications , Psychomotor Disorders/genetics , Psychomotor Disorders/pathology , Retrospective Studies , Tubulin/genetics
4.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34462350

ABSTRACT

Intraventricular hemorrhage (IVH) results in periventricular inflammation, hypomyelination of the white matter, and hydrocephalus in premature infants. No effective therapy exists to prevent these disorders. Peroxisome proliferator activated receptor-γ (PPAR-γ) agonists reduce inflammation, alleviate free radical generation, and enhance microglial phagocytosis, promoting clearance of debris and red blood cells. We hypothesized that activation of PPAR-γ would enhance myelination, reduce hydrocephalus, and promote neurological recovery in newborns with IVH. These hypotheses were tested in a preterm rabbit model of IVH; autopsy brain samples from premature infants with and without IVH were analyzed. We found that IVH augmented PPAR-γ expression in microglia of both preterm human infants and rabbit kits. The treatment with PPAR-γ agonist or PPAR-γ overexpression by adenovirus delivery further elevated PPAR-γ levels in microglia, reduced proinflammatory cytokines, increased microglial phagocytosis, and improved oligodendrocyte progenitor cell (OPC) maturation in kits with IVH. Transcriptomic analyses of OPCs identified previously unrecognized PPAR-γ-induced genes for purinergic signaling, cyclic adenosine monophosphate generation, and antioxidant production, which would reprogram these progenitors toward promoting myelination. RNA-sequencing analyses of microglia revealed PPAR-γ-triggered down-regulation of several proinflammatory genes and transcripts having roles in Parkinson's disease and amyotrophic lateral sclerosis, contributing to neurological recovery in kits with IVH. Accordingly, PPAR-γ activation enhanced myelination and neurological function in kits with IVH. This also enhanced microglial phagocytosis of red blood cells but did not reduce hydrocephalus. Treatment with PPAR-γ agonist might enhance myelination and neurological recovery in premature infants with IVH.


Subject(s)
Cerebral Intraventricular Hemorrhage/metabolism , Myelin Proteins/biosynthesis , PPAR gamma/metabolism , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Antiporters/deficiency , Antiporters/metabolism , Cerebral Intraventricular Hemorrhage/pathology , Disease Models, Animal , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Humans , Infant, Premature , Microglia/metabolism , Mitochondrial Diseases/metabolism , Oligodendroglia/pathology , PPAR gamma/agonists , Psychomotor Disorders/metabolism , Rabbits , Rosiglitazone/pharmacology , Sequence Analysis, RNA/methods
5.
Am J Med Genet A ; 185(10): 3068-3073, 2021 10.
Article in English | MEDLINE | ID: mdl-34037307

ABSTRACT

PYCR2 pathogenic variants lead to an autosomal recessive hypomyelinating leukodystrophy 10 (HLD10), characterized by global developmental delay, microcephaly, facial dysmorphism, movement disorder, and hypomyelination. This study identified the first two unrelated Thai patients with HLD10. Patient 1 harbored the novel compound heterozygous variants, c.257T>G (p.Val86Gly) and c.400G>A (p.Val134Met), whereas patient 2 possessed the homozygous variant, c.400G>A (p.Val134Met), in PYCR2. Haplotype analysis revealed that the two families' members shared a 2.3 Mb region covering the c.400G>A variant, indicating a common ancestry. The variant was estimated to age 1450 years ago. Since the c.400G>A was detected in three out of four mutant alleles and with a common ancestry, this variant might be common in Thai patients. We also reviewed the phenotype and genotype of all 35 previously reported PYCR2 patients and found that majorities of cases were homozygous with a consanguineous family history, except patient 1 and another reported case who were compound heterozygous. All patients had microcephaly and developmental delay. Hypotonia and peripheral spasticity were common. Hypomyelination or delayed myelination was a typical radiographic feature. Here, we report the first two Thai patients with HLD10 with the novel PYCR2 variants expanding the genotypic spectrum and suggest that the c.400G>A might be a common mutation in Thai patients.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Developmental Disabilities/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Microcephaly/genetics , Mitochondrial Diseases/genetics , Movement Disorders/genetics , Psychomotor Disorders/genetics , Pyrroline Carboxylate Reductases/genetics , Adolescent , Alleles , Amino Acid Transport Systems, Acidic/genetics , Antiporters/genetics , Child , Child, Preschool , Developmental Disabilities/complications , Developmental Disabilities/pathology , Female , Genotype , Haplotypes/genetics , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Central Nervous System Demyelinating Diseases/pathology , Homozygote , Humans , Male , Microcephaly/complications , Microcephaly/pathology , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Movement Disorders/complications , Movement Disorders/pathology , Mutation , Pedigree , Phenotype , Psychomotor Disorders/complications , Psychomotor Disorders/pathology , Young Adult
7.
Neuropediatrics ; 51(2): 160-163, 2020 04.
Article in English | MEDLINE | ID: mdl-31766059

ABSTRACT

CASE: We are reporting the third unrelated case of cerebral aspartate-glutamate carrier isoform 1 (AGC1) deficiency. Patient is a 21-month-old Yemeni male who presented with refractory seizure disorder and developmental arrest. Neuroimaging showed cerebral volume loss and diminished N-acetylaspartate (NAA) peak. Whole exome sequencing revealed a homozygous novel missense variant in the SLC25A12 gene. Patient's seizure frequency abated drastically following initiation of ketogenic diet. DISCUSSION AND CONCLUSION: Cerebral AGC1 deficiency results in dysfunction of mitochondrial malate aspartate shuttle, thereby prohibiting myelin synthesis. There are significant phenotypic commonalities between our patient and previously reported cases including intractable epilepsy, psychomotor delay, cerebral atrophy, and diminished NAA peak. Our report also provides evidence regarding beneficial effect of ketogenic diet in this rare neurometabolic epilepsy.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Diet, Ketogenic , Hereditary Central Nervous System Demyelinating Diseases/diagnosis , Mitochondrial Diseases/diagnosis , Mitochondrial Membrane Transport Proteins/genetics , Psychomotor Disorders/diagnosis , Adult , Amino Acid Transport Systems, Acidic/genetics , Antiporters/genetics , Drug Resistant Epilepsy/diet therapy , Hereditary Central Nervous System Demyelinating Diseases/diet therapy , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/physiopathology , Humans , Male , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Mutation, Missense , Protein Isoforms , Psychomotor Disorders/diet therapy , Psychomotor Disorders/genetics , Psychomotor Disorders/physiopathology , Exome Sequencing , Young Adult
8.
Physiol Behav ; 212: 112688, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31622610

ABSTRACT

The aim of the present study was to test the hypothesis that vesicular glutamate transporter 3 (VGluT3) deficiency is associated with cognitive impairments. Male VGluT3 knockout (KO) and wild type (WT) mice were exposed to a behavioral test battery covering paradigms based on spontaneous exploratory behavior and reinforcement-based learning tests. Reversal learning was examined to test the cognitive flexibility. The VGluT3 KO mice clearly exhibited the ability to learn. The social recognition memory of KO mice was intact. The y-maze test revealed weaker working memory of VGluT3 KO mice. No significant learning impairments were noticed in operant conditioning or holeboard discrimination paradigm. In avoidance-based learning tests (Morris water maze and active avoidance), KO mice exhibited slightly slower learning process compared to WT mice, but not a complete learning impairment. In tests based on simple associations (operant conditioning, avoidance learning) an attenuation of cognitive flexibility was observed in KO mice. In conclusion, knocking out VGluT3 results in mild disturbances in working memory and learning flexibility. Apparently, this glutamate transporter is not a major player in learning and memory formation in general. Based on previous characteristics of VGluT3 KO mice we would have expected a stronger deficit. The observed hypolocomotion did not contribute to the mild cognitive disturbances herein reported, either.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/physiology , Avoidance Learning/physiology , Memory, Short-Term/physiology , Amino Acid Transport Systems, Acidic/genetics , Animals , Cognition/physiology , Conditioning, Operant/physiology , Discrimination Learning/physiology , Male , Maze Learning , Mice , Mice, Knockout , Motor Activity , Reversal Learning/physiology
9.
Int J Mol Sci ; 20(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514314

ABSTRACT

Aspartate-Glutamate Carrier 1 (AGC1) deficiency is a rare neurological disease caused by mutations in the solute carrier family 25, member 12 (SLC25A12) gene, encoding for the mitochondrial aspartate-glutamate carrier isoform 1 (AGC1), a component of the malate-aspartate NADH shuttle (MAS), expressed in excitable tissues only. AGC1 deficiency patients are children showing severe hypotonia, arrested psychomotor development, seizures and global hypomyelination. While the effect of AGC1 deficiency in neurons and neuronal function has been deeply studied, little is known about oligodendrocytes and their precursors, the brain cells involved in myelination. Here we studied the effect of AGC1 down-regulation on oligodendrocyte precursor cells (OPCs), using both in vitro and in vivo mouse disease models. In the cell model, we showed that a reduced expression of AGC1 induces a deficit of OPC proliferation leading to their spontaneous and precocious differentiation into oligodendrocytes. Interestingly, this effect seems to be related to a dysregulation in the expression of trophic factors and receptors involved in OPC proliferation/differentiation, such as Platelet-Derived Growth Factor α (PDGFα) and Transforming Growth Factor ßs (TGFßs). We also confirmed the OPC reduction in vivo in AGC1-deficent mice, as well as a proliferation deficit in neurospheres from the Subventricular Zone (SVZ) of these animals, thus indicating that AGC1 reduction could affect the proliferation of different brain precursor cells. These data clearly show that AGC1 impairment alters myelination not only by acting on N-acetyl-aspartate production in neurons but also on OPC proliferation and suggest new potential therapeutic targets for the treatment of AGC1 deficiency.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Mitochondria/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antiporters/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Down-Regulation , Gene Silencing , Lactates/metabolism , Lateral Ventricles/metabolism , Membrane Potential, Mitochondrial , Mice , Neurons/metabolism , Platelet-Derived Growth Factor , Reactive Oxygen Species/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
10.
Int J Mol Sci ; 20(14)2019 Jul 23.
Article in English | MEDLINE | ID: mdl-31340538

ABSTRACT

WW domain-containing oxidoreductase (Wwox) is a putative tumor suppressor. Several germline mutations of Wwox have been associated with infant neurological disorders characterized by epilepsy, growth retardation, and early death. Less is known, however, about the pathological link between Wwox mutations and these disorders or the physiological role of Wwox in brain development. In this study, we examined age-related expression and histological localization of Wwox in forebrains as well as the effects of loss of function mutations in the Wwox gene in the immature cortex of a rat model of lethal dwarfism with epilepsy (lde/lde). Immunostaining revealed that Wwox is expressed in neurons, astrocytes, and oligodendrocytes. lde/lde cortices were characterized by a reduction in neurite growth without a reduced number of neurons, severe reduction in myelination with a reduced number of mature oligodendrocytes, and a reduction in cell populations of astrocytes and microglia. These results indicate that Wwox is essential for normal development of neurons and glial cells in the cerebral cortex.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Cerebral Cortex/metabolism , Dwarfism/genetics , Epilepsy/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Mitochondrial Diseases/genetics , Neurogenesis/genetics , Psychomotor Disorders/genetics , Tumor Suppressor Proteins/genetics , WW Domain-Containing Oxidoreductase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antiporters/genetics , Antiporters/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Cell Count , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Disease Models, Animal , Dwarfism/metabolism , Dwarfism/pathology , Epilepsy/metabolism , Epilepsy/pathology , Gene Expression Regulation, Developmental , Germ-Line Mutation , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Hereditary Central Nervous System Demyelinating Diseases/pathology , Male , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Prosencephalon/growth & development , Prosencephalon/metabolism , Prosencephalon/pathology , Psychomotor Disorders/metabolism , Psychomotor Disorders/pathology , Rats , Rats, Transgenic , Signal Transduction , Tumor Suppressor Proteins/deficiency , WW Domain-Containing Oxidoreductase/deficiency
11.
J Neurosci ; 39(23): 4434-4447, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30926748

ABSTRACT

Noise-induced excitotoxicity is thought to depend on glutamate. However, the excitotoxic mechanisms are unknown, and the necessity of glutamate for synapse loss or regeneration is unclear. Despite absence of glutamatergic transmission from cochlear inner hair cells in mice lacking the vesicular glutamate transporter-3 (Vglut3KO ), at 9-11 weeks, approximately half the number of synapses found in Vglut3WT were maintained as postsynaptic AMPA receptors juxtaposed with presynaptic ribbons and voltage-gated calcium channels (CaV1.3). Synapses were larger in Vglut3KO than Vglut3WT In Vglut3WT and Vglut3+/- mice, 8-16 kHz octave-band noise exposure at 100 dB sound pressure level caused a threshold shift (∼40 dB) and a loss of synapses (>50%) at 24 h after exposure. Hearing threshold and synapse number partially recovered by 2 weeks after exposure as ribbons became larger, whereas recovery was significantly better in Vglut3WT Noise exposure at 94 dB sound pressure level caused auditory threshold shifts that fully recovered in 2 weeks, whereas suprathreshold hearing recovered faster in Vglut3WT than Vglut3+/- These results, from mice of both sexes, suggest that spontaneous repair of synapses after noise depends on the level of Vglut3 protein or the level of glutamate release during the recovery period. Noise-induced loss of presynaptic ribbons or postsynaptic AMPA receptors was not observed in Vglut3KO , demonstrating its dependence on vesicular glutamate release. In Vglut3WT and Vglut3+/-, noise exposure caused unpairing of presynaptic ribbons and presynaptic CaV1.3, but not in Vglut3KO where CaV1.3 remained clustered with ribbons at presynaptic active zones. These results suggest that, without glutamate release, noise-induced presynaptic Ca2+ influx was insufficient to disassemble the active zone. However, synapse volume increased by 2 weeks after exposure in Vglut3KO , suggesting glutamate-independent mechanisms.SIGNIFICANCE STATEMENT Hearing depends on glutamatergic transmission mediated by Vglut3, but the role of glutamate in synapse loss and repair is unclear. Here, using mice of both sexes, we show that one copy of the Vglut3 gene is sufficient for noise-induced threshold shift and loss of ribbon synapses, but both copies are required for normal recovery of hearing function and ribbon synapse number. Impairment of the recovery process in mice having only one functional copy suggests that glutamate release may promote synapse regeneration. At least one copy of the Vglut3 gene is necessary for noise-induced synapse loss. Although the excitotoxic mechanism remains unknown, these findings are consistent with the presumption that glutamate is the key mediator of noise-induced synaptopathy.


Subject(s)
Amino Acid Transport Systems, Acidic/physiology , Glutamic Acid/physiology , Hair Cells, Auditory, Inner/physiology , Hearing Loss, Noise-Induced/physiopathology , Synapses/physiology , Aging/physiology , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/genetics , Animals , Auditory Threshold/physiology , Calcium/metabolism , Evoked Potentials, Auditory , Exocytosis , Female , Gene Dosage , Genes, Reporter , Hair Cells, Auditory, Outer/physiology , Ion Transport , Male , Mice , Mice, Knockout , Receptors, AMPA/physiology , Recovery of Function , Spiral Ganglion/cytology , Synapses/ultrastructure
12.
Mol Genet Metab ; 125(1-2): 118-126, 2018 09.
Article in English | MEDLINE | ID: mdl-30031689

ABSTRACT

Folate metabolism in the brain is critically important and serves a number of vital roles in nucleotide synthesis, single carbon metabolism/methylation, amino acid metabolism, and mitochondrial translation. Genetic defects in almost every enzyme of folate metabolism have been reported to date, and most have neurological sequelae. We report 2 patients presenting with a neurometabolic disorder associated with biallelic variants in the MTHFS gene, encoding 5,10-methenyltetrahydrofolate synthetase. Both patients presented with microcephaly, short stature, severe global developmental delay, progressive spasticity, epilepsy, and cerebral hypomyelination. Baseline CSF 5-methyltetrahydrolate (5-MTHF) levels were in the low-normal range. The first patient was treated with folinic acid, which resulted in worsening cerebral folate deficiency. Treatment in this patient with a combination of oral L-5-methyltetrahydrofolate and intramuscular methylcobalamin was able to increase CSF 5-MTHF levels, was well tolerated over a 4 month period, and resulted in subjective mild improvements in functioning. Measurement of MTHFS enzyme activity in fibroblasts confirmed reduced activity. The direct substrate of the MTHFS reaction, 5-formyl-THF, was elevated 30-fold in patient fibroblasts compared to control, supporting the hypothesis that the pathophysiology of this disorder is a manifestation of toxicity from this metabolite.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Carbon-Nitrogen Ligases/genetics , Epilepsy/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Microcephaly/genetics , Mitochondrial Diseases/genetics , Psychomotor Disorders/genetics , Amino Acid Transport Systems, Acidic/cerebrospinal fluid , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Antiporters/cerebrospinal fluid , Antiporters/genetics , Antiporters/metabolism , Brain/metabolism , Brain/pathology , Carbon-Nitrogen Ligases/cerebrospinal fluid , Carbon-Nitrogen Ligases/deficiency , Carbon-Nitrogen Ligases/metabolism , Epilepsy/cerebrospinal fluid , Epilepsy/complications , Epilepsy/pathology , Female , Folate Receptor 1/deficiency , Hereditary Central Nervous System Demyelinating Diseases/cerebrospinal fluid , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Humans , Male , Metabolic Diseases/cerebrospinal fluid , Metabolic Diseases/complications , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Microcephaly/cerebrospinal fluid , Microcephaly/complications , Microcephaly/pathology , Mitochondrial Diseases/cerebrospinal fluid , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Nervous System Malformations/cerebrospinal fluid , Nervous System Malformations/complications , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Neuroaxonal Dystrophies , Psychomotor Disorders/cerebrospinal fluid , Psychomotor Disorders/complications , Psychomotor Disorders/metabolism , Tetrahydrofolates/cerebrospinal fluid , Tetrahydrofolates/metabolism
13.
Am J Med Genet A ; 176(6): 1443-1448, 2018 06.
Article in English | MEDLINE | ID: mdl-29696782

ABSTRACT

Early-onset epileptic encephalopathies (EOEEs) are a genetically heterogeneous collection of severe epilepsies often associated with psychomotor regression. Mutations in SZT2, a known seizure threshold regulator gene, are a newly identified cause of EOEE. We present an individual with EOEE, macrocephaly, and developmental regression with compound heterozygous mutations in SZT2 as identified by whole exome sequencing. Serial imaging characterized the novel finding of progressive loss of central myelination. This case expands our clinical understanding of the SZT2-phenotype and emphasizes the role of this gene in the diagnostic investigation for EOEE and leukoencephalopathies.


Subject(s)
Leukoencephalopathies/genetics , Mutation , Nerve Tissue Proteins/genetics , Spasms, Infantile/genetics , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/genetics , Antiporters/deficiency , Antiporters/genetics , Child, Preschool , Developmental Disabilities/genetics , Female , Hereditary Central Nervous System Demyelinating Diseases/diagnostic imaging , Hereditary Central Nervous System Demyelinating Diseases/etiology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Heterozygote , Humans , Infant , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/etiology , Magnetic Resonance Imaging , Megalencephaly/diagnostic imaging , Megalencephaly/genetics , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Psychomotor Disorders/diagnostic imaging , Psychomotor Disorders/etiology , Psychomotor Disorders/genetics , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/etiology
14.
Cell Mol Neurobiol ; 38(1): 37-52, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28776199

ABSTRACT

Maintenance of the homeostasis in a constantly changing environment is a fundamental process of life. Disturbances of the homeostatic balance is defined as stress response and is induced by wide variety of challenges called stressors. Being the main excitatory neurotransmitter of the central nervous system glutamate is important in the adaptation process of stress regulating both the catecholaminergic system and the hypothalamic-pituitary-adrenocortical axis. Data are accumulating about the role of different glutamatergic receptors at all levels of these axes, but little is known about the contribution of different vesicular glutamate transporters (VGluT1-3) characterizing the glutamatergic neurons. Here we summarize basic knowledge about VGluTs, their role in physiological regulation of stress adaptation, as well as their contribution to stress-related psychopathology. Most of our knowledge comes from the VGluT3 knockout mice, as VGluT1 and 2 knockouts are not viable. VGluT3 was discovered later than, and is not as widespread as the VGluT1 and 2. It may co-localize with other transmitters, and participate in retrograde signaling; as such its role might be unique. Previous reports using VGluT3 knockout mice showed enhanced anxiety and innate fear compared to wild type. Moreover, these knockout animals had enhanced resting corticotropin-releasing hormone mRNA levels in the hypothalamus and disturbed glucocorticoid stress responses. In conclusion, VGluT3 participates in stress adaptation regulation. The neuroendocrine changes observed in VGluT3 knockout mice may contribute to their anxious, fearful phenotype.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Stress, Psychological/metabolism , Stress, Psychological/psychology , Amino Acid Transport Systems, Acidic/genetics , Animals , Brain/metabolism , Corticotropin-Releasing Hormone/metabolism , Fear/physiology , Fear/psychology , Glutamic Acid/metabolism , Humans , Mice , Mice, Knockout , Neural Pathways/metabolism , Stress, Psychological/genetics , Vesicular Glutamate Transport Proteins/physiology
16.
Neurology ; 89(17): 1821-1828, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-28931644

ABSTRACT

OBJECTIVE: To identify the gene defect in patients with hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC) who are negative for TUBB4A mutations. METHODS: We performed homozygosity mapping and whole exome sequencing (WES) to detect the disease-causing variant. We used a Taqman assay for population screening. We developed a luciferase reporter construct to investigate the effect of the promoter mutation on expression. RESULTS: Sixteen patients from 14 families from different countries fulfilling the MRI criteria for H-ABC exhibited a similar, severe clinical phenotype, including lack of development and a severe epileptic encephalopathy. The majority of patients had a known Roma ethnic background. Single nucleotide polymorphism array analysis in 5 patients identified one large overlapping homozygous region on chromosome 13. WES in 2 patients revealed a homozygous deletion in the promoter region of UFM1. Sanger sequencing confirmed homozygosity for this variant in all 16 patients. All patients shared a common haplotype, indicative of a founder effect. Screening of 1,000 controls from different European Roma panels demonstrated an overall carrier rate of the mutation of 3%-25%. Transfection assays showed that the deletion significantly reduced expression in specific CNS cell lines. CONCLUSIONS: UFM1 encodes ubiquitin-fold modifier 1 (UFM1), a member of the ubiquitin-like family involved in posttranslational modification of proteins. Its exact biological role is unclear. This study associates a UFM1 gene defect with a disease and sheds new light on possible UFM1 functional networks.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Basal Ganglia/pathology , Cerebellum/pathology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Mitochondrial Diseases/genetics , Polymorphism, Single Nucleotide/genetics , Proteins/genetics , Psychomotor Disorders/genetics , Adolescent , Adult , Amino Acid Transport Systems, Acidic/genetics , Antiporters/genetics , Atrophy/etiology , Basal Ganglia/diagnostic imaging , Cell Line, Tumor/pathology , Cerebellum/diagnostic imaging , Child , Child, Preschool , DNA Mutational Analysis , Family Health , Female , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Central Nervous System Demyelinating Diseases/diagnostic imaging , Humans , Image Processing, Computer-Assisted , Italy , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnostic imaging , Psychomotor Disorders/complications , Psychomotor Disorders/diagnostic imaging , Transfection , Tubulin/genetics , Young Adult
17.
Am J Hum Genet ; 100(6): 969-977, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575651

ABSTRACT

Progressive limb spasticity and cerebellar ataxia are frequently found together in clinical practice and form a heterogeneous group of degenerative disorders that are classified either as pure spastic ataxia or as complex spastic ataxia with additional neurological signs. Inheritance is either autosomal dominant or autosomal recessive. Hypomyelinating features on MRI are sometimes seen with spastic ataxia, but this is usually mild in adults and severe and life limiting in children. We report seven individuals with an early-onset spastic-ataxia phenotype. The individuals come from three families of different ethnic backgrounds. Affected members of two families had childhood onset disease with very slow progression. They are still alive in their 30s and 40s and show predominant ataxia and cerebellar atrophy features on imaging. Affected members of the third family had a similar but earlier-onset presentation associated with brain hypomyelination. Using a combination of homozygozity mapping and exome sequencing, we mapped this phenotype to deleterious nonsense or homeobox domain missense mutations in NKX6-2. NKX6-2 encodes a transcriptional repressor with early high general and late focused CNS expression. Deficiency of its mouse ortholog results in widespread hypomyelination in the brain and optic nerve, as well as in poor motor coordination in a pattern consistent with the observed human phenotype. In-silico analysis of human brain expression and network data provides evidence that NKX6-2 is involved in oligodendrocyte maturation and might act within the same pathways of genes already associated with central hypomyelination. Our results support a non-redundant developmental role of NKX6-2 in humans and imply that NKX6-2 mutations should be considered in the differential diagnosis of spastic ataxia and hypomyelination.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Central Nervous System Demyelinating Diseases/genetics , Homeodomain Proteins/genetics , Intellectual Disability/complications , Intellectual Disability/genetics , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Muscle Spasticity/complications , Muscle Spasticity/genetics , Mutation/genetics , Optic Atrophy/complications , Optic Atrophy/genetics , Psychomotor Disorders/complications , Psychomotor Disorders/genetics , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/genetics , Adult , Amino Acid Sequence , Amino Acid Transport Systems, Acidic/genetics , Antiporters/genetics , Brain/embryology , Brain/metabolism , Child , Female , Gene Regulatory Networks , Homeodomain Proteins/chemistry , Humans , Infant , Male , Pedigree , Phenotype , Young Adult
18.
J Neurochem ; 142(1): 132-139, 2017 07.
Article in English | MEDLINE | ID: mdl-28429368

ABSTRACT

ARALAR/AGC1 (aspartate-glutamate mitochondrial carrier 1) is an important component of the NADH malate-aspartate shuttle (MAS). AGC1-deficiency is a rare disease causing global cerebral hypomyelination, developmental arrest, hypotonia, and epilepsy (OMIM ID #612949); the aralar-KO mouse recapitulates the major findings in humans. This study was aimed at understanding the impact of ARALAR-deficiency in brain lactate levels as a biomarker. We report that lactate was equally abundant in wild-type and aralar-KO mouse brain in vivo at postnatal day 17. We find that lactate production upon mitochondrial blockade depends on up-regulation of lactate formation in astrocytes rather than in neurons. However, ARALAR-deficiency decreased cell respiration in neurons, not astrocytes, which maintained unchanged respiration and lactate production. As the primary site of ARALAR-deficiency is neuronal, this explains the lack of accumulation of brain lactate in ARALAR-deficiency in humans and mice. On the other hand, we find that the cytosolic and mitochondrial components of the glycerol phosphate shuttle are present in astrocytes with similar activities. This suggests that glycerol phosphate shuttle is the main NADH shuttle in astrocytes and explains the absence of effects of ARALAR-deficiency in these cells.


Subject(s)
Aggrecans/genetics , Aggrecans/metabolism , Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Hereditary Central Nervous System Demyelinating Diseases/genetics , Lactic Acid/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neurons/metabolism , Psychomotor Disorders/genetics , Amino Acid Transport Systems, Acidic/genetics , Animals , Antiporters/genetics , Astrocytes/metabolism , Brain Chemistry/genetics , Glucose/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption/genetics
19.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1422-1435, 2017 06.
Article in English | MEDLINE | ID: mdl-28235644

ABSTRACT

The mitochondrial aspartate-glutamate carrier isoform 1 (AGC1) catalyzes a Ca2+-stimulated export of aspartate to the cytosol in exchange for glutamate, and is a key component of the malate-aspartate shuttle which transfers NADH reducing equivalents from the cytosol to mitochondria. By sustaining the complete glucose oxidation, AGC1 is thought to be important in providing energy for cells, in particular in the CNS and muscle where this protein is mainly expressed. Defects in the AGC1 gene cause AGC1 deficiency, an infantile encephalopathy with delayed myelination and reduced brain N-acetylaspartate (NAA) levels, the precursor of myelin synthesis in the CNS. Here, we show that undifferentiated Neuro2A cells with down-regulated AGC1 display a significant proliferation deficit associated with reduced mitochondrial respiration, and are unable to synthesize NAA properly. In the presence of high glutamine oxidation, cells with reduced AGC1 restore cell proliferation, although oxidative stress increases and NAA synthesis deficit persists. Our data suggest that the cellular energetic deficit due to AGC1 impairment is associated with inappropriate aspartate levels to support neuronal proliferation when glutamine is not used as metabolic substrate, and we propose that delayed myelination in AGC1 deficiency patients could be attributable, at least in part, to neuronal loss combined with lack of NAA synthesis occurring during the nervous system development.


Subject(s)
Amino Acid Transport Systems/biosynthesis , Aspartic Acid/analogs & derivatives , Cell Proliferation , Down-Regulation , Mitochondrial Proteins/biosynthesis , Neurons/metabolism , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Antiporters/deficiency , Antiporters/genetics , Antiporters/metabolism , Aspartic Acid/biosynthesis , Cell Line , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Neurons/pathology , Psychomotor Disorders/genetics , Psychomotor Disorders/metabolism , Psychomotor Disorders/pathology
20.
Mol Vis ; 22: 1198-1212, 2016.
Article in English | MEDLINE | ID: mdl-27746674

ABSTRACT

PURPOSE: To characterize the vision phenotype of mice lacking Aralar/AGC1/Slc25a12, the mitochondrial aspartate-glutamate carrier mutated in global cerebral hypomyelination (OMIM 612949). METHODS: We tested overnight dark-adapted control and aralar-deficient mice for the standard full electroretinogram (ERG) response. The metabolic stress of dark-adaptation was reduced by 5 min illumination after which the ERG response was monitored in darkness. We used the electrical response to two identical saturating light flashes (paired-flash stimulation) to isolate the inner retina and photoreceptor responses. Retinal morphology was examined with hematoxylin and eosin staining, immunohistochemistry of antibodies against retinal cells, and 4',6-diamidino-2-phenylindole (DAPI) labeling. RESULTS: Aralar plays a pivotal role in retina metabolism as aralar provides de novo synthesis pathway for glutamine, protects glutamate from oxidation, and is required for efficient glucose oxidative metabolism. Aralar-deficient mice are not blind as their retinas have light-evoked activity. However, we report an approximate 50% decrease in the ERG amplitude response in the light-evoked activity of dark-adapted retinas from aralar-deficient mice, in spite of normal retina histology. The defective response is partly reversed by exposure to a brief illumination period, which lowers the metabolic stress of dark-adaptation. The metabolic stress and ERG alteration takes place primarily in photoreceptors, but the response to two flashes applied in fast succession also revealed an alteration in synaptic transmission consistent with an imbalance of glutamate and an energy deficit in the inner retina neurons. CONCLUSIONS: We propose that compromised glucose oxidation and altered glutamine and glutamate metabolism in the absence of aralar are responsible for the phenotype reported.


Subject(s)
Aggrecans/physiology , Glucose/metabolism , Glutamine/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Retina/metabolism , Vision Disorders/metabolism , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antiporters/deficiency , Antiporters/metabolism , Dark Adaptation , Electroretinography , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Photic Stimulation , Psychomotor Disorders/metabolism , Retina/physiopathology , Retinal Bipolar Cells/physiology , Synaptic Transmission , Vision Disorders/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...