Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Stem Cell Res ; 39: 101522, 2019 08.
Article in English | MEDLINE | ID: mdl-31401456

ABSTRACT

Genetic polymorphism of apolipoprotein E (APOE) confers differential susceptibility to late-onset Alzheimer's disease (LOAD). The ε3 allele of APOE, the most common isoform, does not represent a risk factor for LOAD. In contrast, the ε4 allele is the strongest genetic risk factor for this disease. Here, we present the characterization of four iPSC lines generated from dermal fibroblasts of diagnosed sporadic AD patients using Sendai viral vectors encoding OCT4, SOX2, KLF4 and c-MYC. The iPSCs expressed endogenous pluripotency markers, could be differentiated into the three germ layers, maintained the original genotypes, and were free from Sendai vectors and reprogramming factors.


Subject(s)
Embryoid Bodies/cytology , Apolipoproteins E/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Genotyping Techniques/methods , Humans , Immunohistochemistry , Karyotyping , Kruppel-Like Factor 4 , Microsatellite Repeats/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus/genetics
2.
Neurochem Int ; 123: 95-100, 2019 02.
Article in English | MEDLINE | ID: mdl-29859229

ABSTRACT

Hyperekplexia or startle disease is a dysfunction of inhibitory glycinergic neurotransmission characterized by an exaggerated startle in response to trivial tactile or acoustic stimuli. Although rare, this disorder can have serious consequences, including sudden infant death. One of the most frequent causes of hyperekplexia are mutations in the SLC6A5 gene, encoding the neuronal glycine transporter 2 (GlyT2), a key component of inhibitory glycinergic presynapses involved in synaptic glycine recycling though sodium and chloride-dependent co-transport. Most GlyT2 mutations detected so far are recessive, but two dominant missense mutations have been described. The detailed analysis of these mutations has revealed structural cues on the quaternary structure of GlyT2, and opens the possibility that novel selective pharmacochaperones have potential therapeutic effects in hyperekplexia.


Subject(s)
Glycine Plasma Membrane Transport Proteins/genetics , Hyperekplexia/genetics , Mutation/genetics , Animals , Glycine Plasma Membrane Transport Proteins/metabolism , Humans , Hyperekplexia/metabolism , Neurons/metabolism , Receptors, Glycine/genetics , Synaptic Transmission/genetics
3.
J Biol Chem ; 290(4): 2150-65, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25480793

ABSTRACT

Hyperekplexia or startle disease is a rare clinical syndrome characterized by an exaggerated startle in response to trivial tactile or acoustic stimuli. This neurological disorder can have serious consequences in neonates, provoking brain damage and/or sudden death due to apnea episodes and cardiorespiratory failure. Hyperekplexia is caused by defective inhibitory glycinergic neurotransmission. Mutations in the human SLC6A5 gene encoding the neuronal GlyT2 glycine transporter are responsible for the presynaptic form of the disease. GlyT2 mediates synaptic glycine recycling, which constitutes the main source of releasable transmitter at glycinergic synapses. Although the majority of GlyT2 mutations detected so far are recessive, a dominant negative mutant that affects GlyT2 trafficking does exist. In this study, we explore the properties and structural alterations of the S512R mutation in GlyT2. We analyze its dominant negative effect that retains wild-type GlyT2 in the endoplasmic reticulum (ER), preventing surface expression. We show that the presence of an arginine rather than serine 512 provoked transporter misfolding, enhanced association to the ER-chaperone calnexin, altered association with the coat-protein complex II component Sec24D, and thereby impeded ER exit. The S512R mutant formed oligomers with wild-type GlyT2 causing its retention in the ER. Overexpression of calnexin rescued wild-type GlyT2 from the dominant negative effect of the mutant, increasing the amount of transporter that reached the plasma membrane and dampening the interaction between the wild-type and mutant GlyT2. The ability of chemical chaperones to overcome the dominant negative effect of the disease mutation on the wild-type transporter was demonstrated in heterologous cells and primary neurons.


Subject(s)
Glycine Plasma Membrane Transport Proteins/genetics , Mutation , Stiff-Person Syndrome/genetics , Animals , Biotinylation , COS Cells , Calnexin/metabolism , Cerebral Cortex/metabolism , Chlorocebus aethiops , Densitometry , Dogs , Endoplasmic Reticulum/metabolism , Genes, Dominant , Glycine/chemistry , Glycine Plasma Membrane Transport Proteins/metabolism , Humans , Madin Darby Canine Kidney Cells , Molecular Chaperones/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Stiff-Person Syndrome/metabolism , Synaptic Transmission
4.
J Biol Chem ; 289(49): 34308-24, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25315779

ABSTRACT

Fast inhibitory glycinergic transmission occurs in spinal cord, brainstem, and retina to modulate the processing of motor and sensory information. After synaptic vesicle fusion, glycine is recovered back to the presynaptic terminal by the neuronal glycine transporter 2 (GlyT2) to maintain quantal glycine content in synaptic vesicles. The loss of presynaptic GlyT2 drastically impairs the refilling of glycinergic synaptic vesicles and severely disrupts neurotransmission. Indeed, mutations in the gene encoding GlyT2 are the main presynaptic cause of hyperekplexia in humans. Here, we show a novel endogenous regulatory mechanism that can modulate GlyT2 activity based on a compartmentalized interaction between GlyT2, neuronal plasma membrane Ca(2+)-ATPase (PMCA) isoforms 2 and 3, and Na(+)/Ca(2+)-exchanger 1 (NCX1). This GlyT2·PMCA2,3·NCX1 complex is found in lipid raft subdomains where GlyT2 has been previously found to be fully active. We show that endogenous PMCA and NCX activities are necessary for GlyT2 activity and that this modulation depends on lipid raft integrity. Besides, we propose a model in which GlyT2·PMCA2-3·NCX complex would help Na(+)/K(+)-ATPase in controlling local Na(+) increases derived from GlyT2 activity after neurotransmitter release.


Subject(s)
Glycine Plasma Membrane Transport Proteins/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Sensory Receptor Cells/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , Gene Expression Regulation , Glycine Plasma Membrane Transport Proteins/genetics , Intercellular Signaling Peptides and Proteins , Male , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Peptides/pharmacology , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Plasma Membrane Calcium-Transporting ATPases/genetics , Presynaptic Terminals/drug effects , Primary Cell Culture , Protein Binding , Rats , Rats, Wistar , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism , Synaptic Transmission , Thiourea/analogs & derivatives , Thiourea/pharmacology , beta-Cyclodextrins/pharmacology
5.
PLoS One ; 8(5): e63230, 2013.
Article in English | MEDLINE | ID: mdl-23650557

ABSTRACT

The neuronal transporter GlyT2 is a polytopic, 12-transmembrane domain, plasma membrane glycoprotein involved in the removal and recycling of synaptic glycine from inhibitory synapses. Mutations in the human GlyT2 gene (SLC6A5) that cause deficient glycine transport or defective GlyT2 trafficking are the second most common cause of hyperekplexia or startle disease. In this study we examined several aspects of GlyT2 biogenesis that involve the endoplasmic reticulum chaperone calnexin (CNX). CNX binds transiently to an intermediate under-glycosylated transporter precursor and facilitates GlyT2 processing. In cells expressing GlyT2, transporter accumulation and transport activity were attenuated by siRNA-mediated CNX knockdown and enhanced by CNX overexpression. GlyT2 binding to CNX was mediated by glycan and polypeptide-based interactions as revealed by pharmacological approaches and the behavior of GlyT2 N-glycan-deficient mutants. Moreover, transporter folding appeared to be stabilized by N-glycans. Co-expression of CNX and a fully non-glycosylated mutant rescues glycine transport but not mutant surface expression. Hence, CNX discriminates between different conformational states of GlyT2 displaying a lectin-independent chaperone activity. GlyT2 wild-type and mutant transporters were finally degraded in the lysosome. Our findings provide further insight into GlyT2 biogenesis, and a useful framework for the study of newly synthesized GlyT2 transporters bearing hyperekplexia mutations.


Subject(s)
Calnexin/metabolism , Glycine Plasma Membrane Transport Proteins/biosynthesis , Amino Acid Substitution , Animals , COS Cells , Calnexin/genetics , Chlorocebus aethiops , Glucosidases/antagonists & inhibitors , Glucosidases/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycosylation , Kinetics , Mannosidases/antagonists & inhibitors , Mannosidases/metabolism , Mice , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational/drug effects , Proteolysis , Rats , Thapsigargin/pharmacology , Tunicamycin/pharmacology , Unfolded Protein Response
6.
J Biol Chem ; 287(34): 28986-9002, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22753417

ABSTRACT

Hyperekplexia or startle disease is characterized by an exaggerated startle response, evoked by tactile or auditory stimuli, producing hypertonia and apnea episodes. Although rare, this orphan disorder can have serious consequences, including sudden infant death. Dominant and recessive mutations in the human glycine receptor (GlyR) α1 gene (GLRA1) are the major cause of this disorder. However, recessive mutations in the presynaptic Na(+)/Cl(-)-dependent glycine transporter GlyT2 gene (SLC6A5) are rapidly emerging as a second major cause of startle disease. In this study, systematic DNA sequencing of SLC6A5 revealed a new dominant GlyT2 mutation: pY705C (c.2114A→G) in transmembrane domain 11, in eight individuals from Spain and the United Kingdom. Curiously, individuals harboring this mutation show significant variation in clinical presentation. In addition to classical hyperekplexia symptoms, some individuals had abnormal respiration, facial dysmorphism, delayed motor development, or intellectual disability. We functionally characterized this mutation using molecular modeling, electrophysiology, [(3)H]glycine transport, cell surface expression, and cysteine labeling assays. We found that the introduced cysteine interacts with the cysteine pair Cys-311-Cys-320 in the second external loop of GlyT2. This interaction impairs transporter maturation through the secretory pathway, reduces surface expression, and inhibits transport function. Additionally, Y705C presents altered H(+) and Zn(2+) dependence of glycine transport that may affect the function of glycinergic neurotransmission in vivo.


Subject(s)
Genes, Dominant , Genetic Diseases, Inborn , Glycine Plasma Membrane Transport Proteins , Mutation, Missense , Nerve Tissue Proteins , Nervous System Diseases , Amino Acid Substitution , Animals , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Glycine/genetics , Glycine/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Humans , Ion Transport/genetics , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Presynaptic Terminals , Protein Transport/genetics , Spain , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL
...