Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Glia ; 71(3): 509-523, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36354016

RESUMEN

Healthy myelin sheaths consist of multiple compacted membrane layers closely encasing the underlying axon. The ultrastructure of CNS myelin requires specialized structural myelin proteins, including the transmembrane-tetraspan proteolipid protein (PLP) and the Ig-CAM myelin-associated glycoprotein (MAG). To better understand their functional relevance, we asked to what extent the axon/myelin-units display similar morphological changes if PLP or MAG are lacking. We thus used focused ion beam-scanning electron microscopy (FIB-SEM) to re-investigate axon/myelin-units side-by-side in Plp- and Mag-null mutant mice. By three-dimensional reconstruction and morphometric analyses, pathological myelin outfoldings extend up to 10 µm longitudinally along myelinated axons in both models. More than half of all assessed outfoldings emerge from internodal myelin. Unexpectedly, three-dimensional reconstructions demonstrated that both models displayed complex axonal pathology underneath the myelin outfoldings, including axonal sprouting. Axonal anastomosing was additionally observed in Plp-null mutant mice. Importantly, normal-appearing axon/myelin-units displayed significantly increased axonal diameters in both models according to quantitative assessment of electron micrographs. These results imply that healthy CNS myelin sheaths facilitate normal axonal diameters and shape, a function that is impaired when structural myelin proteins PLP or MAG are lacking.


Asunto(s)
Sistema Nervioso Central , Proteína Proteolipídica de la Mielina , Vaina de Mielina , Glicoproteína Asociada a Mielina , Animales , Ratones , Axones/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Noqueados , Microscopía Electrónica de Rastreo , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Glicoproteína Asociada a Mielina/genética , Proteína Proteolipídica de la Mielina/genética
2.
Ann Neurol ; 89(4): 711-725, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33410190

RESUMEN

OBJECTIVE: Multiple sclerosis (MS) is a chronic neuroinflammatory and neurodegenerative disease of unknown etiology. Although the prevalent view regards a CD4+ -lymphocyte autoimmune reaction against myelin at the root of the disease, recent studies propose autoimmunity as a secondary reaction to idiopathic brain damage. To gain knowledge about this possibility we investigated the presence of axonal and myelinic morphological alterations, which could implicate imbalance of axon-myelin units as primary event in MS pathogenesis. METHODS: Using high resolution imaging histological brain specimens from patients with MS and non-neurological/non-MS controls, we explored molecular changes underpinning imbalanced interaction between axon and myelin in normal appearing white matter (NAWM), a region characterized by normal myelination and absent inflammatory activity. RESULTS: In MS brains, we detected blister-like swellings formed by myelin detachment from axons, which were substantially less frequently retrieved in non-neurological/non-MS controls. Swellings in MS NAWM presented altered glutamate receptor expression, myelin associated glycoprotein (MAG) distribution, and lipid biochemical composition of myelin sheaths. Changes in tethering protein expression, widening of nodes of Ranvier and altered distribution of sodium channels in nodal regions of otherwise normally myelinated axons were also present in MS NAWM. Finally, we demonstrate a significant increase, compared with controls, in citrullinated proteins in myelin of MS cases, pointing toward biochemical modifications that may amplify the immunogenicity of MS myelin. INTERPRETATION: Collectively, the impaired interaction of myelin and axons potentially leads to myelin disintegration. Conceptually, the ensuing release of (post-translationally modified) myelin antigens may elicit a subsequent immune attack in MS. ANN NEUROL 2021;89:711-725.


Asunto(s)
Axones/patología , Esclerosis Múltiple/patología , Vaina de Mielina/patología , Sustancia Blanca/patología , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Dermatoglifia del ADN , Femenino , Humanos , Inmunohistoquímica , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Imagen Molecular , Esclerosis Múltiple/diagnóstico , Glicoproteína Asociada a Mielina/biosíntesis , Glicoproteína Asociada a Mielina/genética , Neuroimagen , Nódulos de Ranvier/patología , Receptores de Glutamato/biosíntesis , Canales de Sodio/metabolismo
3.
J Neurosci ; 40(48): 9293-9305, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33087477

RESUMEN

Aralar/AGC1/Slc25a12, the mitochondrial aspartate-glutamate carrier expressed in neurons, is the regulatory component of the NADH malate-aspartate shuttle. AGC1 deficiency is a neuropediatric rare disease characterized by hypomyelination, hypotonia, developmental arrest, and epilepsy. We have investigated whether ß-hydroxybutyrate (ßOHB), the main ketone body (KB) produced in ketogenic diet (KD), is neuroprotective in aralar-knock-out (KO) neurons and mice. We report that ßOHB efficiently recovers aralar-KO neurons from deficits in basal-stimulated and glutamate-stimulated respiration, effects requiring ßOHB entry into the neuron, and protects from glutamate excitotoxicity. Aralar-deficient mice were fed a KD to investigate its therapeutic potential early in development, but this approach was unfeasible. Therefore, aralar-KO pups were treated without distinction of gender with daily intraperitoneal injections of ßOHB during 5 d. This treatment resulted in a recovery of striatal markers of the dopaminergic system including dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC)/DA ratio, and vesicular monoamine transporter 2 (VMAT2) protein. Regarding postnatal myelination, myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) myelin proteins were markedly increased in the cortices of ßOHB-treated aralar-KO mice. Although brain Asp and NAA levels did not change by ßOHB administration, a 4-d ßOHB treatment to aralar-KO, but not to control, neurons led to a substantial increase in Asp (3-fold) and NAA (4-fold) levels. These results suggest that the lack of increase in brain Asp and NAA is possibly because of its active utilization by the aralar-KO brain and the likely involvement of neuronal NAA in postnatal myelination in these mice. The effectiveness of ßOHB as a therapeutic treatment in AGC1 deficiency deserves further investigation.SIGNIFICANCE STATEMENTAralar deficiency induces a fatal phenotype in humans and mice and is associated with impaired neurodevelopment, epilepsy, and hypomyelination. In neurons, highly expressing aralar, its deficiency causes a metabolic blockade hampering mitochondrial energetics and respiration. Here, we find that ßOHB, the main metabolic product in KD, recovers defective mitochondrial respiration bypassing the metabolic failure in aralar-deficient neurons. ßOHB oxidation in mitochondria boosts the synthesis of cytosolic aspartate (Asp) and NAA, which is impeded by aralar deficiency, presumably through citrate-malate shuttle. In aralar-knock-out (KO) mice, ßOHB recovers from the drastic drop in specific dopaminergic and myelin markers. The ßOHB-induced myelin synthesis occurring together with the marked increment in neuronal NAA synthesis supports the role of NAA as a lipid precursor during postnatal myelination.


Asunto(s)
Ácido 3-Hidroxibutírico/fisiología , Agrecanos/fisiología , Encéfalo/fisiología , Dieta Cetogénica , Vías Nerviosas/fisiología , Neuronas/fisiología , Ácido 3-Hidroxibutírico/administración & dosificación , Ácido 3-Hidroxibutírico/farmacología , Agrecanos/genética , Aminoácidos/metabolismo , Animales , Dopamina/fisiología , Femenino , Ácido Glutámico/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/genética , Vaina de Mielina/fisiología , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/fisiología , Consumo de Oxígeno/fisiología , Respiración/efectos de los fármacos , Proteínas de Transporte Vesicular de Monoaminas/fisiología
4.
J Neurosci ; 39(1): 63-77, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30446529

RESUMEN

Sulfatides and gangliosides are raft-associated glycolipids essential for maintaining myelinated nerve integrity. Mice deficient in sulfatide (cerebroside sulfotransferase knock-out, CST-/-) or complex gangliosides (ß-1,4-N-acetylegalactosaminyltransferase1 knock-out, GalNAc-T-/-) display prominent disorganization of proteins at the node of Ranvier (NoR) in early life and age-dependent neurodegeneration. Loss of neuronal rather than glial complex gangliosides underpins the GalNAc-T-/- phenotype, as shown by neuron- or glial-specific rescue, whereas sulfatide is principally expressed and functional in glial membranes. The similarities in NoR phenotype of CST-/-, GalNAc-T-/-, and axo-glial protein-deficient mice suggests that these glycolipids stabilize membrane proteins including neurofascin155 (NF155) and myelin-associated glycoprotein (MAG) at axo-glial junctions. To assess the functional interactions between sulfatide and gangliosides, CST-/- and GalNAc-T-/- genotypes were interbred. CST-/-× GalNAc-T-/- mice develop normally to postnatal day 10 (P10), but all die between P20 and P25, coinciding with peak myelination. Ultrastructural, immunohistological, and biochemical analysis of either sex revealed widespread axonal degeneration and disruption to the axo-glial junction at the NoR. In addition to sulfatide-dependent loss of NF155, CST-/- × GalNAc-T-/- mice exhibited a major reduction in MAG protein levels in CNS myelin compared with WT and single-lipid-deficient mice. The CST-/- × GalNAc-T-/- phenotype was fully restored to that of CST-/- mice by neuron-specific expression of complex gangliosides, but not by their glial-specific expression nor by the global expression of a-series gangliosides. These data indicate that sulfatide and complex b-series gangliosides on the glial and neuronal membranes, respectively, act in concert to promote NF155 and MAG in maintaining the stable axo-glial interactions essential for normal nerve function.SIGNIFICANCE STATEMENT Sulfatides and complex gangliosides are membrane glycolipids with important roles in maintaining nervous system integrity. Node of Ranvier maintenance in particular requires stable compartmentalization of multiple membrane proteins. The axo-glial adhesion molecules neurofascin155 (NF155) and myelin-associated glycoprotein (MAG) require membrane microdomains containing either sulfatides or complex gangliosides to localize and function effectively. The cooperative roles of these microdomains and associated proteins are unknown. Here, we show vital interdependent roles for sulfatides and complex gangliosides because double (but not single) deficiency causes a rapidly lethal phenotype at an early age. These findings suggest that sulfatides and complex gangliosides on opposing axo-glial membranes are responsible for essential tethering of the axo-glial junction proteins NF155 and MAG, which interact to maintain the nodal complex.


Asunto(s)
Axones/fisiología , Gangliósidos/metabolismo , Gangliósidos/fisiología , Vaina de Mielina/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sulfoglicoesfingolípidos/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Femenino , Genotipo , Esperanza de Vida , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/fisiología , N-Acetilgalactosaminiltransferasas/genética , Factores de Crecimiento Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Nódulos de Ranvier/fisiología , Sulfotransferasas/genética , Sulfotransferasas/fisiología
5.
J Neurochem ; 147(6): 764-783, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30261098

RESUMEN

The close association of myelinated axons and their myelin sheaths involves numerous intercellular molecular interactions. For example, myelin-associated glycoprotein (MAG) mediates myelin-to-axon adhesion and signalling via molecules on the axonal surface. However, knowledge about intracellular binding partners of myelin proteins, including MAG, has remained limited. The two splice isoforms of MAG, S- and L-MAG, display distinct cytoplasmic domains and spatiotemporal expression profiles. We used yeast two-hybrid screening to identify interaction partners of L-MAG and found the dynein light chain DYNLL1 (also termed dynein light chain 8). DYNLL1 homodimers are known to facilitate dimerization of target proteins. L-MAG and DYNLL1 associate with high affinity, as confirmed with recombinant proteins in vitro. Structural analyses of the purified complex indicate that the DYNLL1-binding segment is localized close to the L-MAG C terminus, next to the Fyn kinase Tyr phosphorylation site. The crystal structure of the complex between DYNLL1 and its binding segment on L-MAG shows 2 : 2 binding in a parallel arrangement, indicating a heterotetrameric complex. The homology between L-MAG and previously characterized DYNLL1-ligands is limited, and some details of binding site interactions are unique for L-MAG. The structure of the complex between the entire L-MAG cytoplasmic domain and DYNLL1, as well as that of the extracellular domain of MAG, were modelled based on small-angle X-ray scattering data, allowing structural insights into L-MAG interactions on both membrane surfaces. Our data imply that DYNLL1 dimerizes L-MAG, but not S-MAG, through the formation of a specific 2 : 2 heterotetramer. This arrangement is likely to affect, in an isoform-specific manner, the functions of MAG in adhesion and myelin-to-axon signalling. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Read the Editorial Highlight for this article on page 712.


Asunto(s)
Dineínas/biosíntesis , Glicoproteína Asociada a Mielina/biosíntesis , Animales , Axones/fisiología , Sitios de Unión , Dineínas Citoplasmáticas , Dineínas/química , Dineínas/genética , Espacio Extracelular/metabolismo , Ratones , Modelos Moleculares , Glicoproteína Asociada a Mielina/química , Glicoproteína Asociada a Mielina/genética , Fibras Nerviosas/metabolismo , Fibras Nerviosas/ultraestructura , Neuroglía/fisiología , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/metabolismo , Dispersión de Radiación , Nervio Ciático/citología , Nervio Ciático/metabolismo , Rayos X
6.
J Neurovirol ; 23(3): 376-384, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27981441

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a neurotropic virus linked to a range of acute and chronic neurological disorders affecting distinct regions of the brain. Unusually, HSV-1 entry into cells requires the interaction of viral proteins glycoprotein D (gD) and glycoprotein B (gB) with distinct cellular receptor proteins. Several different gD and gB receptors have been identified, including TNFRSF14/HVEM and PVRL1/nectin 1 as gD receptors and PILRA, MAG, and MYH9 as gB receptors. We investigated the expression of these receptor molecules in different areas of the adult and developing human brain using online transcriptome databases. Whereas all HSV-1 receptors showed distinct expression patterns in different brain areas, the Allan Brain Atlas (ABA) reported increased expression of both gD and gB receptors in the hippocampus. Specifically, for PVRL1, TNFRFS14, and MYH9, the differential z scores for hippocampal expression, a measure of relative levels of increased expression, rose to 2.9, 2.9, and 2.5, respectively, comparable to the z score for the archetypical hippocampus-enriched mineralocorticoid receptor (NR3C2, z = 3.1). These data were confirmed at the Human Brain Transcriptome (HBT) database, but HBT data indicate that MAG expression is also enriched in hippocampus. The HBT database allowed the developmental pattern of expression to be investigated; we report that all HSV1 receptors markedly increase in expression levels between gestation and the postnatal/adult periods. These results suggest that differential receptor expression levels of several HSV-1 gD and gB receptors in the adult hippocampus are likely to underlie the susceptibility of this brain region to HSV-1 infection.


Asunto(s)
Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno/genética , Receptores Virales/genética , Receptores Virales/metabolismo , Adulto , Anciano de 80 o más Años , Encéfalo/virología , Mapeo Encefálico , Preescolar , Bases de Datos Genéticas , Femenino , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/metabolismo , Humanos , Lactante , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Nectinas/genética , Nectinas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transcriptoma , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
7.
Biochim Biophys Acta Gen Subj ; 1861(10): 2455-2461, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28709864

RESUMEN

BACKGROUND: The human natural killer-1 (HNK-1) carbohydrate, a unique trisaccharide possessing sulfated glucuronic acid in a non-reducing terminus (HSO3-3GlcAß1-3Galß1-4GlcNAc-), is highly expressed in the nervous system and its spatiotemporal expression is strictly regulated. Mice deficient in the gene encoding a key enzyme, GlcAT-P, of the HNK-1 biosynthetic pathway exhibit almost complete disappearance of the HNK-1 epitope in the brain, significant reduction of long-term potentiation, and aberration of spatial learning and memory formation. In addition to its physiological roles in higher brain function, the HNK-1 carbohydrate has attracted considerable attention as an autoantigen associated with peripheral demyelinative neuropathy, which relates to IgM paraproteinemia, because of high immunogenicity. It has been suggested, however, that serum autoantibodies in IgM anti-myelin-associated glycoprotein (MAG) antibody-associated neuropathy patients show heterogeneous reactivity to the HNK-1 epitope. SCOPE OF REVIEW: We have found that structurally distinct HNK-1 epitopes are expressed in specific proteins in the nervous system. Here, we overview the current knowledge of the involvement of these HNK-1 epitopes in the regulation of neural plasticity and discuss the impact of different HNK-1 antigens of anti-MAG neuropathy patients. MAJOR CONCLUSIONS: We identified the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptor subunit GluA2 and aggrecan as HNK-1 carrier proteins. The HNK-1 epitope on GluA2 and aggrecan regulates neural plasticity in different ways. Furthermore, we found the clinical relationship between reactivity of autoantibodies to the different HNK-1 epitopes and progression of anti-MAG neuropathy. GENERAL SIGNIFICANCE: The HNK-1 epitope is indispensable for the acquisition of normal neuronal function and can be a good target for the establishment of diagnostic criteria for anti-MAG neuropathy.


Asunto(s)
Antígenos CD57/química , Epítopos/química , Glicoproteína Asociada a Mielina/inmunología , Plasticidad Neuronal , Paraproteinemias/inmunología , Enfermedades del Sistema Nervioso Periférico/inmunología , Agrecanos/metabolismo , Animales , Autoanticuerpos/biosíntesis , Antígenos CD57/genética , Antígenos CD57/inmunología , Epítopos/genética , Epítopos/inmunología , Glucuronosiltransferasa/deficiencia , Glucuronosiltransferasa/genética , Humanos , Inmunoglobulina M/biosíntesis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ratones , Ratones Noqueados , Glicoproteína Asociada a Mielina/genética , Paraproteinemias/genética , Paraproteinemias/patología , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Receptores AMPA/genética , Receptores AMPA/inmunología
8.
J Biol Chem ; 290(32): 19833-43, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26105052

RESUMEN

Varicella-zoster virus (VZV) is a member of the human Herpesvirus family that causes varicella (chicken pox) and zoster (shingles). VZV latently infects sensory ganglia and is also responsible for encephalomyelitis. Myelin-associated glycoprotein (MAG), a member of the sialic acid (SA)-binding immunoglobulin-like lectin family, is mainly expressed in neural tissues. VZV glycoprotein B (gB) associates with MAG and mediates membrane fusion during VZV entry into host cells. The SA requirements of MAG when associating with its ligands vary depending on the specific ligand, but it is unclear whether the SAs on gB are involved in the association with MAG. In this study, we found that SAs on gB are essential for the association with MAG as well as for membrane fusion during VZV infection. MAG with a point mutation in the SA-binding site did not bind to gB and did not mediate cell-cell fusion or VZV entry. Cell-cell fusion and VZV entry mediated by the gB-MAG interaction were blocked by sialidase treatment. N-glycosylation or O-glycosylation inhibitors also inhibited the fusion and entry mediated by gB-MAG interaction. Furthermore, gB with mutations in N-glycosylation sites, i.e. asparagine residues 557 and 686, did not associate with MAG, and the cell-cell fusion efficiency was low. Fusion between the viral envelope and cellular membrane is essential for host cell entry by herpesviruses. Therefore, these results suggest that SAs on gB play important roles in MAG-mediated VZV infection.


Asunto(s)
Células Epiteliales/metabolismo , Herpesvirus Humano 3/metabolismo , Glicoproteína Asociada a Mielina/metabolismo , Neuroglía/metabolismo , Polisacáridos/química , Ácidos Siálicos/química , Proteínas del Envoltorio Viral/química , Línea Celular Tumoral , Células Epiteliales/patología , Células Epiteliales/virología , Glicosilación , Células HEK293 , Herpesvirus Humano 3/química , Herpesvirus Humano 3/genética , Interacciones Huésped-Patógeno , Humanos , Fusión de Membrana , Glicoproteína Asociada a Mielina/química , Glicoproteína Asociada a Mielina/genética , Neuraminidasa/química , Neuraminidasa/genética , Neuraminidasa/metabolismo , Neuroglía/patología , Neuroglía/virología , Mutación Puntual , Polisacáridos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ácidos Siálicos/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
9.
Biochem Soc Trans ; 44(4): 1066-72, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528753

RESUMEN

STAR (signal transduction and activation of RNA) proteins regulate splicing of target genes that have roles in neural connectivity, survival and myelination in the vertebrate nervous system. These regulated splicing targets include mRNAs such as the Neurexins (Nrxn), SMN2 (survival of motor neuron) and MAG (myelin-associated glycoprotein). Recent work has made it possible to identify and validate STAR protein splicing targets in vivo by using genetically modified mouse models. In this review, we will discuss the importance of STAR protein splicing targets in the CNS (central nervous system).


Asunto(s)
Sistema Nervioso Central/metabolismo , Empalme del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Animales , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular Neuronal/genética , Humanos , Glicoproteína Asociada a Mielina/genética , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa , Filogenia , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/clasificación , Proteínas de Unión al ARN/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética
10.
Brain ; 138(Pt 9): 2521-36, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26179919

RESUMEN

Pelizaeus-Merzbacher disease is an X-linked hypomyelinating leukodystrophy caused by mutations or rearrangements in PLP1. It presents in infancy with nystagmus, jerky head movements, hypotonia and developmental delay evolving into spastic tetraplegia with optic atrophy and variable movement disorders. A clinically similar phenotype caused by recessive mutations in GJC2 is known as Pelizaeus-Merzbacher-like disease. Both genes encode proteins associated with myelin. We describe three siblings of a consanguineous family manifesting the typical infantile-onset Pelizaeus-Merzbacher disease-like phenotype slowly evolving into a form of complicated hereditary spastic paraplegia with mental retardation, dysarthria, optic atrophy and peripheral neuropathy in adulthood. Magnetic resonance imaging and spectroscopy were consistent with a demyelinating leukodystrophy. Using genetic linkage and exome sequencing, we identified a homozygous missense c.399C>G; p.S133R mutation in MAG. This gene, previously associated with hereditary spastic paraplegia, encodes myelin-associated glycoprotein, which is involved in myelin maintenance and glia-axon interaction. This mutation is predicted to destabilize the protein and affect its tertiary structure. Examination of the sural nerve biopsy sample obtained in childhood in the oldest sibling revealed complete absence of myelin-associated glycoprotein accompanied by ill-formed onion-bulb structures and a relatively thin myelin sheath of the affected axons. Immunofluorescence, cell surface labelling, biochemical analysis and mass spectrometry-based proteomics studies in a variety of cell types demonstrated a devastating effect of the mutation on post-translational processing, steady state expression and subcellular localization of myelin-associated glycoprotein. In contrast to the wild-type protein, the p.S133R mutant was retained in the endoplasmic reticulum and was subjected to endoplasmic reticulum-associated protein degradation by the proteasome. Our findings identify involvement of myelin-associated glycoprotein in this family with a disorder affecting the central and peripheral nervous system, and suggest that loss of the protein function is responsible for the unique clinical phenotype.


Asunto(s)
Mutación/genética , Glicoproteína Asociada a Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/genética , Adulto , Conexinas/genética , Análisis Mutacional de ADN , Retículo Endoplásmico/metabolismo , Salud de la Familia , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Masculino , Modelos Moleculares , Proteína Proteolipídica de la Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Transporte de Proteínas/genética , Proteómica , Proteínas S100/metabolismo , Nervio Sural/patología , Adulto Joven
11.
J Cell Sci ; 126(Pt 1): 209-20, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23132925

RESUMEN

In the injured adult mammalian central nervous system (CNS), products are generated that inhibit neuronal sprouting and regeneration. In recent years, most attention has focused on the myelin-associated inhibitory proteins (MAIs) Nogo-A, OMgp, and myelin-associated glycoprotein (MAG). Binding of MAIs to neuronal cell-surface receptors leads to activation of RhoA, growth cone collapse, and neurite outgrowth inhibition. In the present study, we identify low-density lipoprotein (LDL) receptor-related protein-1 (LRP1) as a high-affinity, endocytic receptor for MAG. In contrast with previously identified MAG receptors, binding of MAG to LRP1 occurs independently of terminal sialic acids. In primary neurons, functional inactivation of LRP1 with receptor-associated protein, depletion by RNA interference (RNAi) knock-down, or LRP1 gene deletion is sufficient to significantly reverse MAG and myelin-mediated inhibition of neurite outgrowth. Similar results are observed when LRP1 is antagonized in PC12 and N2a cells. By contrast, inhibiting LRP1 does not attenuate inhibition of neurite outgrowth caused by chondroitin sulfate proteoglycans. Mechanistic studies in N2a cells showed that LRP1 and p75NTR associate in a MAG-dependent manner and that MAG-mediated activation of RhoA may involve both LRP1 and p75NTR. LRP1 derivatives that include the complement-like repeat clusters CII and CIV bind MAG and other MAIs. When CII and CIV were expressed as Fc-fusion proteins, these proteins, purified full-length LRP1 and shed LRP1 all attenuated the inhibition of neurite outgrowth caused by MAG and CNS myelin in primary neurons. Collectively, our studies identify LRP1 as a novel MAG receptor that functions in neurite outgrowth inhibition.


Asunto(s)
Sistema Nervioso Central/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Vaina de Mielina/metabolismo , Glicoproteína Asociada a Mielina/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuritas/metabolismo , Animales , Células CHO , Células COS , Línea Celular , Cricetinae , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Espectrometría de Masas , Vaina de Mielina/genética , Glicoproteína Asociada a Mielina/genética , Proteínas del Tejido Nervioso , Células PC12 , Unión Proteica , Ratas , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
12.
J Neurosci Res ; 93(2): 285-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25327365

RESUMEN

The γ-aminobutyric acid (GABA) type B receptor has been implicated in glial cell development in the peripheral nervous system (PNS), although the exact function of GABA signaling is not known. To investigate GABA and its B receptor in PNS development and degeneration, we studied the expression of the GABAB receptor, GABA, and glutamic acid decarboxylase GAD65/67 in both development and injury in fetal dissociated dorsal root ganglia (DRG) cell cultures and in the rat sciatic nerve. We found that GABA, GAD65/67, and the GABAB receptor were expressed in premyelinating and nonmyelinating Schwann cells throughout development and after injury. A small population of myelinated sensory fibers displayed all of these molecules at the node of Ranvier, indicating a role in axon-glia communication. Functional studies using GABAB receptor agonists and antagonists were performed in fetal DRG primary cultures to study the function of this receptor during development. The results show that GABA, via its B receptor, is involved in the myelination process but not in Schwann cell proliferation. The data from adult nerves suggest additional roles in axon-glia communication after injury.


Asunto(s)
Vaina de Mielina/metabolismo , Nódulos de Ranvier/metabolismo , Receptores de GABA-B/metabolismo , Nervio Ciático , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos , GABAérgicos/farmacología , Ganglios Espinales/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-B/genética , Nervio Ciático/citología , Nervio Ciático/embriología , Nervio Ciático/crecimiento & desarrollo , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología
13.
RNA ; 19(7): 948-57, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23704325

RESUMEN

Myelin-associated glycoprotein (MAG) is a major component of myelin in the vertebrate central nervous system. MAG is present in the periaxonal region of the myelin structure, where it interacts with neuronal proteins to inhibit axon outgrowth and protect neurons from degeneration. Two alternatively spliced isoforms of Mag mRNA have been identified. The mRNA encoding the shorter isoform, known as S-MAG, contains a termination codon in exon 12, while the mRNA encoding the longer isoform, known as L-MAG, skips exon 12 and produces a protein with a longer C-terminal region. L-MAG is required in the central nervous system. How inclusion of Mag exon 12 is regulated is not clear. In a previous study, we showed that heteronuclear ribonucleoprotein A1 (hnRNP A1) contributes to Mag exon 12 skipping. Here, we show that hnRNP A1 interacts with an element that overlaps the 5' splice site of Mag exon 12. The element has a reduced ability to interact with the U1 snRNP compared with a mutant that improves the splice site consensus. An evolutionarily conserved secondary structure is present surrounding the element. The structure modulates interaction with both hnRNP A1 and U1. Analysis of splice isoforms produced from a series of reporter constructs demonstrates that the hnRNP A1-binding site and the secondary structure both contribute to exclusion of Mag exon 12.


Asunto(s)
Empalme Alternativo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Glicoproteína Asociada a Mielina/metabolismo , Conformación de Ácido Nucleico , ARN Mensajero/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Exones , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Secuencias Invertidas Repetidas , Ratones , Glicoproteína Asociada a Mielina/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , Sitios de Empalme de ARN , ARN Mensajero/genética , Empalmosomas/genética , Empalmosomas/metabolismo
14.
Exp Cell Res ; 322(1): 108-21, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24246222

RESUMEN

PACAP and its cognate peptide VIP participate in various biological functions, including myelin maturation and synthesis. However, defining whether these peptides affect peripheral expression of myelin proteins still remains unanswered. To address this issue, we assessed whether PACAP or VIP contribute to regulate the expression of three myelin proteins (MAG, MBP and MPZ, respectively) using the rat schwannoma cell line (RT4-P6D2T), a well-established model to study myelin gene expression. In addition, we endeavored to partly unravel the underlying molecular mechanisms involved. Expression of myelin-specific proteins was assessed in cells grown either in normal serum (10% FBS) or serum starved and treated with or without 100 nM PACAP or VIP. Furthermore, through pharmacological approach using the PACAP/VIP receptor antagonist (PACAP6-38) or specific pathway (MAPK or PI3K) inhibitors we defined the relative contribution of receptors and/or signaling pathways on the expression of myelin proteins. Our data show that serum starvation (24h) significantly increased both MAG, MBP and MPZ expression. Concurrently, we observed increased expression of endogenous PACAP and related receptors. Treatment with PACAP or VIP further exacerbated starvation-induced expression of myelin markers, suggesting that serum withdrawal might sensitize cells to peptide activity. Stimulation with either peptides increased phosphorylation of Akt at Ser473 residue but had no effect on phosphorylated Erk-1/2. PACAP6-38 (10 µM) impeded starvation- or peptide-induced expression of myelin markers. Similar effects were obtained after pretreatment with the PI3K inhibitor (wortmannin, 10 µM) but not the MAPKK inhibitor (PD98059, 50 µM). Together, the present finding corroborate the hypothesis that PACAP and VIP might contribute to the myelinating process preferentially via the canonical PI3K/Akt signaling pathway, providing the basis for future studies on the role of these peptides in demyelinating diseases.


Asunto(s)
Proteínas de la Mielina/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/fisiología , Células de Schwann/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología , Animales , Línea Celular Tumoral , Activación Enzimática/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Células de Schwann/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
J Neurosci ; 33(21): 8990-9002, 2013 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23699510

RESUMEN

The pathological mechanisms underlying neurological deficits observed in individuals born prematurely are not completely understood. A common form of injury in the preterm population is periventricular white matter injury (PWMI), a pathology associated with impaired brain development. To mitigate or eliminate PWMI, there is an urgent need to understand the pathological mechanism(s) involved on a neurobiological, structural, and functional level. Recent clinical data suggest that a percentage of premature infants experience relative hyperoxia. Using a hyperoxic model of premature brain injury, we have previously demonstrated that neonatal hyperoxia exposure in the mouse disrupts development of the white matter (WM) by delaying the maturation of the oligodendroglial lineage. In the present study, we address the question of how hyperoxia-induced alterations in WM development affect overall WM integrity and axonal function. We show that neonatal hyperoxia causes ultrastructural changes, including: myelination abnormalities (i.e., reduced myelin thickness and abnormal extramyelin loops) and axonopathy (i.e., altered neurofilament phosphorylation, paranodal defects, and changes in node of Ranvier number and structure). This disruption of axon-oligodendrocyte integrity results in the lasting impairment of conduction properties in the adult WM. Understanding the pathology of premature PWMI injury will allow for the development of interventional strategies to preserve WM integrity and function.


Asunto(s)
Axones/patología , Encéfalo/patología , Hiperoxia/patología , Fibras Nerviosas Mielínicas/patología , Oligodendroglía/patología , 2',3'-Nucleótido Cíclico Fosfodiesterasas/genética , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Potenciales de Acción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Axones/ultraestructura , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Transmisión , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/genética , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligodendroglía/ultraestructura
16.
J Biol Chem ; 288(47): 34009-34018, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24129569

RESUMEN

In addition to functioning as an activator of fibrinolysis, tissue-type plasminogen activator (tPA) interacts with neurons and regulates multiple aspects of neuronal cell physiology. In this study, we examined the mechanism by which tPA initiates cell signaling in PC12 and N2a neuron-like cells. We demonstrate that enzymatically active and inactive tPA (EI-tPA) activate ERK1/2 in a biphasic manner. Rapid ERK1/2 activation is dependent on LDL receptor-related protein-1 (LRP1). In the second phase, ERK1/2 is activated by tPA independently of LRP1. The length of the LRP1-dependent phase varied inversely with the tPA concentration. Rapid ERK1/2 activation in response to EI-tPA and activated α2-macroglobulin (α2M*) required the NMDA receptor and Trk receptors, which assemble with LRP1 into a single pathway. Assembly of this signaling system may have been facilitated by the bifunctional adapter protein, PSD-95, which associated with LRP1 selectively in cells treated with EI-tPA or α2M*. Myelin-associated glycoprotein binds to LRP1 with high affinity but failed to induce phosphorylation of TrkA or ERK1/2. Instead, myelin-associated glycoprotein recruited p75 neurotrophin receptor (p75NTR) into a complex with LRP1 and activated RhoA. p75NTR was not recruited by other LRP1 ligands, including EI-tPA and α2M*. Lactoferrin functioned as an LRP1 signaling antagonist, inhibiting Trk receptor phosphorylation and ERK1/2 activation in response to EI-tPA. These results demonstrate that LRP1-initiated cell signaling is ligand-dependent. Proteins that activate cell signaling by binding to LRP1 assemble different co-receptor systems. Ligand-specific co-receptor recruitment provides a mechanism by which one receptor, LRP1, may trigger different signaling responses.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Glicoproteína Asociada a Mielina/metabolismo , Receptores de LDL/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lactoferrina/genética , Lactoferrina/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Glicoproteína Asociada a Mielina/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Ratas , Receptores de Factores de Crecimiento , Receptores de LDL/genética , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Activador de Tejido Plasminógeno/genética , Proteínas Supresoras de Tumor/genética , alfa-Macroglobulinas/genética , alfa-Macroglobulinas/metabolismo
17.
Cereb Cortex ; 23(9): 2044-57, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22772651

RESUMEN

Oligodendrocyte genes and white matter tracts have been implicated in the pathophysiology of schizophrenia and may play an important etiopathogenic role in cognitive dysfunction in schizophrenia. The objective of the present study in 60 chronic schizophrenia patients individually matched to 60 healthy controls was to determine whether 1) white matter tract integrity influences cognitive performance, 2) oligodendrocyte gene variants influence white matter tract integrity and cognitive performance, and 3) effects of oligodendrocyte gene variants on cognitive performance are mediated via white matter tract integrity. We used the partial least-squares multivariate approach to ascertain relationships among oligodendrocyte gene variants, integrity of cortico-cortical and subcortico-cortical white matter tracts, and cognitive performance. Robust relationships among oligodendrocyte gene variants, white matter tract integrity, and cognitive performance were found in both patients and controls. We also showed that effects of gene variants on cognitive performance were mediated by the integrity of white matter tracts. Our results were strengthened by bioinformatic analyses of gene variant function. To our knowledge, this is the first study that has brought together these lines of investigation in the same population and highlights the importance of the oligodendrocyte/white matter pathway in schizophrenia, particularly as it pertains to cognitive function.


Asunto(s)
Corteza Cerebral/patología , Cognición , Esquizofrenia/genética , Esquizofrenia/patología , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Receptores ErbB/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Glicoproteína Asociada a Mielina/genética , Proteínas del Tejido Nervioso/genética , Neurregulina-1/genética , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía , Polimorfismo de Nucleótido Simple , Receptor ErbB-4 , Adulto Joven
18.
PLoS Genet ; 7(1): e1001269, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21253564

RESUMEN

In mice, Quaking (Qk) is required for myelin formation; in humans, it has been associated with psychiatric disease. QK regulates the stability, subcellular localization, and alternative splicing of several myelin-related transcripts, yet little is known about how QK governs these activities. Here, we show that QK enhances Hnrnpa1 mRNA stability by binding a conserved 3' UTR sequence with high affinity and specificity. A single nucleotide mutation in the binding site eliminates QK-dependent regulation, as does reduction of QK by RNAi. Analysis of exon expression across the transcriptome reveals that QK and hnRNP A1 regulate an overlapping subset of transcripts. Thus, a simple interpretation is that QK regulates a large set of oligodendrocyte precursor genes indirectly by increasing the intracellular concentration of hnRNP A1. Together, the data show that hnRNP A1 is an important QK target that contributes to its control of myelin gene expression.


Asunto(s)
Regiones no Traducidas 3' , Regulación de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Oligodendroglía/metabolismo , Proteínas de Unión al ARN/genética , Empalme Alternativo , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Secuencia Conservada , Exones , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Ratones , Glicoproteína Asociada a Mielina/genética , Oligodendroglía/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Estabilidad del ARN , ARN Interferente Pequeño/genética , Ratas , Alineación de Secuencia
19.
Dev Dyn ; 242(7): 847-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23592243

RESUMEN

BACKGROUND: The mammalian central nervous system is incapable of substantial axon regeneration after injury partially due to the presence of myelin-associated inhibitory molecules including Nogo-A and myelin associated glycoprotein (MAG). In contrast, axolotl salamanders are capable of considerable axon regrowth during spinal cord regeneration. RESULTS: Here, we show that Nogo-A and MAG, and their receptor, Nogo receptor (NgR), are present in the axolotl genome and are broadly expressed in the central nervous system (CNS) during development, adulthood, and importantly, during regeneration. Furthermore, we show that Nogo-A and NgR are co-expressed in Sox2 positive neural progenitor cells. CONCLUSIONS: These expression patterns suggest myelin-associated proteins are permissive for neural development and regeneration in axolotls.


Asunto(s)
Ambystoma mexicanum/metabolismo , Proteínas Anfibias/metabolismo , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Regeneración de la Medula Espinal/fisiología , Proteínas Anfibias/genética , Animales , Proteínas de la Mielina/genética , Glicoproteína Asociada a Mielina/genética , Proteínas Nogo , Traumatismos de la Médula Espinal/genética , Regeneración de la Medula Espinal/genética
20.
Genes (Basel) ; 15(9)2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39336794

RESUMEN

Background and objectives: Hereditary spastic paraplegia (HSP) is characterized by unsteady gait, motor incoordination, speech impairment, abnormal eye movement, progressive spasticity and lower limb weakness. Spastic paraplegia 75 (SPG75) results from a mutation in the gene that encodes myelin associated glycoprotein (MAG). Only a limited number of MAG variants associated with SPG75 in families of European, Middle Eastern, North African, Turkish and Palestinian ancestry have been documented so far. This study aims to provide further insight into the clinical and molecular manifestations of HSP. Methods: Using whole-exome sequencing, we investigated a consanguineous Pakistani family where three individuals presented with clinical signs of HSP. Sanger sequencing was used to carry out segregation analysis on available family members, and a minigene splicing assay was utilized to evaluate the effect of the splicing variant. Results: We identified a novel homozygous pathogenic splice donor variant in MAG (c.46 + 1G > T) associated with SPG75. RNA analysis revealed exon skipping that resulted in the loss of a start codon for ENST00000361922.8 isoform. Affected individuals exhibited variable combinations of nystagmus, developmental delay, cognitive impairments, spasticity, dysarthria, delayed gait and ataxia. The proband displayed a quadrupedal stride, and his siblings experienced frequent falls and ataxic gait as one of the prominent features that have not been previously reported in SPG75. Conclusions: Thus, the present study presents an uncommon manifestation of SPG75, the first from the Pakistani population, and broadens the spectrum of MAG variants.


Asunto(s)
Consanguinidad , Glicoproteína Asociada a Mielina , Linaje , Paraplejía Espástica Hereditaria , Humanos , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/patología , Masculino , Pakistán , Femenino , Glicoproteína Asociada a Mielina/genética , Mutación , Niño , Adulto , Secuenciación del Exoma , Homocigoto
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda