Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Int J Biol Macromol ; 258(Pt 1): 128780, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38104688

RESUMEN

This review is an effort towards the development of substrate reduction therapy using cerebroside sulfotransferase (CST) as a target protein for the development of inhibitors intended to treat pathophysiological condition resulting from the accumulation of sulfatide, a product from the catalytic action of CST. Accumulation of sulfatides leads to progressive impairment and destruction of the myelin structure, disruption of normal physiological transmission of electrical impulse between nerve cells, axonal loss in the central and peripheral nervous system and cumulatively gives a clinical manifestation of metachromatic leukodystrophy. Thus, there is a need to develop specific and potent CST inhibitors to positively control sulfatide accumulation. Structural similarity and computational studies revealed that LYS85, SER172 and HIS141 are key catalytic residues that determine the catalytic action of CST through the transfer of sulfuryl group from the donor PAPS to the acceptor galactosylceramide. Computational studies revealed catalytic site of CST consists two binding site pocket including PAPS binding pocket and substrate binding pocket. Specific substrate site residues in CST can be targeted to develop specific CST inhibitors. This review also explores the challenges of CST-directed substrate reduction therapy as well as the opportunities available in natural products for inhibitor development.


Asunto(s)
Leucodistrofia Metacromática , Sulfotransferasas , Humanos , Leucodistrofia Metacromática/metabolismo , Sulfoglicoesfingolípidos , Vaina de Mielina/metabolismo , Neuronas/metabolismo
2.
Neurotherapeutics ; 20(5): 1347-1368, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37525026

RESUMEN

Metachromatic leukodystrophy (MLD) is a severe demyelinating, autosomal recessive genetic leukodystrophy. The disease is underpinned by mutations in the arylsulfatase A gene (ARSA), resulting in deficient activity of the arylsulfatase A lysosomal enzyme and consequential accumulation of galactosylceramide-3-O-sulfate (sulfatide) in the brain. Using an ex vivo murine-derived organotypic cerebellar slice culture model, we demonstrate that sulfatide induces demyelination in a concentration-dependent manner. Interestingly, our novel data demonstrate that sulfatide-induced demyelination is underpinned by PARP-1 activation, oligodendrocyte loss, pro-inflammatory cytokine expression, astrogliosis, and microgliosis. Moreover, such sulfatide-induced effects can be attenuated by the treatment with the poly (ADP-ribose) polymerase 1 (PARP-1) inhibitor Olaparib (IC50∼100 nM) suggesting that this small molecule may be neuroprotective and limit toxin-induced demyelination. Our data support the idea that sulfatide is a key driver of demyelination and neuroinflammation in MLD and suggest that PARP-1 inhibitors have therapeutic utility in the sphere of rare demyelinating disease.


Asunto(s)
Enfermedades Desmielinizantes , Leucodistrofia Metacromática , Animales , Ratones , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Cerebrósido Sulfatasa/genética , Cerebrósido Sulfatasa/metabolismo , Sulfoglicoesfingolípidos/metabolismo , Enfermedades Neuroinflamatorias , Inhibidores de Poli(ADP-Ribosa) Polimerasas
3.
Int J Mol Sci ; 24(11)2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37298156

RESUMEN

Metachromatic leukodystrophy (MLD) is a hereditary neurodegenerative disease characterized by demyelination and motor and cognitive impairments due to deficiencies of the lysosomal enzyme arylsulfatase A (ARSA) or the saposin B activator protein (SapB). Current treatments are limited; however, gene therapy using adeno-associated virus (AAV) vectors for ARSA delivery has shown promising results. The main challenges for MLD gene therapy include optimizing the AAV dosage, selecting the most effective serotype, and determining the best route of administration for ARSA delivery into the central nervous system. This study aims to evaluate the safety and efficacy of AAV serotype 9 encoding ARSA (AAV9-ARSA) gene therapy when administered intravenously or intrathecally in minipigs, a large animal model with anatomical and physiological similarities to humans. By comparing these two administration methods, this study contributes to the understanding of how to improve the effectiveness of MLD gene therapy and offers valuable insights for future clinical applications.


Asunto(s)
Leucodistrofia Metacromática , Enfermedades Neurodegenerativas , Humanos , Animales , Porcinos , Cerebrósido Sulfatasa/genética , Cerebrósido Sulfatasa/metabolismo , Porcinos Enanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Sistema Nervioso Central/metabolismo , Esterasas
4.
Neuroimage Clin ; 37: 103296, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36563646

RESUMEN

BACKGROUND AND PURPOSE: Metachromatic leukodystrophy (MLD) is a lysosomal enzyme deficiency disorder leading to demyelination and subsequently to a progressive decline in cognitive and motor function. It affects mainly white matter where changes during the course of the disease can be visualized on T2-weighted MRI as hyperintense areas. Associated changes in brain metabolism can be quantified by MR spectroscopy (MRS) and may give complementary information as biomarkers for disease characterisation and progression. Our study aimed to further investigate the correlation of MRS with clinical parameters for motor and cognitive function by using a model free MRS analysis approach that would be precise and straightforward to implement. MATERIALS AND METHODS: 53 MRS datasets derived from 29 patients (10 late-infantile, 19 juvenile) and 12 controls were acquired using a semi-LASER CSI sequence covering a slice through the centrum semiovale above the corpus callosum. We defined four regions of interest in the white matter (frontal white matter [FWM] and the cortico-spinal tract [CST] area, each left and right) and one in cortical grey matter. Spectra were analysed using a model and fitting free approach by calculating the definite integral of 10 intervals which were distributed along the whole spectrum. These 10 intervals were orientated towards the main peaks of the metabolites N-acetylaspartate (NAA), creatine, myo-inositol, choline, glutamine/glutamate and aspartate to approximately attribute changes in the intervals to corresponding metabolites. Their ratios to the main creatine peak integral were correlated with clinical parameters assessing motor and cognitive abilities. Furthermore, in a post-hoc analysis, NAA levels of a subset of 21 MR datasets were correlated to NAA levels in urine measured by 1H (proton) nuclear magnetic resonance (NMR) spectroscopy. The applied interval integration method was validated in the control cohort against the standard approach, using spectral profile templates of known metabolites (LCModel). Both methods showed good agreement, with coefficients of variance being slightly lower for our approach compared to the related LCModel results. Moreover, the new approach was able to extract information out of the frequency range around the main peaks of aspartate and glutamine where LCModel showed only few usable values for the respective metabolites. RESULTS: MLD spectra clearly differed from controls. The most pronounced differences were found in white matter (much less in grey matter), with larger values corresponding to main peaks of myo-inositol, choline and aspartate, and smaller values associated with NAA and glutamine. Late-infantile patients had more severe changes compared to later-onset patients, especially in intervals corresponding to NAA, aspartate, myo-inositol, choline and glutamine. There was a high correlation of several intervals in the corticospinal tract region with motor function (with the most relevant interval corresponding to NAA peak with a correlation coefficient of -0.75; p < 0.001), while cognitive function, by means of IQ, was found to be most correlating in frontal white matter corresponding to the NAA peak (r = 0.84, p < 0.001). The post-hoc analysis showed that the main NAA peak interval correlated negatively with the NAA in urine (r = -0.584, p < 0.001). CONCLUSION: The applied model and fitting free interval integration approach to analyse MRS data of a semi-LASER sequence at 3T suits well to detect and quantify pathological changes in MLD patients through the different courses of the disease and correlates well with clinical symptoms while showing smaller dimensions of variation compared to the more sophisticated single metabolite analysis using LCModel. NAA seems the most clinically meaningful biomarker to use in this context. Its correlation with urine measurements further underlines its potential as a clinically and biologically useful parameter of disease progression in MLD.


Asunto(s)
Glutamina , Leucodistrofia Metacromática , Humanos , Glutamina/metabolismo , Creatina/metabolismo , Leucodistrofia Metacromática/diagnóstico por imagen , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Ácido Aspártico , Espectroscopía de Resonancia Magnética/métodos , Encéfalo/patología , Colina/metabolismo , Inositol/metabolismo
5.
Open Biol ; 12(7): 210371, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35857900

RESUMEN

Metachromatic leukodystrophy is a neurological lysosomal deposit disease that affects public health despite its low incidence in the population. Currently, few reports are available on pathophysiological events related to enzyme deficiencies and subsequent sulfatide accumulation. This research aims to examine the use of metformin as an alternative treatment to counteract these effects. This was evaluated in human Schwann cells (HSCs) transfected or non-transfected with CRISPR-Cas9, and later treated with sulfatides and metformin. This resulted in transfected HSCs showing a significant increase in cell reactive oxygen species (ROS) production when exposed to 100 µM sulfatides (p = 0.0007), compared to non-transfected HSCs. Sulfatides at concentrations of 10 to 100 µM affected mitochondrial bioenergetics in transfected HSCs. Moreover, these analyses showed that transfected cells showed a decrease in basal and maximal respiration rates after exposure to 100 µM sulfatide. However, maximal and normal mitochondrial respiratory capacity decreased in cells treated with both sulfatide and metformin. This study has provided valuable insights into bioenergetic and mitochondrial effects of sulfatides in HSCs for the first time. Treatment with metformin (500 µM) restored the metabolic activity of these cells and decreased ROS production.


Asunto(s)
Leucodistrofia Metacromática , Metformina , Sistemas CRISPR-Cas , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Metformina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células de Schwann/metabolismo , Sulfoglicoesfingolípidos/metabolismo
6.
Lipids Health Dis ; 21(1): 32, 2022 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-35351138

RESUMEN

BACKGROUND: Recent findings show that extracellular vesicle constituents can exert short- and long-range biological effects on neighboring cells in the brain, opening an exciting avenue for investigation in the field of neurodegenerative diseases. Although it is well documented that extracellular vesicles contain many lipids and are enriched in sphingomyelin, cholesterol, phosphatidylserines and phosphatidylinositols, no reports have addressed the lipidomic profile of brain derived EVs in the context of Metachromatic Leukodystrophy, a lysosomal storage disease with established metabolic alterations in sulfatides. METHODS: In this study, we isolated and characterized the lipid content of brain-derived EVs using the arylsulfatase A knockout mouse as a model of the human condition. RESULTS: Our results suggest that biogenesis of brain-derived EVs is a tightly regulated process in terms of size and protein concentration during postnatal life. Our lipidomic analysis demonstrated that sulfatides and their precursors (ceramides) as well as other lipids including fatty acids are altered in an age-dependent manner in EVs isolated from the brain of the knockout mouse. CONCLUSIONS: In addition to the possible involvement of EVs in the pathology of Metachromatic Leukodystrophy, our study underlines that measuring lipid signatures in EVs may be useful as biomarkers of disease, with potential application to other genetic lipidoses.


Asunto(s)
Vesículas Extracelulares , Leucodistrofia Metacromática , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Lipidómica , Ratones
7.
Sci Rep ; 11(1): 14486, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34262084

RESUMEN

Krabbe disease (KD) and metachromatic leukodystrophy (MLD) are caused by accumulation of the glycolipids galactosylceramide (GalCer) and sulfatide and their toxic metabolites psychosine and lysosulfatide, respectively. We discovered a potent and selective small molecule inhibitor (S202) of ceramide galactosyltransferase (CGT), the key enzyme for GalCer biosynthesis, and characterized its use as substrate reduction therapy (SRT). Treating a KD mouse model with S202 dose-dependently reduced GalCer and psychosine in the central (CNS) and peripheral (PNS) nervous systems and significantly increased lifespan. Similarly, treating an MLD mouse model decreased sulfatides and lysosulfatide levels. Interestingly, lower doses of S202 partially inhibited CGT and selectively reduced synthesis of non-hydroxylated forms of GalCer and sulfatide, which appear to be the primary source of psychosine and lysosulfatide. Higher doses of S202 more completely inhibited CGT and reduced the levels of both non-hydroxylated and hydroxylated forms of GalCer and sulfatide. Despite the significant benefits observed in murine models of KD and MLD, chronic CGT inhibition negatively impacted both the CNS and PNS of wild-type mice. Therefore, further studies are necessary to elucidate the full therapeutic potential of CGT inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Leucodistrofia de Células Globoides/tratamiento farmacológico , Leucodistrofia Metacromática/tratamiento farmacológico , N-Acilesfingosina Galactosiltransferasa/antagonistas & inhibidores , N-Acilesfingosina Galactosiltransferasa/metabolismo , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Galactosilceramidas/metabolismo , Balactosiltransferasa de Gangliósidos/genética , Balactosiltransferasa de Gangliósidos/metabolismo , Humanos , Leucodistrofia de Células Globoides/mortalidad , Leucodistrofia Metacromática/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Psicosina/análogos & derivados , Psicosina/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfotransferasas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
8.
J Mol Neurosci ; 71(5): 1112-1130, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33185815

RESUMEN

Metachromatic leukodystrophy (MLD) is a neurodegenerative disorder characterized by progressive demyelination due to deficiency of the enzyme arylsulfatase A (ARSA) in leukocytes, and consequently leads to impaired degradation and accumulation of cerebroside-3-sulfate (sulfatide). This study aimed to sequence the ARSA gene in a total of 43 patients with metachromatic leukodystrophy descendant from 40 Egyptian families. In addition, four carrier parents from two families with children who had died from MLD came to the clinic for genetic analysis. Prenatal diagnosis was performed for four families with molecularly diagnosed MLD sibs. Different mutations were characterized in our cohort, including missense, nonsense, splice, and deletion. Overall, 21 different mutations in the ARSA gene were detected, with 12 novel mutations, i.e. p.Arg60Pro, p.Tyr65*, p.Val112Asp, p.Arg116*, p.Gly124Asp, p.Pro193Ser, p.Gln238*, p.Gln456*, p.Thr276Lys, and p.Gly311Arg, in addition to two new acceptor splice-site mutations 685-1G > A and c.954_956 delCTT. The amniotic fluid samples revealed two carrier fetuses with heterozygous monoallelic mutations, and two affected fetuses had the homozygous biallelic mutations. In conclusion, the current study sheds light on the underlying ARSA gene defect, with an expansion of the mutation spectrum. To our knowledge, this is the first molecular study of MLD among the Egyptian population.


Asunto(s)
Cerebrósido Sulfatasa/genética , Leucodistrofia Metacromática/genética , Fenotipo , Niño , Preescolar , Humanos , Lactante , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Mutación
9.
J Child Neurol ; 36(2): 148-151, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32991243

RESUMEN

Phelan-McDermid syndrome or 22q13.3 deletion syndrome is a rare neurodevelopmental disorder characterized by neonatal hypotonia, severe speech delay, moderate to profound intellectual disability, and minor dysmorphic features. Regression of developmental milestones is often recognized as characteristic of this syndrome. We report a 6-year-old patient with Phelan-McDermid syndrome who presented with rapid neurologic deterioration secondary to metachromatic leukodystrophy due to a mutation of the arylsulfatase A gene (ARSA) on the other allele of 22q13.3. Metachromatic leukodystrophy was diagnosed later after clinical deterioration. Currently, there are no guidelines for screening Phelan-McDermid syndrome patients for metachromatic leukodystrophy. We propose screening for urine sulfatides at the time of Phelan-McDermid syndrome diagnosis to identify patients with pre-symptomatic or early symptomatic metachromatic leukodystrophy as it is important to facilitate discussion of treatment options and prognosis and provide medical surveillance for associated complications.


Asunto(s)
Trastornos de los Cromosomas/complicaciones , Leucodistrofia Metacromática/complicaciones , Leucodistrofia Metacromática/diagnóstico , Arilsulfatasas/sangre , Encéfalo/diagnóstico por imagen , Niño , Deleción Cromosómica , Cromosomas Humanos Par 22 , Diagnóstico Diferencial , Femenino , Humanos , Leucodistrofia Metacromática/metabolismo , Espectroscopía de Resonancia Magnética , Sulfoglicoesfingolípidos/orina
10.
Glia ; 67(6): 1196-1209, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30980503

RESUMEN

X-linked adrenoleukodystrophy (X-ALD) and metachromatic leukodystrophy (MLD) are two relatively common examples of hereditary demyelinating diseases caused by a dysfunction of peroxisomal or lysosomal lipid degradation. In both conditions, accumulation of nondegraded lipids leads to the destruction of cerebral white matter. Because of their high lipid content, oligodendrocytes are considered key to the pathophysiology of these leukodystrophies. However, the response to allogeneic stem cell transplantation points to the relevance of cells related to the hematopoietic lineage. In the present study, we aimed to better characterize the pathogenetic role of microglia in the above-mentioned diseases. Applying recently established microglia markers to human autopsy cases of X-ALD and MLD we were able to delineate distinct lesion stages in evolving demyelinating lesions. The immune-phenotype of microglia was altered already early in lesion evolution, and microglia loss preceded full-blown myelin degeneration both in X-ALD and MLD. DNA fragmentation indicating phagocyte death was observed in areas showing microglia loss. The morphology and dynamics of phagocyte decay differed between the diseases and between lesion stages, hinting at distinct pathways of programmed cell death. In summary, the present study shows an early and severe damage to microglia in the pathogenesis of X-ALD and MLD. This hints at a central pathophysiologic role of these cells in the diseases and provides evidence for an ongoing transfer of toxic substrates primarily enriched in myelinating cells to microglia.


Asunto(s)
Adrenoleucodistrofia/patología , Leucodistrofia Metacromática/patología , Microglía/patología , Vaina de Mielina/patología , Adolescente , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/metabolismo , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Masculino , Microglía/metabolismo , Persona de Mediana Edad , Vaina de Mielina/genética , Vaina de Mielina/metabolismo
11.
J Neurol Neurosurg Psychiatry ; 89(1): 105-111, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28889092

RESUMEN

OBJECTIVE: To determine whether proton magnetic resonance spectroscopic imaging is useful in predicting clinical course of patients with metachromatic leukodystrophy (MLD), an inherited white matter disorder treatable with haematopoietic cell transplantation (HCT). METHODS: 21 patients with juvenile or adult MLD (12 HCT-treated) were compared with 16 controls in the same age range. Clinical outcome was determined as good, moderate or poor. Metabolites were quantified in white matter, and significance of metabolite concentrations at baseline for outcome prediction was assessed using logistic regression analysis. Evolution of metabolic changes was assessed for patients with follow-up examinations. RESULTS: In this retrospective study, 16 patients with baseline scans were included, 5 with good, 3 with moderate and 8 with poor outcome, and 16 controls. We observed significant group differences for all metabolite concentrations in white matter (p<0.001). Compared with controls, patients had decreased N-acetylaspartate and glutamate, and increased myo-inositol and lactate, most pronounced in patients with poor outcome (post hoc, all p<0.05). Logistic regression showed complete separation of data. Creatine could distinguish poor from moderate and good outcome, the sum of glutamate and glutamine could distinguish good from moderate and poor outcome, and N-acetylaspartate could distinguish all outcome groups. For 13 patients (8 with baseline scans), one or more follow-up examinations were evaluated, revealing stabilisation or even partial normalisation of metabolites in patients with moderate and good outcome, clearly visible in the ratio of choline/N-acetylaspartate. CONCLUSION: In MLD, quantitative spectroscopic imaging at baseline is predictive for outcome and aids in determining eligibility for HCT.


Asunto(s)
Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Espectroscopía de Resonancia Magnética , Adolescente , Adulto , Ácido Aspártico/análogos & derivados , Niño , Preescolar , Creatina/metabolismo , Femenino , Ácido Glutámico/metabolismo , Humanos , Leucodistrofia Metacromática/terapia , Masculino , Pronóstico , Estudios Retrospectivos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
12.
Hum Mol Genet ; 26(22): 4506-4518, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28973395

RESUMEN

Hypomyelinating leukodystrophies are heritable disorders defined by lack of development of brain myelin, but the cellular mechanisms of hypomyelination are often poorly understood. Mutations in TUBB4A, encoding the tubulin isoform tubulin beta class IVA (Tubb4a), result in the symptom complex of hypomyelination with atrophy of basal ganglia and cerebellum (H-ABC). Additionally, TUBB4A mutations are known to result in a broad phenotypic spectrum, ranging from primary dystonia (DYT4), isolated hypomyelination with spastic quadriplegia, and an infantile onset encephalopathy, suggesting multiple cell types may be involved. We present a study of the cellular effects of TUBB4A mutations responsible for H-ABC (p.Asp249Asn), DYT4 (p.Arg2Gly), a severe combined phenotype with hypomyelination and encephalopathy (p.Asn414Lys), as well as milder phenotypes causing isolated hypomyelination (p.Val255Ile and p.Arg282Pro). We used a combination of histopathological, biochemical and cellular approaches to determine how these different mutations may have variable cellular effects in neurons and/or oligodendrocytes. Our results demonstrate that specific mutations lead to either purely neuronal, combined neuronal and oligodendrocytic or purely oligodendrocytic defects that closely match their respective clinical phenotypes. Thus, the DYT4 mutation that leads to phenotypes attributable to neuronal dysfunction results in altered neuronal morphology, but with unchanged tubulin quantity and polymerization, with normal oligodendrocyte morphology and myelin gene expression. Conversely, mutations associated with isolated hypomyelination (p.Val255Ile and p.Arg282Pro) and the severe combined phenotype (p.Asn414Lys) resulted in normal neuronal morphology but were associated with altered oligodendrocyte morphology, myelin gene expression, and microtubule dysfunction. The H-ABC mutation (p.Asp249Asn) that exhibits a combined neuronal and myelin phenotype had overlapping cellular defects involving both neuronal and oligodendrocyte cell types in vitro. Only mutations causing hypomyelination phenotypes showed altered microtubule dynamics and acted through a dominant toxic gain of function mechanism. The DYT4 mutation had no impact on microtubule dynamics suggesting a distinct mechanism of action. In summary, the different clinical phenotypes associated with TUBB4A reflect the selective and specific cellular effects of the causative mutations. Cellular specificity of disease pathogenesis is relevant to developing targeted treatments for this disabling condition.


Asunto(s)
Neuronas/patología , Oligodendroglía/patología , Tubulina (Proteína)/genética , Adolescente , Adulto , Atrofia/patología , Ganglios Basales/patología , Encéfalo/metabolismo , Encéfalo/patología , Catarata/congénito , Catarata/genética , Catarata/metabolismo , Catarata/patología , Cerebelo/patología , Niño , Preescolar , Femenino , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Imagen por Resonancia Magnética , Masculino , Microtúbulos/patología , Persona de Mediana Edad , Mutación , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Fenotipo , Tubulina (Proteína)/metabolismo , Adulto Joven
13.
J Control Release ; 253: 1-10, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28215668

RESUMEN

The lysosomal storage disorder (LSD) metachromatic leukodystrophy (MLD) is caused by a deficiency of the soluble, lysosomal hydrolase arylsulfatase A (ASA). The disease is characterized by accumulation of 3-O-sulfogalactosylceramide (sulfatide), progressive demyelination of the nervous system and premature death. Enzyme replacement therapy (ERT), based on regular intravenous injections of recombinant functional enzyme, is in clinical use for several LSDs. For MLD and other LSDs with central nervous system (CNS) involvement, however, ERT is limited by the blood-brain barrier (BBB) restricting transport of therapeutic enzymes from the blood to the brain. In the present study, the potential of different types of surfactant-coated biodegradable nanoparticles to increase brain delivery of ASA was evaluated. Three different strategies to bind ASA to nanoparticle surfaces were compared: (1) adsorption, (2) high-affinity binding via the streptavidin-biotin system, and (3) covalent binding. Adsorption allowed binding of high amounts of active ASA. However, in presence of phosphate-buffered saline or serum rapid and complete desorption occurred, rendering this strategy ineffective for in vivo applications. In contrast, stable immobilization with negligible dissociation was achieved by high-affinity and covalent binding. Consequently, we analyzed the brain targeting of two stably nanoparticle-bound ASA formulations in ASA-/- mice, an animal model of MLD. Compared to free ASA, injected as a control, the biodistribution of nanoparticle-bound ASA was altered in peripheral organs, but no increase of brain levels was detectable. The failure to improve brain delivery suggests that the ASA glycoprotein interferes with processes required to target surfactant-coated nanoparticles to brain capillary endothelial cells.


Asunto(s)
Encéfalo/metabolismo , Cerebrósido Sulfatasa/administración & dosificación , Nanopartículas/administración & dosificación , Tensoactivos/administración & dosificación , Animales , Avidina/química , Biotinilación , Cerebrósido Sulfatasa/química , Cerebrósido Sulfatasa/genética , Cerebrósido Sulfatasa/farmacocinética , Femenino , Ácido Láctico/química , Leucodistrofia Metacromática/tratamiento farmacológico , Leucodistrofia Metacromática/metabolismo , Ratones Noqueados , Nanopartículas/química , Poloxámero/administración & dosificación , Poloxámero/química , Poloxámero/farmacocinética , Poliésteres/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polisorbatos/administración & dosificación , Polisorbatos/química , Polisorbatos/farmacocinética , Albúmina Sérica Humana/química , Tensoactivos/química , Tensoactivos/farmacocinética
15.
Nat Commun ; 6: 8966, 2015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26608817

RESUMEN

Glia play critical roles in maintaining the structure and function of the nervous system; however, the specific contribution that astroglia make to neurodegeneration in human disease states remains largely undefined. Here we use Alexander disease, a serious degenerative neurological disorder caused by astrocyte dysfunction, to identify glial-derived NO as a signalling molecule triggering astrocyte-mediated neuronal degeneration. We further find that NO acts through cGMP signalling in neurons to promote cell death. Glial cells themselves also degenerate, via the DNA damage response and p53. Our findings thus define a specific mechanism for glial-induced non-cell autonomous neuronal cell death, and identify a potential therapeutic target for reducing cellular toxicity in Alexander disease, and possibly other neurodegenerative disorders with glial dysfunction.


Asunto(s)
Enfermedad de Alexander/metabolismo , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Adolescente , Adulto , Enfermedad de Alexander/genética , Enfermedad de Alexander/patología , Animales , Astrocitos/patología , Western Blotting , Muerte Celular , Niño , Preescolar , Modelos Animales de Enfermedad , Drosophila , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Lactante , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/patología , Organismos Modificados Genéticamente , Estrés Oxidativo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
16.
Mol Ther ; 23(9): 1519-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26061647

RESUMEN

Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disorder resulting from a functional deficiency of arylsulfatase A (ARSA), an enzyme that catalyzes desulfation of 3-O-sulfogalactosylceramide (sulfatide). Lack of active ARSA leads to the accumulation of sulfatide in oligodendrocytes, Schwann cells and some neurons and triggers progressive demyelination, the neuropathological hallmark of MLD. Several therapeutic approaches have been explored, including enzyme replacement, autologous hematopoietic stem cell-based gene therapy, intracerebral gene therapy or cell-based gene delivery into the central nervous system (CNS). However, long-term treatment of the blood-brain-barrier protected CNS remains challenging. Here we used MLD patient-derived induced pluripotent stem cells (iPSCs) to generate long-term self-renewing neuroepithelial stem cells and astroglial progenitors for cell-based ARSA replacement. Following transplantation of ARSA-overexpressing precursors into ARSA-deficient mice we observed a significant reduction of sulfatide storage up to a distance of 300 µm from grafted cells. Our data indicate that neural precursors generated via reprogramming from MLD patients can be engineered to ameliorate sulfatide accumulation and may thus serve as autologous cell-based vehicle for continuous ARSA supply in MLD-affected brain tissue.


Asunto(s)
Sistema Nervioso Central/metabolismo , Cerebrósido Sulfatasa/genética , Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Sulfoglicoesfingolípidos/metabolismo , Animales , Axones/metabolismo , Encéfalo/metabolismo , Diferenciación Celular , Supervivencia Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Cerebrósido Sulfatasa/metabolismo , Proteínas de Unión al ADN/deficiencia , Modelos Animales de Enfermedad , Orden Génico , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Lentivirus/genética , Ratones , Ratones Noqueados , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Transducción Genética
17.
Mol Genet Metab ; 115(1): 48-52, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25796965

RESUMEN

Metachromatic Leukodystrophy (MLD; MIM# 250100) is a rare inherited lysosomal storage disorder caused by the deficiency of Arylsulfatase A (ARSA). The enzymatic defect results in the accumulation of the ARSA substrate that is particularly relevant in myelin forming cells and leads to progressive dysmyelination and dysfunction of the central and peripheral nervous system. Sulfatide accumulation has also been reported in various visceral organs, although little is known about the potential clinical consequences of such accumulation. Different forms of MLD-associated gallbladder disease have been described, and there is one reported case of an MLD patient presenting with functional consequences of sulfatide accumulation in the kidney. Here we describe a wide cohort of MLD patients in whom a tendency to sub-clinical metabolic acidosis was observed. Furthermore in some of them we report episodes of metabolic acidosis of different grades of severity developed in acute clinical conditions of various origin. Importantly, we finally show how a careful acid-base balance monitoring and prompt correction of imbalances might prevent severe consequences of acidosis.


Asunto(s)
Acidosis/complicaciones , Leucodistrofia Metacromática/complicaciones , Leucodistrofia Metacromática/metabolismo , Monitoreo Fisiológico , Equilibrio Ácido-Base , Desequilibrio Ácido-Base , Acidosis/sangre , Acidosis/prevención & control , Acidosis/orina , Niño , Preescolar , Estudios de Cohortes , Estudios de Seguimiento , Genotipo , Humanos , Lactante , Estudios Retrospectivos , Factores de Tiempo
18.
Biochim Biophys Acta ; 1851(8): 1026-39, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25701718

RESUMEN

The rapid development of analytical technology has made lipidomics an exciting new area and this review will focus more on modern approaches to lipidomics than on earlier technology. Although not fully comprehensive for all possible brain lipids, the intent is to at least provide a reference for the analysis of classes of lipids found in brain and nervous tissue. We will discuss problems posed by the brain because of its structural and functional heterogeneity, the development changes it undergoes (myelination, aging, pathology etc.) and its cellular heterogeneity (neurons, glia etc.). Section 2 will discuss the various ways in which brain tissue can be extracted to yield lipids for analysis and section 3 will cover a wide range of techniques used to analyze brain lipids such as chromatography and mass-spectrometry. In Section 4 we will discuss ways of analyzing some of the specific biologically active brain lipids found in very small amounts except in pathological conditions and section 5 looks to the future of experimental lipidomic modification in the brain. This article is part of a Special Issue entitled Brain Lipids.


Asunto(s)
Encefalopatías Metabólicas/metabolismo , Ácidos Grasos/análisis , Glucolípidos/análisis , Leucodistrofia Metacromática/metabolismo , Esclerosis Múltiple/metabolismo , Esfingolípidos/análisis , Animales , Encéfalo/metabolismo , Encéfalo/patología , Química Encefálica , Encefalopatías Metabólicas/patología , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Ácidos Grasos/química , Glucolípidos/química , Humanos , Leucodistrofia Metacromática/patología , Esclerosis Múltiple/patología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingolípidos/química
19.
J Biol Chem ; 290(11): 7040-53, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25605750

RESUMEN

The membrane-bound receptor for platelet-derived growth factor A (PDGFRα) is crucial for controlling the production of oligodendrocytes (OLs) for myelination, but regulation of its activity during OL differentiation is largely unknown. We have examined the effect of increased sulfated content of galactosylceramides (sulfatides) on the regulation of PDGFRα in multipotential neural precursors (NPs) that are deficient in arylsulfatase A (ASA) activity. This enzyme is responsible for the lysosomal hydrolysis of sulfatides. We show that sulfatide accumulation significantly impacts the formation of OLs via deregulation of PDGFRα function. PDGFRα is less associated with detergent-resistant membranes in ASA-deficient cells and showed a significant decrease in AKT phosphorylation. Rescue experiments with ASA showed a normalization of the ratio of long versus short sulfatides, restored PDGFRα levels, corrected its localization to detergent-resistant membranes, increased AKT phosphorylation, and normalized the production of OLs in ASA-deficient NPs. Moreover, our studies identified a novel mechanism that regulates the secretion of PDGFRα in NPs, in glial cells, and in the brain cortex via exosomal shedding. Our study provides a first step in understanding the role of sulfatides in regulating PDGFRα levels in OLs and its impact in myelination.


Asunto(s)
Cerebrósido Sulfatasa/genética , Ácidos Grasos/metabolismo , Leucodistrofia Metacromática/patología , Células-Madre Neurales/patología , Oligodendroglía/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sulfoglicoesfingolípidos/metabolismo , Animales , Células Cultivadas , Cerebrósido Sulfatasa/metabolismo , Exosomas/genética , Exosomas/metabolismo , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Oligodendroglía/citología , Oligodendroglía/metabolismo , Proteolisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Transcripción Genética
20.
Ann Neurol ; 75(1): 127-37, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24242821

RESUMEN

OBJECTIVE: To facilitate development of novel disease-modifying therapies for lysosomal storage disorder (LSDs) characterized by nervous system involvement such as metachromatic leukodystrophy (MLD), molecular markers for monitoring disease progression and therapeutic response are needed. To this end, we sought to identify blood transcripts associated with the progression of MLD. METHODS: Genome-wide expression analysis was performed in primary T lymphocytes of 24 patients with MLD compared to 24 age- and sex-matched healthy controls. Genes associated with MLD were identified, confirmed on a quantitative polymerase chain reaction platform, and replicated in an independent patient cohort. mRNA and protein expression of the prioritized gene family of metallothioneins was evaluated in postmortem patient brains and in mouse models representing 6 other LSDs. Metallothionein expression during disease progression and in response to specific treatment was evaluated in 1 of the tested LSD mouse models. Finally, a set of in vitro studies was planned to dissect the biological functions exerted by this class of molecules. RESULTS: Metallothionein genes were significantly overexpressed in T lymphocytes and brain of patients with MLD and generally marked nervous tissue damage in the LSDs here evaluated. Overexpression of metallothioneins correlated with measures of disease progression in mice and patients, whereas their levels decreased in mice upon therapeutic treatment. In vitro studies indicated that metallothionein expression is regulated in response to oxidative stress and inflammation, which are biochemical hallmarks of lysosomal storage diseases. INTERPRETATION: Metallothioneins are potential markers of neurologic disease processes and treatment response in LSDs.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Leucodistrofia Metacromática/metabolismo , Enfermedades por Almacenamiento Lisosomal/metabolismo , Metalotioneína/química , Simulación de Dinámica Molecular , Animales , Biomarcadores/metabolismo , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Leucodistrofia Metacromática/diagnóstico , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades por Almacenamiento Lisosomal/patología , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...