Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci Res ; 93(3): 424-32, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25332157

RESUMEN

Mucopolysaccharidosis (MPS) type IIIB is a genetic deficiency of α-N-acetylglucosaminidase, inducing accumulation of partially degraded heparan sulfate (HS) oligosaccharides in tissues. In the central nervous system, this accumulation is associated with microglial activation, neurodegeneration, and oxidative stress. We have already shown that HS activates microglial cells through toll-like receptor 4 (TLR4) and triggers neuroinflammation. The present study investigates whether oxidative stress is a direct consequence of inflammation or is an independent event directly caused by HS accumulation. The present study addresses causative links between oxidative stress and inflammation by analyzing the corresponding markers in the cortex of control mice, MPSIIIB mice (with neuroinflammation), and double mutant TLR4 knockout MPSIIIB mice (without neuroinflammation at early stages). Results showed that, although inflammation was not present in the cortex of 10-day-old double mutant MPSIIIB/TLR4(-/-) mice, the enzymatic activity of total superoxide dismutase (SOD) was already greater than in control animals. Moreover, at 3 and 8 months of age, the total enzymatic activities of glutathione peroxidase, SOD, and carbonyl protein levels in the cortex of MPSIIIB/TLR4(-/-) mice were similar to those measured in MPSIIIB mice and were higher than those in controls. The results indicate that the oxidative stress present at a very early stage in the brain of MPSIIIB mice is not the consequence of neuroinflammation. Insofar as it has an impact on the development of neurological disease, reducing oxidative stress might prevent or slow the progression of MPSIIIB.


Asunto(s)
Encéfalo/metabolismo , Inflamación/metabolismo , Mucopolisacaridosis III/metabolismo , Estrés Oxidativo/fisiología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glutatión Peroxidasa/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , Mucopolisacaridosis III/patología , NADPH Oxidasas/metabolismo , Superóxido Dismutasa/metabolismo , Receptor Toll-Like 4/genética
2.
Hum Mol Genet ; 21(7): 1481-95, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22156940

RESUMEN

Cell pathology in lysosomal storage diseases is characterized by the formation of distended vacuoles with characteristics of lysosomes. Our previous studies in mucopolysaccharidosis type IIIB (MPSIIIB), a disease in which a genetic defect induces the accumulation of undigested heparan sulfate (HS) fragments, led to the hypothesis that abnormal lysosome formation was related to events occurring at the Golgi level. We reproduced the enzyme defect of MPSIIIB in HeLa cells using tetracycline-inducible expression of shRNAs directed against α-N-acetylglucosaminidase (NAGLU) and addressed this hypothesis. HeLa cells deprived of NAGLU accumulated abnormal lysosomes. The Golgi matrix protein GM130 was over-expressed. The cis- and medial-Golgi compartments were distended, elongated and formed circularized ribbons. The Golgi microtubule network was enlarged with increased amounts of AKAP450, a partner of GM130 controlling this network. GM130 down-regulation prevented pathology in HeLa cells deprived of NAGLU, whereas GM130 over-expression in control HeLa cells mimicked the pathology of deprived cells. We concluded that abnormal lysosomes forming in cells accumulating HS fragments were the consequence of GM130 gain-of-function and subsequent alterations of the Golgi ribbon architecture. These results indicate that GM130 functions are modulated by HS glycosaminoglycans and therefore possibly controlled by extracellular cues.


Asunto(s)
Autoantígenos/metabolismo , Proteínas de la Membrana/metabolismo , Mucopolisacaridosis III/patología , Acetilglucosaminidasa/antagonistas & inhibidores , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Lisosomas/patología , Microtúbulos/ultraestructura , Modelos Biológicos , Vacuolas/ultraestructura
3.
Hum Mol Genet ; 20(18): 3653-66, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21685203

RESUMEN

By providing access to affected neurons, human induced pluripotent stem cells (iPSc) offer a unique opportunity to model human neurodegenerative diseases. We generated human iPSc from the skin fibroblasts of children with mucopolysaccharidosis type IIIB. In this fatal lysosomal storage disease, defective α-N-acetylglucosaminidase interrupts the degradation of heparan sulfate (HS) proteoglycans and induces cell disorders predominating in the central nervous system, causing relentless progression toward severe mental retardation. Partially digested proteoglycans, which affect fibroblast growth factor signaling, accumulated in patient cells. They impaired isolation of emerging iPSc unless exogenous supply of the missing enzyme cleared storage and restored cell proliferation. After several passages, patient iPSc starved of an exogenous enzyme continued to proliferate in the presence of fibroblast growth factor despite HS accumulation. Survival and neural differentiation of patient iPSc were comparable with unaffected controls. Whereas cell pathology was modest in floating neurosphere cultures, undifferentiated patient iPSc and their neuronal progeny expressed cell disorders consisting of storage vesicles and severe disorganization of Golgi ribbons associated with modified expression of the Golgi matrix protein GM130. Gene expression profiling in neural stem cells pointed to alterations of extracellular matrix constituents and cell-matrix interactions, whereas genes associated with lysosome or Golgi apparatus functions were downregulated. Taken together, these results suggest defective responses of patient undifferentiated stem cells and neurons to environmental cues, which possibly affect Golgi organization, cell migration and neuritogenesis. This could have potential consequences on post-natal neurological development, once HS proteoglycan accumulation becomes prominent in the affected child brain.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Lisosomas/metabolismo , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/fisiopatología , Neuronas/citología , Acetilglucosaminidasa/genética , Acetilglucosaminidasa/metabolismo , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas/enzimología , Masculino , Modelos Biológicos , Mucopolisacaridosis III/enzimología , Mucopolisacaridosis III/genética , Mutación , Neuronas/enzimología , Neuronas/metabolismo
4.
Mol Ther ; 19(2): 251-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21139569

RESUMEN

Recent trials in patients with neurodegenerative diseases documented the safety of gene therapy based on adeno-associated virus (AAV) vectors deposited into the brain. Inborn errors of the metabolism are the most frequent causes of neurodegeneration in pre-adulthood. In Sanfilippo syndrome, a lysosomal storage disease in which heparan sulfate oligosaccharides accumulate, the onset of clinical manifestation is before 5 years. Studies in the mouse model showed that gene therapy providing the missing enzyme α-N-acetyl-glucosaminidase to brain cells prevents neurodegeneration and improves behavior. We now document safety and efficacy in affected dogs. Animals received eight deposits of a serotype 5 AAV vector, including vector prepared in insect Sf9 cells. As shown previously in dogs with the closely related Hurler syndrome, immunosuppression was necessary to prevent neuroinflammation and elimination of transduced cells. In immunosuppressed dogs, vector was efficiently delivered throughout the brain, induced α-N-acetyl-glucosaminidase production, cleared stored compounds and storage lesions. The suitability of the procedure for clinical application was further assessed in Hurler dogs, providing information on reproducibility, tolerance, appropriate vector type and dosage, and optimal age for treatment in a total number of 25 treated dogs. Results strongly support projects of human trials aimed at assessing this treatment in Sanfilippo syndrome.


Asunto(s)
Encéfalo/metabolismo , Terapia Genética/métodos , Mucopolisacaridosis III/terapia , Mucopolisacaridosis I/terapia , Acetilglucosaminidasa/genética , Animales , Encéfalo/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Reacción en Cadena de la Polimerasa
5.
Am J Pathol ; 177(6): 2984-99, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21037080

RESUMEN

The accumulation of intracellular storage vesicles is a hallmark of lysosomal storage diseases. Neither the identity nor origin of these implicated storage vesicles have yet been established. The vesicles are often considered as lysosomes, endosomes, and/or autophagosomes that are engorged with undigested materials. Our studies in the mouse model of mucopolysaccharidosis type IIIB, a lysosomal storage disease that induces neurodegeneration, showed that large storage vesicles in cortical neurons did not receive material from either the endocytic or autophagy pathway, which functioned normally. Storage vesicles expressed GM130, a Golgi matrix protein, which mediates vesicle tethering in both pre- and cis-Golgi compartments. However, other components of the tethering/fusion complex were not associated with GM130 on storage vesicles, likely accounting for both the resistance of the vesicles to brefeldin A and the alteration of Golgi ribbon architecture, which comprised distended cisterna connected to LAMP1-positive storage vesicles. We propose that alteration in the GM130-mediated control of vesicle trafficking in pre-Golgi and Golgi compartments affects Golgi biogenesis and gives rise to a dead-end storage compartment. Vesicle accumulation, Golgi disorganization, and alterations of other GM130 functions may account for neuron dysfunction and death.


Asunto(s)
Vesículas Citoplasmáticas/patología , Aparato de Golgi/patología , Neuronas/ultraestructura , Animales , Autofagia/fisiología , Transporte Biológico/fisiología , Células Cultivadas , Vesículas Citoplasmáticas/metabolismo , Modelos Animales de Enfermedad , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Lisosomas/patología , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mucopolisacaridosis III/complicaciones , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/patología , Neuronas/metabolismo , Neuronas/patología , Neuronas/fisiología
6.
Am J Med Genet A ; 155A(1): 58-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21204211

RESUMEN

Sanfilippo syndrome, or mucopolysaccharidosis type III (MPSIII) is a lysosomal storage disease with predominant neurological manifestations in affected children. It is considered heterogeneous with respect to prevalence, clinical presentation, biochemistry (four biochemical forms of the disease referred to as MPSIIIA, B, C, and D are known), and causative mutations. The perspective of therapeutic options emphasizes the need for better knowledge of MPSIII incidence and natural history. We performed parallel retrospective epidemiological studies of patients diagnosed with MSPIII in France (n = 128), UK (n = 126), and Greece (n = 20) from 1990 to 2006. Incidences ranged from 0.68 per 100,000 live-births in France to 1.21 per 100,000 live-births in UK. MPSIIIA, which predominates in France and UK, was absent in Greece, where most patients have MPSIIIB. The study confirmed the large allelic heterogeneity of MPSIIIA and MPSIIIB and detected several yet undescribed mutations. Analysis of clinical manifestations at diagnosis and over a 6-7 years follow-up indicated that almost all patients, whatever the disease subtype, expressed neurological manifestations before the age of 5 years, including language acquisition delay, cognitive delay, and/or abnormal behavior. In contrast to relatively homogeneous early onset manifestations, disease progression showed significant variation depending on subtype and age at diagnosis. Different severities of disease progressions and different allele distribution between France and UK suggested that mutations are not equally deleterious, although genotype-phenotype correlation could not be established. Notwithstanding the rapidity of further clinical deterioration, all MPSIII patients suffer early onset devastating neurological manifestations that deserve early treatment when available.


Asunto(s)
Hidrolasas/genética , Mucopolisacaridosis III/epidemiología , Mucopolisacaridosis III/genética , Adolescente , Factores de Edad , Niño , Preescolar , Progresión de la Enfermedad , Francia/epidemiología , Grecia/epidemiología , Humanos , Hidrolasas/metabolismo , Incidencia , Lactante , Hígado/metabolismo , Mucopolisacaridosis III/patología , Mutación/genética , Estudios Retrospectivos , Reino Unido/epidemiología
7.
Front Neurol ; 12: 640547, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054689

RESUMEN

Mucopolysaccharidosis type I (MPS I) is caused by a deficiency of the lysosomal hydroxylase alpha-l-iduronidase (IDUA). The resulting accumulation of dermatan and heparan sulfate induces intellectual disabilities and pre-mature death, and only a few treatment options are available. In a previous study, we demonstrated the feasibility, safety, and efficacy of gene therapy by injecting recombinant adeno-associated viral vector serotype (AAV)2/5-IDUA into the brain of a canine model of MPS I. We report on a quantitative proteomic analysis of control dogs and untreated dogs with MPS I cerebrospinal fluid (CSF) that had been collected throughout the study in the MPS I dogs. Mass spectrometry (MS) analysis identified numerous proteins present at altered levels in MPS I CSF samples. Quantitative immunoblotting, performed on CSF from healthy controls, untreated MPS I dogs, and MPS I dogs early treated and late treated by gene therapy, confirmed the MS data for a subset of proteins with higher abundance (neuronal pentraxin 1, chitinase 3-like 1, monocyte differentiation antigen CD14, and insulin-like growth factor-binding protein 2). Scoring of the results shows that the expression levels of these proteins are close to those of the control group for dogs that underwent gene therapy early in life but not for older treated animals. Our results disclose four novel predictive biomarker candidates that might be valuable in monitoring the course of the neurological disease in MPS patients at diagnosis, during clinical follow-up, and after treatment.

8.
Front Immunol ; 12: 655478, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040605

RESUMEN

Mucopolysaccharidosis type IIIB syndrome (Sanfilippo disease) is a rare autosomic recessif disorder caused by mutations in the α-N-acetylglucosaminidase (NAGLU) gene coding for a lysosomal enzyme, leading to neurodegeneration and progressive deterioration of cognitive abilities in affected children. To supply the missing enzyme, several recent human gene therapy trials relied on the deposit of adeno-associated virus (AAV) vectors directly into the brain. We reported safety and efficacy of an intracerebral therapy in a phase 1/2 clinical trial (https://clinicaltrials.gov/ct2/show/NCT03300453), with a recombinant AAV serotype 2/5 (rAAV2/5) coding human NAGLU in four children with MPS IIIB syndrome receiving immunosuppression. It was reported that AAV-mediated gene therapies might elicit a strong host immune response resulting in decreased transgene expression. To address this issue, we performed a comprehensive analysis of cellular immunity and cytokine patterns generated against the therapeutic enzyme in the four treated children over 5.5 years of follow-up. We report the emergence of memory and polyfunctional CD4+ and CD8+ T lymphocytes sensitized to the transgene soon after the start of therapy, and appearing in peripheral blood in waves throughout the follow-up. However, this response had no apparent impact on CNS transgene expression, which remained stable 66 months after surgery, possibly a consequence of the long-term immunosuppressive treatment. We also report that gene therapy did not trigger neuroinflammation, evaluated through the expression of cytokines and chemokines in patients' CSF. Milder disease progression in the youngest patient was found associated with low level and less differentiated circulating NAGLU-specific T cells, together with the lack of proinflammatory cytokines in the CSF. Findings in this study support a systematic and comprehensive immunomonitoring approach for understanding the impact immune reactions might have on treatment safety and efficacy of gene therapies.


Asunto(s)
Acetilglucosaminidasa/inmunología , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Inmunidad Celular , Mucopolisacaridosis III/complicaciones , Transgenes/inmunología , Acetilglucosaminidasa/genética , Niño , Citocinas/metabolismo , Vías de Administración de Medicamentos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Memoria Inmunológica , Activación de Linfocitos , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes/genética
9.
Hum Gene Ther ; 32(19-20): 1251-1259, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34405688

RESUMEN

We report the safety (primary endpoint) and efficacy (secondary endpoint) of a novel intracerebral gene therapy at 5.5 years of follow-up in children with Sanfilippo B. An uncontrolled, phase 1/2 clinical trial was performed in four patients aged 20, 26, 30, and 53 months. Treatment consisted of 16 intracerebral and cerebellar deposits of a recombinant adeno-associated viral vector encoding human α-N-acetylglucosaminidase (rAAV2/5-hNAGLU) plus immunosuppression. An intermediate report at 30 months was previously published. Thirty treatment-emergent adverse events were reported between 30 and 66 months after surgery, including three classified as severe with no serious drug reactions. At 5.5 years, NAGLU activity was persistently detected in the lumbar cerebrospinal fluid (18% of unaffected control level). Circulating T cells reacting against NAGLU peptides were present, indicating a lack of acquired tolerance. Patients 2, 3, and 4 showed progressive brain atrophy and neurocognitive evolution that did not differ from untreated Sanfilippo A/B children. Patient 1, enrolled at 20 months of age, had a milder disease with normal brain imaging and a significantly better cognitive outcome than the three other patients and untreated patients, although not equivalent to normal children. After 5.5 years, the primary endpoint of this study was achieved with a good safety profile of the proposed treatment. We have also observed sustained enzyme production in the brain and absence of immunological tolerance. Cognitive benefit was not confirmed in the three oldest patients. Milder disease in the youngest patient supports further investigations of adeno-associated vector-mediated intracerebral gene therapy in Sanfilippo B.


Asunto(s)
Mucopolisacaridosis III , Encéfalo/diagnóstico por imagen , Preescolar , Estudios de Seguimiento , Terapia Genética , Humanos , Lactante , Recién Nacido , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Linfocitos T
10.
J Neurosci Res ; 88(1): 202-13, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19658197

RESUMEN

Behavioral manifestations mark the onset of disease expression in children with mucopolysaccharidosis type III (MPSIII, Sanfilippo syndrome), a genetic disorder resulting from interruption of the lysosomal degradation of heparan sulfate. In the mouse model of MPSIII type B (MPSIIIB), cortical neuron pathology and dysfunction occur several months before neuronal loss and are primarily cell autonomous. The gene coding for GAP43, a neurite growth potentiator, is overexpressed in the MPSIIIB mouse cortex, and neurite dystrophy was reported in other types of lysosomal storage diseases. We therefore examined the development of the neuritic trees in pure populations of MPSIIIB mouse embryo cortical neurons grown for up to 12 days in primary culture. Dynamic observation of living neurons and quantification of neurite growth parameters indicated more frequent neurite elongation and branching and less frequent neurite retraction, resulting in a relative overgrowth of MPSIIIB neuron neuritic trees, involving both dendrites and axons, compared with normal controls. Neurite overgrowth was concomitant with more than twofold increased expression of GAP43 mRNAs and proteins. Correction of the genetic defect leads to expression of the missing lysosomal enzyme, normal GAP43 mRNA expression, and reduced neurite outgrowth. These results indicate that heparan sulfate oligosaccharide storage modifies GAP43 expression in MPSIIIB cortical neurons with potential consequences for neurite development and neuronal functions that may be relevant to clinical manifestations.


Asunto(s)
Corteza Cerebral/metabolismo , Proteína GAP-43/metabolismo , Mucopolisacaridosis III/metabolismo , Neuritas/metabolismo , Animales , Western Blotting , Forma de la Célula/fisiología , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Proteína GAP-43/genética , Expresión Génica , Vectores Genéticos/metabolismo , Lentivirus/metabolismo , Ratones , Mucopolisacaridosis III/genética , Plasticidad Neuronal/fisiología , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Factores de Tiempo
11.
Biochem Soc Trans ; 38(6): 1442-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21118104

RESUMEN

Biochemical disorders in lysosomal storage diseases consist of the interruption of metabolic pathways involved in the recycling of the degradation products of one or several types of macromolecules. The progressive accumulation of these primary storage products is the direct consequence of the genetic defect and represents the initial pathogenic event. Downstream consequences for the affected cells include the accumulation of secondary storage products and the formation of histological storage lesions, which appear as intracellular vacuoles that represent the pathological hallmark of lysosomal storage diseases. Relationships between storage products and storage lesions are not simple and are still largely not understood. Primary storage products induce malfunction of the organelles where they accumulate, these being primarily, but not only, lysosomes. Consequences for cell metabolism and intracellular trafficking combine the effects of primary storage product toxicity and the compensatory mechanisms activated to protect the cell. Induced disorders extend far beyond the primarily interrupted metabolic pathway.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/metabolismo , Lisosomas/metabolismo , Redes y Vías Metabólicas , Animales , Autoantígenos/metabolismo , Transporte Biológico , Encéfalo/metabolismo , Encéfalo/patología , Aparato de Golgi/patología , Humanos , Enfermedades por Almacenamiento Lisosomal/patología , Lisosomas/patología , Proteínas de la Membrana/metabolismo
12.
Mol Ther ; 17(6): 992-1002, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19240691

RESUMEN

Brachial plexus injury is frequent after traffic accident in adults or shoulder dystocia in newborns. Whereas surgery can restore arm movements, therapeutic options are missing for sensory defects. Dorsal root (DR) ganglion neurons convey sensory information to the central nervous system (CNS) through a peripheral and a central axon. Central axons severed through DR section or avulsion during brachial plexus injury inefficiently regenerate and do not reenter the spinal cord. We show that a combination of microsurgery and gene therapy circumvented the functional barrier to axonal regrowth at the peripheral and CNS interface. After cervical DR section in rats, microsurgery restored anatomical continuity through a nerve graft that laterally connected the injured DR to an intact DR. Gene transfer to cells in the nerve graft induced the local release of neurotrophin-3 (NT-3) and glial cell line-derived neurotrophic factor (GDNF) and stimulated axonal regrowth. Central DR ganglion axons efficiently regenerated and invaded appropriate areas of the spinal cord dorsal horn, leading to partial recovery of nociception and proprioception. Microsurgery created conditions for functional restoration of DR ganglion central axons, which were improved in combination with gene therapy. This combination treatment provides means to reduce disability due to somatosensory defects after brachial plexus injury.


Asunto(s)
Ganglios Espinales/lesiones , Ganglios Espinales/cirugía , Terapia Genética/métodos , Traumatismos de la Médula Espinal/cirugía , Traumatismos de la Médula Espinal/terapia , Raíces Nerviosas Espinales/lesiones , Raíces Nerviosas Espinales/cirugía , Animales , Electrofisiología , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Inmunohistoquímica , Masculino , Microscopía Electrónica de Transmisión , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Neurotrofina 3/genética , Neurotrofina 3/fisiología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas F344 , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Raíces Nerviosas Espinales/metabolismo
13.
Mol Cell Neurosci ; 41(1): 8-18, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19386237

RESUMEN

The interruption of the lysosomal degradation of heparan sulfate oligosaccharides has deleterious consequences on the central nervous system in children or in animals with mucopolysaccharidosis type III (Sanfilippo syndrome). Behavioural manifestations are prominent at disease onset, suggesting possible early synaptic defects in cortical neurons. We report that synaptophysin, the most abundant protein of the synaptic vesicle membrane, was detected at low levels in the rostral cortex of MPSIII type B mice as early as 10 days after birth. This defect preceded other disease manifestations, was associated with normal neuron and synapse density and corrected after gene transfer inducing re-expression of the missing lysosomal enzyme. Clearance of heparan sulfate oligosaccharides in cultured embryonic MPSIIIB cortical neurons or treatment with proteasome inhibitors restored normal synaptophysin levels indicating that heparan sulfate oligosaccharides activate the degradation of synaptophysin by the proteasome with consequences on synaptic vesicle components that are relevant to clinical manifestations.


Asunto(s)
Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/fisiopatología , Complejo de la Endopetidasa Proteasomal/metabolismo , Sinaptofisina/metabolismo , Acetilglucosaminidasa/genética , Acetilglucosaminidasa/metabolismo , Animales , Conducta/fisiología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Niño , Femenino , Proteína GAP-43/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucopolisacaridosis III/patología , Neuronas/citología , Neuronas/metabolismo , Proteínas R-SNARE/metabolismo , Sinaptofisina/genética
14.
Orphanet J Rare Dis ; 15(1): 3, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31907071

RESUMEN

BACKGROUND: The European Medicine Agency granted marketing approval to 164 orphan medicinal products for rare diseases, among which 28 products intended for the treatment of hereditary metabolic diseases. Taking advantage of its privileged connection with 69 healthcare centres of excellence in this field, MetabERN, the European Reference Network for hereditary metabolic diseases, performed a survey asking health care providers from 18 European countries whether these products are available on the market, reimbursed and therefore accessible for prescription, and actually delivered in their centre. RESULTS: Responses received from 52 centres (75%) concerned the design of treatment plans, the access to marketed products, and the barriers to delivery. Treatment options are always discussed with patients, who are often involved in their treatment plan. Most products (26/28) are available in most countries (15/18). Among the 15 broadly accessible products (88.5% of the centres), 9 are delivered to most patients (mean 70.1%), and the others to only few (16.5%). Among the 10 less accessible products (40.2% of the centres), 6 are delivered to many patients (66.7%), and 4 are rarely used (6.3%). Information was missing for 3 products. Delay between prescription and delivery is on average one month. Beside the lack of availability or accessibility, the most frequent reasons for not prescribing a treatment are patients' clinical status, characteristic, and personal choice. CONCLUSIONS: Data collected from health care providers in the MetabERN network indicate that two-third of the orphan medicines approved by EMA for the treatment of hereditary metabolic diseases are accessible to treating patients, although often less than one-half of the patients with the relevant conditions actually received the approved product to treat their disease. Thus, in spite of the remarkable achievement of many products, patients concerned by EMA-approved orphan medicinal products have persistent unmet needs, which deserve consideration. The enormous investments made by the companies to develop products, and the high financial burden for the Member States to purchase these products emphasize the importance of a scrupulous appreciation of treatment value involving all stakeholders at early stage of development, before marketing authorization, and during follow up.


Asunto(s)
Enfermedades Metabólicas/tratamiento farmacológico , Errores Innatos del Metabolismo/tratamiento farmacológico , Producción de Medicamentos sin Interés Comercial/métodos , Aprobación de Drogas , Humanos , Enfermedades Raras , Encuestas y Cuestionarios
15.
Stem Cells ; 26(10): 2564-75, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18635866

RESUMEN

Stem cell-based therapies hold therapeutic promise for degenerative motor neuron diseases, such as amyotrophic lateral sclerosis, and for spinal cord injury. Fetal neural progenitors present less risk of tumor formation than embryonic stem cells but inefficiently differentiate into motor neurons, in line with their low expression of motor neuron-specific transcription factors and poor response to soluble external factors. To overcome this limitation, we genetically engineered fetal rat spinal cord neurospheres to express the transcription factors HB9, Nkx6.1, and Neurogenin2. Enforced expression of the three factors rendered neural precursors responsive to Sonic hedgehog and retinoic acid and directed their differentiation into cholinergic motor neurons that projected axons and formed contacts with cocultured myotubes. When transplanted in the injured adult rat spinal cord, a model of acute motor neuron degeneration, the engineered precursors transiently proliferated, colonized the ventral horn, expressed motor neuron-specific differentiation markers, and projected cholinergic axons in the ventral root. We conclude that genetic engineering can drive the differentiation of fetal neural precursors into motor neurons that efficiently engraft in the spinal cord. The strategy thus holds promise for cell replacement in motor neuron and related diseases. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Evolución Molecular Dirigida , Ingeniería Genética , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Axones/metabolismo , Biomarcadores/metabolismo , Comunicación Celular , Diferenciación Celular , Movimiento Celular , Colina/metabolismo , Técnicas de Cocultivo , Humanos , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Especificidad de Órganos , Ratas , Traumatismos de la Médula Espinal/patología , Raíces Nerviosas Espinales/patología , Trasplante de Células Madre , Factores de Transcripción/metabolismo
16.
Orphanet J Rare Dis ; 14(1): 119, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142374

RESUMEN

BACKGROUND: MetabERN is one of the 24 European Reference Networks created according to the European Union directive 2011/24/EU on patient's rights in cross border healthcare. MetabERN associates 69 centres in 18 countries, which provide care for patients with Hereditary Metabolic Diseases, and have the mission to reinforce research and provide training for health professionals in this field. MetabERN performed a survey in December 2017 with the aim to produce an overview documenting research activities and potentials within the network. As the centres are multidisciplinary, separated questionnaires were sent to the clinical, university and laboratory teams. Answers were received from 52 out of the 69 centres of the network, covering 16 countries. A descriptive analysis of the information collected is presented. RESULTS: The answers indicate a marked interest of the respondents for research, who expressed high motivation and commitment, and estimated that the conditions to do research in their institution were mostly satisfactory. They are active in research, which according to several indicators, is competitive and satisfies standards of excellence, as well as the education programs offered in the respondent's universities. Research in the centres is primarily performed in genetics, pathophysiology, and epidemiology, and focuses on issues related to diagnosis. Few respondents declared having activity in human and social sciences, including research on patient's quality of life, patient's awareness, or methods for social support. Infrastructures offering services for medical research were rarely known and used by respondents, including national and international biobanking platforms. In contrast, respondents often participate to patient registries, even beyond their specific field of interest. CONCLUSIONS: Taken as a whole, these results provide an encouraging picture of the research capacities and activities in the MetabERN network, which, with respect to the number and representativeness of the investigated centres, gives a comprehensive picture of research on Hereditary Metabolic Diseases in Europe, as well as the priorities for future actions. Marginal activity in human and social sciences points out the limited multidisciplinary constitution of the responding teams with possible consequences on their current capability to participate to patient's empowerment programs and efficiently collaborate with patient's advocacy groups.


Asunto(s)
Investigación Interdisciplinaria/métodos , Europa (Continente) , Humanos , Calidad de Vida , Encuestas y Cuestionarios
17.
Mol Genet Metab ; 94(1): 135-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18325808

RESUMEN

Mucopolysaccharidosis IIIB is a lysosomal disease characterized by a severe neurological deterioration, the pathophysiological mechanisms of which are poorly understood. Recently FGF pathway was shown to be altered leading us to explore a downstream target involved in brain development: the collapsin response mediator protein-1 (CRMP-1). CRMP-1 transcript level was normal but a cleavage of CRMP-1 was observed with an abnormal expression of the truncated form until adult age. This truncated CRMP-1 protein could play a role in post-natal cortex maturation and be involved in neuronal alterations occurring in lysosomal diseases.


Asunto(s)
Corteza Cerebral/metabolismo , Mucopolisacaridosis III/genética , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética , Animales , Calpaína/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis III/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo
18.
Lancet Neurol ; 16(9): 712-720, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28713035

RESUMEN

BACKGROUND: Mucopolysaccharidosis type IIIB syndrome (also known as Sanfilippo type B syndrome) is a lysosomal storage disease resulting in progressive deterioration of cognitive acquisition after age 2-4 years. No treatment is available for the neurological manifestations of the disease. We sought to assess the safety and efficacy of a novel intracerebral gene therapy. METHODS: Local regulatory authorities in France allowed inclusion of up to four children in this phase 1/2 study. Treatment was 16 intraparenchymal deposits (four in the cerebellum) of a recombinant adenoassociated viral vector serotype 2/5 (rAAV2/5) encoding human α-N-acetylglucosaminidase (NAGLU) plus immunosuppressive therapy. We assessed tolerance, neurocognitive progression, brain growth, NAGLU enzymatic activity in CSF, and specific anti-NAGLU immune response for 30 months after surgery. This trial is registered with EudraCT, number 2012-000856-33, and the International Standard Clinical Trial Registry, number ISRCTN19853672. FINDINGS: Of seven eligible children, the four youngest, from France (n=2), Italy (n=1), and Greece (n=1), aged 20, 26, 30, and 53 months, were included between February, 2012, and February, 2014. 125 adverse events were recorded, of which 117 were treatment emergent and included six classified as severe, but no suspected unexpected serious adverse drug reactions were seen. Vector genomes were detected in blood for 2 days after surgery. Compared with the natural history of mucopolysaccharidosis type III syndromes, neurocognitive progression was improved in all patients, with the youngest patient having function close to that in healthy children. Decrease in developmental quotient was -11·0 points in patient one, -23·0 in patient two, -29·0 in patient three, and -17·0 in patient four, compared with -37·7 in the natural history of the disease. NAGLU activity was detected in lumbar CSF and was 15-20% of that in unaffected children. Circulating T lymphocytes that proliferated and produced tumour necrosis factor α upon ex-vivo exposure to NAGLU antigens were detectable at 1-12 months and 3-12 months, respectively, but not at 30 months in three of four patients. INTERPRETATION: Intracerebral rAVV2/5 was well tolerated and induced sustained enzyme production in the brain. The initial specific anti-NAGLU immune response that later subsided suggested acquired immunological tolerance. The best results being obtained in the youngest patient implies a potential window of opportunity. Longer follow-up is needed to further assess safety outcomes and persistence of improved cognitive development. FUNDING: Association Française Contre les Myopathies, Vaincre les Maladies Lysosomales, Institut Pasteur, and UniQure.


Asunto(s)
Acetilglucosaminidasa , Encéfalo/enzimología , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/farmacología , Mucopolisacaridosis III/terapia , Evaluación de Resultado en la Atención de Salud , Acetilglucosaminidasa/genética , Preescolar , Terapia Genética/efectos adversos , Vectores Genéticos/administración & dosificación , Humanos , Inmunosupresores/uso terapéutico , Lactante , Mucopolisacaridosis III/tratamiento farmacológico , Síndrome
19.
J Neurosci ; 24(45): 10229-39, 2004 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-15537895

RESUMEN

Sanfilippo syndrome is a mucopolysaccharidosis (MPS) caused by a lysosomal enzyme defect interrupting the degradation pathway of heparan sulfates. Affected children develop hyperactivity, aggressiveness, delayed development, and severe neuropathology. We observed relevant behaviors in the mouse model of Sanfilippo syndrome type B (MPSIIIB), in which the gene coding for alpha-N-acetylglucosaminidase (NaGlu) is invalidated. We addressed the feasibility of gene therapy in these animals. Vectors derived from adeno-associated virus serotype 2 (AAV2) or 5 (AAV5) coding for NaGlu were injected at a single site in the putamen of 45 6-week-old MPSIIIB mice. Normal behavior was observed in treated mice. High NaGlu activity, far above physiological levels, was measured in the brain and persisted at 38 weeks of age. NaGlu immunoreactivity was detected in neuron intracellular organelles, including lysosomes. Enzyme activity spread beyond vector diffusion areas. Delivery to the entire brain was reproducibly obtained with both vector types. NaGlu activity was higher and distribution was broader with AAV5-NaGlu than with AAV2-NaGlu vectors. The compensatory increase in the activity of various lysosomal enzymes was improved. The accumulation of gangliosides GM2 and GM3 present before treatment and possibly participating in neuropathology was reversed. Characteristic vacuolations in microglia, perivascular cells, and neurons, which were prominent before the age of treatment, disappeared in areas in which NaGlu was present. However, improvement was only partial in some animals, in contrast to high NaGlu activity. These results indicate that NaGlu delivery from intracerebral sources has the capacity to alleviate most disease manifestations in the MPSIIIB mouse model.


Asunto(s)
Acetilglucosaminidasa/genética , Encéfalo/patología , Cuerpo Estriado , Dependovirus/genética , Gangliósido G(M2)/metabolismo , Gangliósido G(M3)/metabolismo , Terapia Genética , Vectores Genéticos/uso terapéutico , Mucopolisacaridosis III/terapia , Acetilglucosaminidasa/deficiencia , Animales , Encéfalo/enzimología , Dependovirus/clasificación , Conducta Exploratoria , Inyecciones , Lisosomas/enzimología , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucopolisacaridosis III/enzimología , Mucopolisacaridosis III/patología , Neuronas/metabolismo , Putamen
20.
J Mol Biol ; 340(1): 39-47, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15184021

RESUMEN

Pit2 is a member of the Pit family of inorganic phosphate transporters and serves as a gamma-retrovirus receptor in mammals. Pit2 contains two copies of the protein homology domain PD001131, which defines the Pit family. These domains are presumably in opposite topology with respect to the plasma membrane plane. We have mutated a serine residue conserved in almost all of the 192 known PD001131 sequences to alanine in each PD001131 domain of human Pit2. Expression in CHO cells showed that phosphate uptake was affected severely in mutants, whereas susceptibility to virus infection was conserved. We reported previously that the inorganic phosphate concentration affects both phosphate transport mediated by Pit2 and the conformation of cell-surface Pit2 oligomers. Cross-linking experiments in transport-incompetent Pit2 mutants indicated that structural changes induced by phosphate starvation or supply occur independently of the whole transport cycle. These results suggest that the structural reorganisation of cell-surface Pit2 occurred as a consequence of ion binding, a model consistent with the possible involvement of cell-surface Pit2 oligomers in inorganic phosphate sensing.


Asunto(s)
Fosfatos/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células CHO , Membrana Celular/metabolismo , Secuencia Conservada , Cricetinae , Humanos , Virus de la Leucemia Murina/fisiología , Datos de Secuencia Molecular , Mutación , Fosfatos/farmacología , Estructura Terciaria de Proteína , Receptores Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA