Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Neurol ; 86(3): 332-343, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31206741

RESUMEN

Progress in addressing the origins of intellectual and developmental disabilities accelerated with the establishment 50 years ago of the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health and associated Intellectual and Developmental Disabilities Research Centers. Investigators at these Centers have made seminal contributions to understanding human brain and behavioral development and defining mechanisms and treatments of disorders of the developing brain. ANN NEUROL 2019;86:332-343.


Asunto(s)
Academias e Institutos/historia , Discapacidades del Desarrollo , Discapacidad Intelectual , National Institute of Child Health and Human Development (U.S.)/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Estados Unidos
2.
J Neurochem ; 148(5): 669-689, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29770442

RESUMEN

Mucolipidosis type IV (MLIV) is an autosomal recessive, lysosomal storage disorder causing progressively severe intellectual disability, motor and speech deficits, retinal degeneration often culminating in blindness, and systemic disease causing a shortened lifespan. MLIV results from mutations in the gene MCOLN1 encoding the transient receptor potential channel mucolipin-1. It is an ultra-rare disease and is currently known to affect just over 100 diagnosed individuals. The last decade has provided a wealth of research focused on understanding the role of the enigmatic mucolipin-1 protein in cell and brain function and how its absence causes disease. This review explores our current understanding of the mucolipin-1 protein in relation to neuropathogenesis in MLIV and describes recent findings implicating mucolipin-1's important role in mechanistic target of rapamycin and TFEB (transcription factor EB) signaling feedback loops as well as in the function of the greater endosomal/lysosomal system. In addition to addressing the vital role of mucolipin-1 in the brain, we also report new data on the question of whether haploinsufficiency as would be anticipated in MCOLN1 heterozygotes is associated with any evidence of neuron dysfunction or disease. Greater insights into the role of mucolipin-1 in the nervous system can be expected to shed light not only on MLIV disease but also on numerous processes governing normal brain function. This article is part of the Special Issue "Lysosomal Storage Disorders".


Asunto(s)
Mucolipidosis , Animales , Humanos
3.
Hum Mol Genet ; 26(5): 843-859, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28062666

RESUMEN

2-hydroxypropyl-ß-cyclodextrin (CYCLO), a modifier of cholesterol efflux from cellular membrane and endo-lysosomal compartments, reduces lysosomal lipid accumulations and has therapeutic effects in animal models of Niemann-Pick disease type C and several other neurodegenerative states. Here, we investigated CYCLO effects on autophagy in wild-type mice and TgCRND8 mice-an Alzheimer's Disease (AD) model exhibiting ß-amyloidosis, neuronal autophagy deficits leading to protein and lipid accumulation within greatly enlarged autolysosomes. A 14-day intracerebroventricular administration of CYCLO to 8-month-old TgCRND8 mice that exhibit moderately advanced neuropathology markedly diminished the sizes of enlarged autolysosomes and lowered their content of GM2 ganglioside and Aß-immunoreactivity without detectably altering amyloid precursor protein processing or extracellular Aß/ß-amyloid burden. We identified two major actions of CYCLO on autophagy underlying amelioration of lysosomal pathology. First, CYCLO stimulated lysosomal proteolytic activity by increasing cathepsin D activity, levels of cathepsins B and D and two proteins known to interact with cathepsin D, NPC1 and ABCA1. Second, CYCLO impeded autophagosome-lysosome fusion as evidenced by the accumulation of LC3, SQSTM1/p62, and ubiquitinated substrates in an expanded population of autophagosomes in the absence of greater autophagy induction. By slowing substrate delivery to lysosomes, autophagosome maturational delay, as further confirmed by our in vitro studies, may relieve lysosomal stress due to accumulated substrates. These findings provide in vivo evidence for lysosomal enhancing properties of CYCLO, but caution that prolonged interference with cellular membrane fusion/autophagosome maturation could have unfavorable consequences, which might require careful optimization of dosage and dosing schedules.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Amiloidosis/tratamiento farmacológico , Ciclodextrinas/administración & dosificación , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Amiloidosis/metabolismo , Animales , Autofagia/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología
4.
Lancet ; 390(10104): 1758-1768, 2017 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-28803710

RESUMEN

BACKGROUND: Niemann-Pick disease, type C1 (NPC1) is a lysosomal storage disorder characterised by progressive neurodegeneration. In preclinical testing, 2-hydroxypropyl-ß-cyclodextrins (HPßCD) significantly delayed cerebellar Purkinje cell loss, slowed progression of neurological manifestations, and increased lifespan in mouse and cat models of NPC1. The aim of this study was to assess the safety and efficacy of lumbar intrathecal HPßCD. METHODS: In this open-label, dose-escalation phase 1-2a study, we gave monthly intrathecal HPßCD to participants with NPC1 with neurological manifestation at the National Institutes of Health (NIH), Bethesda, MD, USA. To explore the potential effect of 2-week dosing, three additional participants were enrolled in a parallel study at Rush University Medical Center (RUMC), Chicago, IL, USA. Participants from the NIH were non-randomly, sequentially assigned in cohorts of three to receive monthly initial intrathecal HPßCD at doses of 50, 200, 300, or 400 mg per month. A fifth cohort of two participants received initial doses of 900 mg. Participants from RUMC initially received 200 or 400 mg every 2 weeks. The dose was escalated based on tolerance or safety data from higher dose cohorts. Serum and CSF 24(S)-hydroxycholesterol (24[S]-HC), which serves as a biomarker of target engagement, and CSF protein biomarkers were evaluated. NPC Neurological Severity Scores (NNSS) were used to compare disease progression in HPßCD-treated participants relative to a historical comparison cohort of 21 NPC1 participants of similar age range. FINDINGS: Between Sept 21, 2013, and Jan 19, 2015, 32 participants with NPC1 were assessed for eligibility at the National Institutes of Health. 18 patients were excluded due to inclusion criteria not met (six patients), declined to participate (three patients), pursued independent expanded access and obtained the drug outside of the study (three patients), enrolled in the RUMC cohort (one patient), or too late for the trial enrolment (five patients). 14 patients were enrolled and sequentially assigned to receive intrathecal HPßCD at a starting dose of 50 mg per month (three patients), 200 mg per month (three patients), 300 mg per month (three patients), 400 mg per month (three patients), or 900 mg per month (two patients). During the first year, two patients had treatment interrupted for one dose, based on grade 1 ototoxicity. All 14 patients were assessed at 12 months. Between 12 and 18 months, one participant had treatment interrupted at 17 months due to hepatocellular carcinoma, one patient had dose interruption for 2 doses based on caregiver hardship and one patient had treatment interrupted for 1 dose for mastoiditis. 11 patients were assessed at 18 months. Between Dec 11, 2013, and June 25, 2014, three participants were assessed for eligibility and enrolled at RUMC, and were assigned to receive intrathecal HPßCD at a starting dose of 200 mg every 2 weeks (two patients), or 400 mg every two weeks (one patient). There were no dropouts in this group and all 3 patients were assessed at 18 months. Biomarker studies were consistent with improved neuronal cholesterol homoeostasis and decreased neuronal pathology. Post-drug plasma 24(S)-HC area under the curve (AUC8-72) values, an indicator of neuronal cholesterol homoeostasis, were significantly higher than post-saline plasma 24(S)-HC AUC8-72 after doses of 900 mg (p=0·0063) and 1200 mg (p=0·0037). CSF 24(S)-HC concentrations in three participants given either 600 or 900 mg of HPßCD were increased about two fold (p=0·0032) after drug administration. No drug-related serious adverse events were observed. Mid-frequency to high-frequency hearing loss, an expected adverse event, was documented in all participants. When managed with hearing aids, this did not have an appreciable effect on daily communication. The NNSS for the 14 participants treated monthly increased at a rate of 1·22, SEM 0·34 points per year compared with 2·92, SEM 0·27 points per year (p=0·0002) for the 21 patient comparison group. Decreased progression was observed for NNSS domains of ambulation (p=0·0622), cognition (p=0·0040) and speech (p=0·0423). INTERPRETATION: Patients with NPC1 treated with intrathecal HPßCD had slowed disease progression with an acceptable safety profile. These data support the initiation of a multinational, randomised, controlled trial of intrathecal HPßCD. FUNDING: National Institutes of Health, Dana's Angels Research Trust, Ara Parseghian Medical Research Foundation, Hope for Haley, Samantha's Search for the Cure Foundation, National Niemann-Pick Disease Foundation, Support of Accelerated Research for NPC Disease, Vtesse, Janssen Research and Development, a Johnson & Johnson company, and Johnson & Johnson.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/administración & dosificación , Progresión de la Enfermedad , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , 2-Hidroxipropil-beta-Ciclodextrina/efectos adversos , Adolescente , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Calbindinas/líquido cefalorraquídeo , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Proteína 3 de Unión a Ácidos Grasos/líquido cefalorraquídeo , Femenino , Pérdida Auditiva de Alta Frecuencia/inducido químicamente , Humanos , Hidroxicolesteroles/sangre , Hidroxicolesteroles/líquido cefalorraquídeo , Inyecciones Espinales , Masculino , Enfermedad de Niemann-Pick Tipo C/sangre , Enfermedad de Niemann-Pick Tipo C/líquido cefalorraquídeo , Enfermedades Raras/tratamiento farmacológico , Adulto Joven
5.
Am J Pathol ; 187(4): 864-883, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28342444

RESUMEN

Farber disease is a rare autosomal recessive disorder caused by acid ceramidase deficiency that usually presents as early-onset progressive visceral and neurologic disease. To understand the neurologic abnormality, we investigated behavioral, biochemical, and cellular abnormalities in the central nervous system of Asah1P361R/P361R mice, which serve as a model of Farber disease. Behaviorally, the mutant mice had reduced voluntary locomotion and exploration, increased thigmotaxis, abnormal spectra of basic behavioral activities, impaired muscle grip strength, and defects in motor coordination. A few mutant mice developed hydrocephalus. Mass spectrometry revealed elevations of ceramides, hydroxy-ceramides, dihydroceramides, sphingosine, dihexosylceramides, and monosialodihexosylganglioside in the brain. The highest accumulation was in hydroxy-ceramides. Storage compound distribution was analyzed by mass spectrometry imaging and morphologic analyses and revealed involvement of a wide range of central nervous system cell types (eg, neurons, endothelial cells, and choroid plexus cells), most notably microglia and/or macrophages. Coalescing and mostly perivascular granuloma-like accumulations of storage-laden CD68+ microglia and/or macrophages were seen as early as 3 weeks of age and located preferentially in white matter, periventricular zones, and meninges. Neurodegeneration was also evident in specific cerebral areas in late disease. Overall, our central nervous system studies in Asah1P361R/P361R mice substantially extend the understanding of human Farber disease and suggest that this model can be used to advance therapeutic approaches for this currently untreatable disorder.


Asunto(s)
Sistema Nervioso Central/anomalías , Lipogranulomatosis de Farber/complicaciones , Lipogranulomatosis de Farber/patología , Malformaciones del Sistema Nervioso/etiología , Malformaciones del Sistema Nervioso/patología , Ceramidasa Ácida/metabolismo , Animales , Conducta Animal , Sistema Nervioso Central/patología , Cerebelo/patología , Cerebelo/ultraestructura , Cerebro/patología , Cerebro/ultraestructura , Homocigoto , Hidrocefalia/patología , Ratones , Ratones Transgénicos , Actividad Motora , Neuronas/patología , Neuronas/ultraestructura , Fenotipo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingolípidos/metabolismo , Factores de Tiempo
6.
Neurobiol Dis ; 105: 257-270, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28610891

RESUMEN

Mucolipidosis type IV (MLIV) is a lysosomal storage disease exhibiting progressive intellectual disability, motor impairment, and premature death. There is currently no cure or corrective treatment. The disease results from mutations in the gene encoding mucolipin-1, a transient receptor potential channel believed to play a key role in lysosomal calcium egress. Loss of mucolipin-1 and subsequent defects lead to a host of cellular aberrations, including accumulation of glycosphingolipids (GSLs) in neurons and other cell types, microgliosis and, as reported here, cerebellar Purkinje cell loss. Several studies have demonstrated that N-butyldeoxynojirimycin (NB-DNJ, also known as miglustat), an inhibitor of the enzyme glucosylceramide synthase (GCS), successfully delays the onset of motor deficits, improves longevity, and rescues some of the cerebellar abnormalities (e.g., Purkinje cell death) seen in another lysosomal disease known as Niemann-Pick type C (NPC). Given the similarities in pathology between MLIV and NPC, we examined whether miglustat would be efficacious in ameliorating disease progression in MLIV. Using a full mucolipin-1 knockout mouse (Mcoln1-/-), we found that early miglustat treatment delays the onset and progression of motor deficits, delays cerebellar Purkinje cell loss, and reduces cerebellar microgliosis characteristic of MLIV disease. Quantitative mass spectrometry analyses provided new data on the GSL profiles of murine MLIV brain tissue and showed that miglustat partially restored the wild type profile of white matter enriched lipids. Collectively, our findings indicate that early miglustat treatment delays the progression of clinically relevant pathology in an MLIV mouse model, and therefore supports consideration of miglustat as a therapeutic agent for MLIV disease in humans.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Cerebelo/patología , Inhibidores Enzimáticos/uso terapéutico , Gliosis/tratamiento farmacológico , Trastornos del Movimiento/tratamiento farmacológico , Mucolipidosis , Células de Purkinje/efectos de los fármacos , 1-Desoxinojirimicina/uso terapéutico , Animales , Antígenos CD/metabolismo , Recuento de Células , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Gliosis/etiología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos del Movimiento/etiología , Mucolipidosis/complicaciones , Mucolipidosis/genética , Mucolipidosis/patología , Proteínas del Tejido Nervioso/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Células de Purkinje/patología , Retina/patología , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
7.
Hum Mol Genet ; 24(7): 1856-68, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452429

RESUMEN

Deficiency of arylsulfatase G (ARSG) leads to a lysosomal storage disease in mice resembling biochemical and pathological features of the mucopolysaccharidoses and particularly features of mucopolysaccharidosis type III (Sanfilippo syndrome). Here we show that Arsg KO mice share common neuropathological findings with other Sanfilippo syndrome models and patients, but they can be clearly distinguished by the limitation of most phenotypic alterations to the cerebellum, presenting with ataxia as the major neurological finding. We determined in detail the expression of ARSG in the central nervous system and observed highest expression in perivascular macrophages (which are characterized by abundant vacuolization in Arsg KO mice) and oligodendrocytes. To gain insight into possible mechanisms leading to ataxia, the pathology in older adult mice (>12 months) was investigated in detail. This study revealed massive loss of Purkinje cells and gliosis in the cerebellum, and secondary accumulation of glycolipids like GM2 and GM3 gangliosides and unesterified cholesterol in surviving Purkinje cells, as well as neurons of some other brain regions. The abundant presence of ubiquitin and p62-positive aggregates in degenerating Purkinje cells coupled with the absence of significant defects in macroautophagy is consistent with lysosomal membrane permeabilization playing a role in the pathogenesis of Arsg-deficient mice and presumably Sanfilippo disease in general. Our data delineating the phenotype of mucopolysaccharidosis IIIE in a mouse KO model should help in the identification of possible human cases of this disease.


Asunto(s)
Arilsulfatasas/deficiencia , Ataxia/enzimología , Mucopolisacaridosis III/enzimología , Animales , Arilsulfatasas/genética , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Cerebelo/citología , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Femenino , Gliosis/metabolismo , Glucolípidos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/patología , Células de Purkinje/metabolismo
8.
Mol Genet Metab ; 120(1-2): 34-37, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27923544

RESUMEN

The Niemann-Pick family of diseases was poorly understood until Roscoe Brady and his colleagues began their investigations in the 1960s. Following Brady's discovery of the defect in acid sphingomyelinase in Niemann-Pick disease, types A and B, Peter Pentchev, a senior scientist in the group, launched a series of investigations of an unusual lipid storage disease in a spontaneous mouse model. These led initially to identification of the cholesterol trafficking defect in the mouse, and then in human Niemann-Pick disease, type C (NPC). This discovery formed the basis of the standard diagnostic test for NPC for the next three decades. Subsequently, an international collaboration was established, based at the Brady lab at NIH, which culminated in discovery of the NPC1 gene. Roscoe Brady, Peter Pentchev and their colleagues defined and refined the clinical biochemical and pathological phenotypes of NPC in a series of elegant parallel studies. They also identified abnormal oxysterols in NPC; later work has proved such compounds to be sensitive biomarkers of the disease. The dedication of the Brady lab to NPC, and the discoveries that flowed therefrom, provided critical foundations for the current explosion of progress in this disease.


Asunto(s)
Proteínas Portadoras/genética , Colesterol/metabolismo , Glicoproteínas de Membrana/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Animales , Modelos Animales de Enfermedad , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Mutación , Proteína Niemann-Pick C1 , Oxiesteroles
9.
J Neurosci ; 35(21): 8091-106, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019327

RESUMEN

Niemann-Pick Type C1 (NPC1) disease is a rare neurovisceral, cholesterol-sphingolipid lysosomal storage disorder characterized by ataxia, motor impairment, progressive intellectual decline, and dementia. The most prevalent mutation, NPC1(I1061T), encodes a misfolded protein with a reduced half-life caused by ER-associated degradation. Therapies directed at stabilization of the mutant NPC1 protein reduce cholesterol storage in fibroblasts but have not been tested in vivo because of lack of a suitable animal model. Whereas the prominent features of human NPC1 disease are replicated in the null Npc1(-/-) mouse, this model is not amenable to examining proteostatic therapies. The objective of the present study was to develop an NPC1 I1061T knock-in mouse in which to test proteostatic therapies. Compared with the Npc1(-/-) mouse, this Npc1(tm(I1061T)Dso) model displays a less severe, delayed form of NPC1 disease with respect to weight loss, decreased motor coordination, Purkinje cell death, lipid storage, and premature death. The murine NPC1(I1061T) protein has a reduced half-life in vivo, consistent with protein misfolding and rapid ER-associated degradation, and can be stabilized by histone deacetylase inhibition. This novel mouse model faithfully recapitulates human NPC1 disease and provides a powerful tool for preclinical evaluation of therapies targeting NPC1 protein variants with compromised stability.


Asunto(s)
Alelos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Glicoproteínas de Membrana/genética , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología , Animales , Células Cultivadas , Femenino , Técnicas de Sustitución del Gen/métodos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Niemann-Pick C1 , Prevalencia
10.
Brain ; 137(Pt 12): 3300-18, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25270989

RESUMEN

Autophagy, the major lysosomal pathway for the turnover of intracellular organelles is markedly impaired in neurons in Alzheimer's disease and Alzheimer mouse models. We have previously reported that severe lysosomal and amyloid neuropathology and associated cognitive deficits in the TgCRND8 Alzheimer mouse model can be ameliorated by restoring lysosomal proteolytic capacity and autophagy flux via genetic deletion of the lysosomal protease inhibitor, cystatin B. Here we present evidence that macroautophagy is a significant pathway for lipid turnover, which is defective in TgCRND8 brain where lipids accumulate as membranous structures and lipid droplets within giant neuronal autolysosomes. Levels of multiple lipid species including several sphingolipids (ceramide, ganglioside GM3, GM2, GM1, GD3 and GD1a), cardiolipin, cholesterol and cholesteryl esters are elevated in autophagic vacuole fractions and lysosomes isolated from TgCRND8 brain. Lipids are localized in autophagosomes and autolysosomes by double immunofluorescence analyses in wild-type mice and colocalization is increased in TgCRND8 mice where abnormally abundant GM2 ganglioside-positive granules are detected in neuronal lysosomes. Cystatin B deletion in TgCRND8 significantly reduces the number of GM2-positive granules and lowers the levels of GM2 and GM3 in lysosomes, decreases lipofuscin-related autofluorescence, and eliminates giant lipid-containing autolysosomes while increasing numbers of normal-sized autolysosomes/lysosomes with reduced content of undigested components. These findings have identified macroautophagy as a previously unappreciated route for delivering membrane lipids to lysosomes for turnover, a function that has so far been considered to be mediated exclusively through the endocytic pathway, and revealed that autophagic-lysosomal dysfunction in TgCRND8 brain impedes lysosomal turnover of lipids as well as proteins. The amelioration of lipid accumulation in TgCRND8 by removing cystatin B inhibition on lysosomal proteases suggests that enhancing lysosomal proteolysis improves the overall environment of the lysosome and its clearance functions, which may be possibly relevant to a broader range of lysosomal disorders beyond Alzheimer's disease.


Asunto(s)
Autofagia/fisiología , Encéfalo/metabolismo , Metabolismo de los Lípidos/fisiología , Lisosomas/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Animales , Autofagia/genética , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Proteolisis
11.
J Neurosci ; 33(26): 10815-27, 2013 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-23804102

RESUMEN

Protein aggregates are a common pathological feature of neurodegenerative diseases and several lysosomal diseases, but it is currently unclear what aggregates represent for pathogenesis. Here we report the accumulation of intraneuronal aggregates containing the macroautophagy adapter proteins p62 and NBR1 in the neurodegenerative lysosomal disease late-infantile neuronal ceroid lipofuscinosis (CLN2 disease). CLN2 disease is caused by a deficiency in the lysosomal enzyme tripeptidyl peptidase I, which results in aberrant lysosomal storage of catabolites, including the subunit c of mitochondrial ATP synthase (SCMAS). In an effort to define the role of aggregates in CLN2, we evaluated p62 and NBR1 accumulation in the CNS of Cln2(-/-) mice. Although increases in p62 and NBR1 often suggest compromised degradative mechanisms, we found normal ubiquitin-proteasome system function and only modest inefficiency in macroautophagy late in disease. Importantly, we identified that SCMAS colocalizes with p62 in extra-lysosomal aggregates in Cln2(-/-) neurons in vivo. This finding is consistent with SCMAS being released from lysosomes, an event known as lysosomal membrane permeability (LMP). We predicted that LMP and storage release from lysosomes results in the sequestration of this material as cytosolic aggregates by p62 and NBR1. Notably, LMP induction in primary neuronal cultures generates p62-positive aggregates and promotes p62 localization to lysosomal membranes, supporting our in vivo findings. We conclude that LMP is a previously unrecognized pathogenic event in CLN2 disease that stimulates cytosolic aggregate formation. Furthermore, we offer a novel role for p62 in response to LMP that may be relevant for other diseases exhibiting p62 accumulation.


Asunto(s)
Lisosomas/metabolismo , Lipofuscinosis Ceroideas Neuronales/metabolismo , Neuronas/metabolismo , Aminopeptidasas/genética , Animales , Western Blotting , Células Cultivadas , Citosol/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/metabolismo , Membranas/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Proteínas de Complejo Poro Nuclear/metabolismo , Permeabilidad , Proteínas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina Proteasas/genética , Tripeptidil Peptidasa 1
12.
Hum Mol Genet ; 21(16): 3632-46, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22619379

RESUMEN

Niemann-Pick disease type C (NPC) is a lysosomal storage disorder characterized by liver disease and progressive neurodegeneration. Deficiency of either NPC1 or NPC2 leads to the accumulation of cholesterol and glycosphingolipids in late endosomes and early lysosomes. In order to identify pathological mechanisms underlying NPC and uncover potential biomarkers, we characterized liver gene expression changes in an Npc1 mouse model at six ages spanning the pathological progression of the disease. We identified altered gene expression at all ages, including changes in asymptomatic, 1-week-old mice. Biological pathways showing early altered gene expression included: lipid metabolism, cytochrome P450 enzymes involved in arachidonic acid and drug metabolism, inflammation and immune responses, mitogen-activated protein kinase and G-protein signaling, cell cycle regulation, cell adhesion and cytoskeleton remodeling. In contrast, apoptosis and oxidative stress appeared to be late pathological processes. To identify potential biomarkers that could facilitate monitoring of disease progression, we focused on a subset of 103 differentially expressed genes that encode secreted proteins. Further analysis identified two secreted proteins with increased serum levels in NPC1 patients: galectin-3 (LGALS3), a pro-inflammatory molecule, and cathepsin D (CTSD), a lysosomal aspartic protease. Elevated serum levels of both proteins correlated with neurological disease severity and appeared to be specific for NPC1. Expression of Lgals3 and Ctsd was normalized following treatment with 2-hydroxypropyl-ß-cyclodextrin, a therapy that reduces pathological findings and significantly increases Npc1(-/-) survival. Both LGALS3 and CTSD have the potential to aid in diagnosis and serve as biomarkers to monitor efficacy in therapeutic trials.


Asunto(s)
Biomarcadores/sangre , Catepsina D/sangre , Galectina 3/sangre , Hígado/fisiología , Enfermedad de Niemann-Pick Tipo C/sangre , Enfermedad de Niemann-Pick Tipo C/genética , 2-Hidroxipropil-beta-Ciclodextrina , Adolescente , Factores de Edad , Animales , Estudios de Casos y Controles , Catepsina D/genética , Niño , Preescolar , Sistema Enzimático del Citocromo P-450/genética , Modelos Animales de Enfermedad , Femenino , Galectina 3/genética , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular , Metabolismo de los Lípidos/genética , Hígado/patología , Masculino , Ratones , Ratones Mutantes , Análisis por Micromatrices , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo C/mortalidad , Proteínas/genética , Proteínas/metabolismo , Tasa de Supervivencia , Transcriptoma , beta-Ciclodextrinas/farmacología
13.
J Neurodev Disord ; 16(1): 10, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38491427

RESUMEN

We describe a multidisciplinary teamwork approach known as "Operation IDD Gene Team" developed by the Rose F. Kennedy Intellectual and Developmental Disabilities Research Center (RFK IDDRC) at the Albert Einstein College of Medicine. This initiative brings families affected by rare genetic diseases that cause intellectual and developmental disability together with physicians, basic scientists, and their trainees. At team meetings, family members share their child's medical and personal history, physicians describe the broader clinical consequences of the condition, and scientists provide accessible tutorials focused on the fundamental biology of relevant genes. When appropriate, possible treatment approaches are also discussed. The outcomes of team meetings have been overwhelmingly positive, with families not only expressing deep gratitude, but also becoming empowered to establish foundations dedicated to their child's specific condition. Physicians, and in particular the scientists and their trainees, have gained a deeper understanding of challenges faced by affected families, broadening their perspective on how their research can extend beyond the laboratory. Remarkably, research by the scientists following the Gene Team meetings have often included focus on the actual gene variants exhibited by the participating children. As these investigations progress and newly created foundations expand their efforts, national as well as international collaborations are forged. These developments emphasize the importance of rare diseases as windows into previously unexplored molecular and cellular processes, which can offer fresh insights into both normal function as well as more common diseases. Elucidating the mechanisms of and treatments for rare and ultra-rare diseases thus has benefits for all involved-families, physicians, and scientists and their trainees, as well as the broader medical community. While the RFK IDDRC's Operation IDD Gene Team program has focused on intellectual disabilities affecting children, we believe it has the potential to be applied to rare genetic diseases impacting individuals of any age and encompassing a wide variety of developmental disorders affecting multiple organ systems.


Asunto(s)
Encefalopatías , Medicina de Precisión , Niño , Humanos , Enfermedades Raras/genética , Enfermedades Raras/terapia
14.
J Lipid Res ; 54(10): 2800-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23881911

RESUMEN

Niemann-Pick type C (NPC)1 is a rare neurodegenerative disease for which treatment options are limited. A major barrier to development of effective treatments has been the lack of validated biomarkers to monitor disease progression or serve as outcome measures in clinical trials. Using targeted metabolomics to exploit the complex lipid storage phenotype that is the hallmark of NPC1 disease, we broadly surveyed Npc1(-/-) mouse tissues and identified elevated species across multiple sphingolipid classes that increased with disease progression. There was a striking accumulation of sphingoid bases, monohexosylceramides (MCs), and GM2 gangliosides in liver, and sphingoid bases and GM2 and GM3 gangliosides in brain. These lipids were modestly decreased following miglustat treatment, but markedly decreased in response to treatment with 2-hydroxypropyl-ß-cyclodextrin (HP-ß-CD), two drugs that have shown efficacy in NPC1 animal models. Extending these studies to human subjects led to identification of sphingolipid classes that were significantly altered in the plasma of NPC1 patients. Plasma MCs and ceramides were elevated, whereas sphingoid bases were reduced in NPC1 subjects. Intervention with miglustat in NPC1 patients was accompanied by striking alterations in plasma (reductions in GM1 and GM3 gangliosides) and cerebrospinal fluid (CSF) (increased MCs) sphingolipids. Similar alterations were observed in the CSF from the NPC1 feline model following HP-ß-CD treatment. Our findings suggest that these lipid biomarkers may prove useful as outcome measures for monitoring efficacy of therapy in clinical trials.


Asunto(s)
Enfermedad de Niemann-Pick Tipo C/sangre , Esfingolípidos/sangre , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , 2-Hidroxipropil-beta-Ciclodextrina , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Gatos , Cromatografía Líquida de Alta Presión , Evaluación Preclínica de Medicamentos , Femenino , Gangliósidos/sangre , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Especificidad de Órganos , Esfingolípidos/líquido cefalorraquídeo , Sulfoglicoesfingolípidos/sangre , Espectrometría de Masas en Tándem , beta-Ciclodextrinas/farmacología , beta-Ciclodextrinas/uso terapéutico
15.
J Biol Chem ; 287(47): 39349-60, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23035117

RESUMEN

Niemann-Pick disease type C (NPC) and Wolman disease are two members of a family of storage disorders caused by mutations of genes encoding lysosomal proteins. Deficiency in function of either the NPC1 or NPC2 protein in NPC disease or lysosomal acid lipase in Wolman disease results in defective cellular cholesterol trafficking. Lysosomal accumulation of cholesterol and enlarged lysosomes are shared phenotypic characteristics of both NPC and Wolman cells. Utilizing a phenotypic screen of an approved drug collection, we found that δ-tocopherol effectively reduced lysosomal cholesterol accumulation, decreased lysosomal volume, increased cholesterol efflux, and alleviated pathological phenotypes in both NPC1 and Wolman fibroblasts. Reduction of these abnormalities may be mediated by a δ-tocopherol-induced intracellular Ca(2+) response and subsequent enhancement of lysosomal exocytosis. Consistent with a general mechanism for reduction of lysosomal lipid accumulation, we also found that δ-tocopherol reduces pathological phenotypes in patient fibroblasts from other lysosomal storage diseases, including NPC2, Batten (ceroid lipofuscinosis, neuronal 2, CLN2), Fabry, Farber, Niemann-Pick disease type A, Sanfilippo type B (mucopolysaccharidosis type IIIB, MPSIIIB), and Tay-Sachs. Our data suggest that regulated exocytosis may represent a potential therapeutic target for reduction of lysosomal storage in this class of diseases.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Colesterol/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Tocoferoles/farmacología , Enfermedad de Wolman/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Cricetinae , Exocitosis/efectos de los fármacos , Humanos , Lisosomas/patología , Enfermedad de Niemann-Pick Tipo C/patología , Tripeptidil Peptidasa 1 , Enfermedad de Wolman/patología
16.
J Inherit Metab Dis ; 36(3): 491-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23412751

RESUMEN

Niemann-Pick type C disease is an inherited autosomal recessive neurodegenerative disorder characterised by the accumulation of unesterified cholesterol and sphingolipids within the endosomal/lysosomal compartments. It has been observed that the administration of hydroxypropyl-ß-cyclodextrin (HPBCD) delays onset of clinical symptoms and reduces accumulation of cholesterol and gangliosides within neuronal cells. It was assumed that HPBCD exerts its action by readily entering the CNS and directly interacting with neurones and other brain cells to facilitate removal of stored cholesterol from the late endosomal/lysosomal compartment. Here, we present evidence that refutes this hypothesis. We use two well established techniques for accurately measuring brain uptake of solutes from blood and show that there is no significant crossing of HPBCD into the brain. The two techniques are brain in situ perfusion and intraperitoneal injection followed by multi-time-point regression analysis. Neither study demonstrates significant, time-dependent uptake of HPBCD in either adult or neonatal mice. However, the volume of distribution available to HPBCD (0.113 ± 0.010 ml/g) exceeds the accepted values for plasma and vascular volume of the brain. In fact, it is nearly three times larger than that for sucrose (0.039 ± 0.006 ml/g). We propose that this indicates cell surface binding of HPBCD to the endothelium of the cerebral vasculature and may provide a mechanism for the mobilisation and clearance of cholesterol from the CNS.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Colesterol/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedades de Niemann-Pick/tratamiento farmacológico , beta-Ciclodextrinas/uso terapéutico , 2-Hidroxipropil-beta-Ciclodextrina , Animales , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Inyecciones Intraperitoneales , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neuronas/patología , Proteína Niemann-Pick C1 , Enfermedades de Niemann-Pick/genética , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Perfusión , Proteínas/genética , Proteínas/metabolismo , beta-Ciclodextrinas/administración & dosificación
17.
Am J Pathol ; 179(2): 890-902, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21708114

RESUMEN

Niemann-Pick disease type C (NPC) is a severe neurovisceral lysosomal storage disorder caused by defects in NPC1 or NPC2 proteins. Although numerous studies support the primacy of cholesterol storage, neurons of double-mutant mice lacking both NPC1 and an enzyme required for synthesis of all complex gangliosides (ß1,4GalNAc transferase) have been reported to exhibit dramatically reduced cholesterol sequestration. Here we show that NPC2-deficient mice lacking this enzyme also exhibit reduced cholesterol, but that genetically restricting synthesis to only a-series gangliosides fully restores neuronal cholesterol storage to typical disease levels. Examining the subcellular locations of sequestered compounds in neurons lacking NPC1 or NPC2 by confocal microscopy revealed that cholesterol and the two principal storage gangliosides (GM2 and GM3) were not consistently co-localized within the same intracellular vesicles. To determine whether the lack of GM2 and GM3 co-localization was due to differences in synthetic versus degradative pathway expression, we generated mice lacking both NPC1 and lysosomal ß-galactosidase, and therefore unable to generate GM2 and GM3 in lysosomes. Double mutants lacked both gangliosides, indicating that each is the product of endosomal/lysosomal processing. Unexpectedly, GM1 accumulation in double mutants increased compared to single mutants consistent with a direct role for NPC1 in ganglioside salvage. These studies provide further evidence that NPC1 and NPC2 proteins participate in endosomal/lysosomal processing of both sphingolipids and cholesterol.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Gangliósidos/metabolismo , Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Animales , Péptidos y Proteínas de Señalización Intracelular , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/enzimología , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos , Mutación , Neuronas/metabolismo , Proteína Niemann-Pick C1 , Proteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , beta-Galactosidasa/genética
18.
Pediatr Res ; 71(1): 39-45, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22289849

RESUMEN

INTRODUCTION: Mucopolysaccharidosis VI (MPS-VI) is caused by a deficiency in N-acetylgalactosamine-4-sulfatase activity, resulting in lysosomal accumulation of partially degraded glycosaminoglycans (GAGs). Compressive myelopathy in early-onset MPS-VI patients has been partly attributed to thickening of the dura mater following engorgement with GAG. In this study, we therefore tested whether the dural abnormalities could be prevented in a feline model of the disorder. RESULTS: All intrathecal injections (IT-INJs) were well tolerated. MPS-VI cats treated with IT-INJ of recombinant human N-acetylgalactosamine-4-sulfatase (rhASB) exhibited reduced vacuolation in the dural fibroblasts, diminished levels of sulfated-N-acetylhexosamine (HNAc(+S)) in the cerebrospinal fluid (CSF) and no hind-limb paresis. Serum anti-rhASB antibodies remained low in MPS-VI cats treated with intravenous enzyme replacement therapy (IV-ERT) and increased slightly in normal cats treated with IT-INJ of rhASB alone. Anti-rhASB antibodies in CSF remained undetectable. DISCUSSION: These data indicate that repeated IT-INJ of rhASB can safely prevent GAG storage in MPS-VI dura. METHODS: Cats were assigned to three groups: (i) receiving weekly IV-ERT of rhASB from birth plus six monthly IT-INJs of rhASB from age 2 months; (ii) receiving six monthly IT-INJs of vehicle; or (iii) untreated. Additional normal cats received five fortnightly IT-INJs of rhASB or vehicle alone.


Asunto(s)
Duramadre/patología , Glicosaminoglicanos/metabolismo , Mucopolisacaridosis VI/tratamiento farmacológico , N-Acetilgalactosamina-4-Sulfatasa/administración & dosificación , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico , Animales , Gatos , Modelos Animales de Enfermedad , Duramadre/metabolismo , Humanos , Inyecciones Espinales , Mucopolisacaridosis VI/enzimología , Mucopolisacaridosis VI/patología , Mucopolisacaridosis VI/fisiopatología , N-Acetilgalactosamina-4-Sulfatasa/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
19.
Brain ; 134(Pt 11): 3369-83, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21964919

RESUMEN

Mutations in solute carrier family 9 isoform 6 on chromosome Xq26.3 encoding sodium-hydrogen exchanger 6, a protein mainly expressed in early and recycling endosomes are known to cause a complex and slowly progressive degenerative human neurological disease. Three resulting phenotypes have so far been reported: an X-linked Angelman syndrome-like condition, Christianson syndrome and corticobasal degeneration with tau deposition, with each characterized by severe intellectual disability, epilepsy, autistic behaviour and ataxia. Hypothesizing that a sodium-hydrogen exchanger 6 deficiency would most likely disrupt the endosomal-lysosomal system of neurons, we examined Slc9a6 knockout mice with tissue staining and related techniques commonly used to study lysosomal storage disorders. As a result, we found that sodium-hydrogen exchanger 6 depletion leads to abnormal accumulation of GM2 ganglioside and unesterified cholesterol within late endosomes and lysosomes of neurons in selective brain regions, most notably the basolateral nuclei of the amygdala, the CA3 and CA4 regions and dentate gyrus of the hippocampus and some areas of cerebral cortex. In these select neuronal populations, histochemical staining for ß-hexosaminidase activity, a lysosomal enzyme involved in the degradation of GM2 ganglioside, was undetectable. Neuroaxonal dystrophy similar to that observed in lysosomal disease was observed in the cerebellum and was accompanied by a marked and progressive loss of Purkinje cells, particularly in those lacking the expression of Zebrin II. On behavioural testing, Slc9a6 knockout mice displayed a discrete clinical phenotype attributable to motor hyperactivity and cerebellar dysfunction. Importantly, these findings show that sodium-hydrogen exchanger 6 loss of function in the Slc9a6-targeted mouse model leads to compromise of endosomal-lysosomal function similar to lysosomal disease and to conspicuous neuronal abnormalities in specific brain regions, which in concert could provide a unified explanation for the cellular and clinical phenotypes in humans with SLC9A6 mutations.


Asunto(s)
Síndrome de Angelman/genética , Encéfalo/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Síndrome de Angelman/metabolismo , Animales , Ratones , Ratones Noqueados , Neuronas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo
20.
Mol Genet Metab ; 103(3): 268-74, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21482164

RESUMEN

The storage disorder mucopolysaccharidosis type I (MPS I) is caused by a deficiency in lysosomal α-L-iduronidase activity. The inability to degrade glycosaminoglycans (GAG) results in lysosomal accumulation and widespread tissue lesions. Many symptoms of MPS I are amenable to treatment with recombinant human α-L-iduronidase (rhIDU), however, peripherally administered rhIDU does not cross the blood-brain barrier and has no beneficial effects in the central nervous system (CNS). A feline model of MPS I was used to evaluate the CNS effects of rhIDU following repeated intrathecal (IT) administration. Twelve animals were randomized into four groups based on the time of euthanasia and tissue evaluation following three repeat IT administrations of 0.1 mg/kg rhIDU or placebo on Study Days 1, 4 or 5, and 9. Two days after the final IT injection, the mean tissue α-L-iduronidase (IDU) activity in the brains of the two treated animals were approximately 3-times higher (50.1 and 54.9 U/mg protein) than the activity found in normal cat brains (mean of 18.3 U/mg), and remained higher than untreated MPSI brain at 1 month (2.4 and 4.1 U/mg protein) before returning to near-baseline levels after 2 months. This activity corresponded with decreased brain GAG concentrations after 2 days (1.4 and 2.0 µg/mg) and 1 month (0.9 and 1.1 µg/mg) which approached levels observed in normal animals (0.7 µg/mg). Attenuation of GAG, gangliosides GM2 and GM3, and cholesterol reaccumulation was identified at both two days and one month following final IT injection. No adverse effects attributable to IT rhIDU administration were observed. IT rhIDU may be an effective means for providing enzyme replacement therapy for the central manifestations of MPS I.


Asunto(s)
Terapia de Reemplazo Enzimático , Iduronidasa/farmacocinética , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/enzimología , Proteínas Recombinantes/farmacocinética , Animales , Anticuerpos/sangre , Anticuerpos/líquido cefalorraquídeo , Encéfalo/metabolismo , Encéfalo/patología , Gatos , Terapia de Reemplazo Enzimático/efectos adversos , Femenino , Glicosaminoglicanos/metabolismo , Hexosaminidasas/metabolismo , Humanos , Iduronidasa/administración & dosificación , Iduronidasa/efectos adversos , Inyecciones Espinales , Masculino , Mucopolisacaridosis I/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA