Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 16(730): eadf1691, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38232139

RESUMEN

Glycogen synthase 1 (GYS1), the rate-limiting enzyme in muscle glycogen synthesis, plays a central role in energy homeostasis and has been proposed as a therapeutic target in multiple glycogen storage diseases. Despite decades of investigation, there are no known potent, selective small-molecule inhibitors of this enzyme. Here, we report the preclinical characterization of MZ-101, a small molecule that potently inhibits GYS1 in vitro and in vivo without inhibiting GYS2, a related isoform essential for synthesizing liver glycogen. Chronic treatment with MZ-101 depleted muscle glycogen and was well tolerated in mice. Pompe disease, a glycogen storage disease caused by mutations in acid α glucosidase (GAA), results in pathological accumulation of glycogen and consequent autophagolysosomal abnormalities, metabolic dysregulation, and muscle atrophy. Enzyme replacement therapy (ERT) with recombinant GAA is the only approved treatment for Pompe disease, but it requires frequent infusions, and efficacy is limited by suboptimal skeletal muscle distribution. In a mouse model of Pompe disease, chronic oral administration of MZ-101 alone reduced glycogen buildup in skeletal muscle with comparable efficacy to ERT. In addition, treatment with MZ-101 in combination with ERT had an additive effect and could normalize muscle glycogen concentrations. Biochemical, metabolomic, and transcriptomic analyses of muscle tissue demonstrated that lowering of glycogen concentrations with MZ-101, alone or in combination with ERT, corrected the cellular pathology in this mouse model. These data suggest that substrate reduction therapy with GYS1 inhibition may be a promising therapeutic approach for Pompe disease and other glycogen storage diseases.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Ratones , Animales , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Glucógeno Sintasa/metabolismo , Glucógeno Sintasa/farmacología , Ratones Noqueados , Glucógeno/metabolismo , Músculo Esquelético/metabolismo , Terapia de Reemplazo Enzimático/métodos
2.
Cell Rep ; 40(1): 111041, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35793618

RESUMEN

Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.


Asunto(s)
Glucógeno Sintasa , Glucogenólisis , Glicoproteínas , Glucosiltransferasas/metabolismo , Glucógeno/metabolismo , Glucógeno Sintasa/metabolismo , Glicoproteínas/metabolismo , Humanos
3.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35226042

RESUMEN

Delivery of biotherapeutics across the blood-brain barrier (BBB) is a challenge. Many approaches fuse biotherapeutics to platforms that bind the transferrin receptor (TfR), a brain endothelial cell target, to facilitate receptor-mediated transcytosis across the BBB. Here, we characterized the pharmacological behavior of two distinct TfR-targeted platforms fused to iduronate 2-sulfatase (IDS), a lysosomal enzyme deficient in mucopolysaccharidosis type II (MPS II), and compared the relative brain exposures and functional activities of both approaches in mouse models. IDS fused to a moderate-affinity, monovalent TfR-binding enzyme transport vehicle (ETV:IDS) resulted in widespread brain exposure, internalization by parenchymal cells, and significant substrate reduction in the CNS of an MPS II mouse model. In contrast, IDS fused to a standard high-affinity bivalent antibody (IgG:IDS) resulted in lower brain uptake, limited biodistribution beyond brain endothelial cells, and reduced brain substrate reduction. These results highlight important features likely to impact the clinical development of TfR-targeting platforms in MPS II and potentially other CNS diseases.


Asunto(s)
Iduronato Sulfatasa , Mucopolisacaridosis II , Receptores de Transferrina , Proteínas Recombinantes de Fusión , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Iduronato Sulfatasa/metabolismo , Iduronato Sulfatasa/farmacología , Lisosomas/metabolismo , Ratones , Mucopolisacaridosis II/metabolismo , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Distribución Tisular
4.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751752

RESUMEN

We recently developed a blood-brain barrier (BBB)-penetrating enzyme transport vehicle (ETV) fused to the lysosomal enzyme iduronate 2-sulfatase (ETV:IDS) and demonstrated its ability to reduce glycosaminoglycan (GAG) accumulation in the brains of a mouse model of mucopolysaccharidosis (MPS) II. To accurately quantify GAGs, we developed a plate-based high-throughput enzymatic digestion assay coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to simultaneously measure heparan sulfate and dermatan sulfate derived disaccharides in tissue, cerebrospinal fluid (CSF) and individual cell populations isolated from mouse brain. The method offers ultra-high sensitivity enabling quantitation of specific GAG species in as low as 100,000 isolated neurons and a low volume of CSF. With an LOD at 3 ng/mL and LLOQs at 5-10 ng/mL, this method is at least five times more sensitive than previously reported approaches. Our analysis demonstrated that the accumulation of CSF and brain GAGs are in good correlation, supporting the potential use of CSF GAGs as a surrogate biomarker for brain GAGs. The bioanalytical method was qualified through the generation of standard curves in matrix for preclinical studies of CSF, demonstrating the feasibility of this assay for evaluating therapeutic effects of ETV:IDS in future studies and applications in a wide variety of MPS disorders.


Asunto(s)
Biomarcadores/metabolismo , Glicosaminoglicanos/aislamiento & purificación , Iduronato Sulfatasa/genética , Mucopolisacaridosis II/diagnóstico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Cromatografía Liquida , Dermatán Sulfato/farmacología , Disacáridos/química , Modelos Animales de Enfermedad , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/farmacología , Humanos , Iduronato Sulfatasa/metabolismo , Ratones , Mucopolisacaridosis II/genética , Mucopolisacaridosis II/patología , Espectrometría de Masas en Tándem
5.
Sci Transl Med ; 12(545)2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32461331

RESUMEN

Most lysosomal storage diseases (LSDs) involve progressive central nervous system (CNS) impairment, resulting from deficiency of a lysosomal enzyme. Treatment of neuronopathic LSDs remains a considerable challenge, as approved intravenously administered enzyme therapies are ineffective in modifying CNS disease because they do not effectively cross the blood-brain barrier (BBB). We describe a therapeutic platform for increasing the brain exposure of enzyme replacement therapies. The enzyme transport vehicle (ETV) is a lysosomal enzyme fused to an Fc domain that has been engineered to bind to the transferrin receptor, which facilitates receptor-mediated transcytosis across the BBB. We demonstrate that ETV fusions containing iduronate 2-sulfatase (ETV:IDS), the lysosomal enzyme deficient in mucopolysaccharidosis type II, exhibited high intrinsic activity and degraded accumulated substrates in both IDS-deficient cell and in vivo models. ETV substantially improved brain delivery of IDS in a preclinical model of disease, enabling enhanced cellular distribution to neurons, astrocytes, and microglia throughout the brain. Improved brain exposure for ETV:IDS translated to a reduction in accumulated substrates in these CNS cell types and peripheral tissues and resulted in a complete correction of downstream disease-relevant pathologies in the brain, including secondary accumulation of lysosomal lipids, perturbed gene expression, neuroinflammation, and neuroaxonal damage. These data highlight the therapeutic potential of the ETV platform for LSDs and provide preclinical proof of concept for TV-enabled therapeutics to treat CNS diseases more broadly.


Asunto(s)
Barrera Hematoencefálica , Iduronato Sulfatasa , Animales , Encéfalo , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático , Lisosomas , Ratones
6.
Nat Commun ; 9(1): 330, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29362376

RESUMEN

Psychoactive compounds such as chloroquine and amphetamine act by dissipating the pH gradient across intracellular membranes, but the physiological mechanisms that normally regulate organelle pH remain poorly understood. Interestingly, recent human genetic studies have implicated the endosomal Na+/H+ exchanger NHE9 in both autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD). Plasma membrane NHEs regulate cytosolic pH, but the role of intracellular isoforms has remained unclear. We now find that inactivation of NHE9 in mice reproduces behavioral features of ASD including impaired social interaction, repetitive behaviors, and altered sensory processing. Physiological characterization reveals hyperacidic endosomes, a cell-autonomous defect in glutamate receptor expression and impaired neurotransmitter release due to a defect in presynaptic Ca2+ entry. Acute inhibition of synaptic vesicle acidification rescues release but without affecting the primary defect due to loss of NHE9.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno del Espectro Autista/metabolismo , Calcio/metabolismo , Endosomas/metabolismo , Neuronas/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Animales , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Conducta Animal , Modelos Animales de Enfermedad , Electroencefalografía , Endosomas/patología , Femenino , Expresión Génica , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Noqueados , Neuronas/patología , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Cultivo Primario de Células , Intercambiadores de Sodio-Hidrógeno/deficiencia , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/patología
7.
Nat Neurosci ; 14(10): 1285-92, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21874016

RESUMEN

The amount of neurotransmitter stored in a single synaptic vesicle can determine the size of the postsynaptic response, but the factors that regulate vesicle filling are poorly understood. A proton electrochemical gradient (Δµ(H+)) generated by the vacuolar H(+)-ATPase drives the accumulation of classical transmitters into synaptic vesicles. The chemical component of Δµ(H+) (ΔpH) has received particular attention for its role in the vesicular transport of cationic transmitters as well as in protein sorting and degradation. Thus, considerable work has addressed the factors that promote ΔpH. However, synaptic vesicle uptake of the principal excitatory transmitter glutamate depends on the electrical component of Δµ(H+) (Δψ). We found that rat brain synaptic vesicles express monovalent cation/H(+) exchange activity that converts ΔpH into Δψ, and that this promotes synaptic vesicle filling with glutamate. Manipulating presynaptic K(+) at a glutamatergic synapse influenced quantal size, indicating that synaptic vesicle K(+)/H(+) exchange regulates glutamate release and synaptic transmission.


Asunto(s)
Potasio/metabolismo , Terminales Presinápticos/fisiología , Sinapsis/fisiología , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Proteínas de Artrópodos , Ácido Aspártico/farmacocinética , Transporte Biológico , Fenómenos Biofísicos/efectos de los fármacos , Encéfalo/citología , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Cationes/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Gluconatos/farmacología , Ácido Glutámico/farmacocinética , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Ionóforos/farmacología , Potencial de la Membrana Mitocondrial , Monensina/farmacología , Oligopéptidos/farmacología , Terminales Presinápticos/efectos de los fármacos , Cintigrafía , Ratas , Ratas Wistar , Isótopos de Sodio/farmacocinética , Sinapsis/diagnóstico por imagen , Sinapsis/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Sinaptosomas/ultraestructura , Tritio/farmacocinética
8.
J Biol Chem ; 285(14): 10415-23, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19955177

RESUMEN

Prion-infected cells accumulate a heterogeneous population of aberrantly folded PrP conformers, including the disease-causing isoform (PrP(Sc)). We found that specific chemicals can modulate the levels of various PrP conformers in cultured cells. Positively charged polyamidoamines (dendrimers) eliminated protease-resistant (r) PrP(Sc) from prion-infected cells and induced the formation of insoluble, protease-sensitive PrP aggregates (designated PrP(A)). Larger, positively charged polyamidoamines more efficaciously induced the formation of PrP(A) and cleared rPrP(Sc), whereas negatively charged polyamidoamines neither induced PrP(A) nor cleared rPrP(Sc). Although the biochemical properties of PrP(A) were shown to be similar to protease-sensitive (s) PrP(Sc), bioassays of PrP(A) indicated that it is not infectious. Our studies argue that PrP(A) represents an aggregated PrP species that is off-pathway relative to the formation of rPrP(Sc). It remains to be established whether the formation of PrP(A) inhibits the formation of rPrP(Sc) by sequestering PrP(C) in the form of benign, insoluble aggregates.


Asunto(s)
Dendrímeros/farmacología , Nylons/farmacología , Proteínas PrPC/química , Proteínas PrPSc/química , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cromatografía en Gel , Dimerización , Endopeptidasas/metabolismo , Ratones , Ratones Transgénicos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Conformación Proteica , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
9.
Proc Natl Acad Sci U S A ; 104(46): 17971-6, 2007 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17989223

RESUMEN

The phenotypic effect of prions on host cells is influenced by the physical properties of the prion strain and its level of accumulation. In mammalian cell cultures, prion accumulation is determined by the interplay between de novo prion formation, catabolism, cell division, and horizontal cell-to-cell transmission. Understanding this dynamic enables the analytical modeling of protein-based heritability and infectivity. Here, we quantitatively measured these competing effects in a subline of neuroblastoma (N2a) cells and propose a concordant reaction mechanism to explain the kinetics of prion propagation. Our results show that cell division leads to a predictable reduction in steady-state prion levels but not to complete clearance. Scrapie-infected N2a cells were capable of accumulating different steady-state levels of prions, dictated partly by the rate of cell division. We also show that prions in this subline of N2a cells are transmitted primarily from mother to daughter cells, rather than horizontal cell-to-cell transmission. We quantitatively modeled our kinetic results based on a mechanism that assumes a subpopulation of prions is capable of self-catalysis, and the levels of this subpopulation reach saturation in fully infected cells. Our results suggest that the apparent effectiveness of antiprion compounds in culture may be strongly influenced by the growth phase of the target cells.


Asunto(s)
División Celular , Priones/metabolismo , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Cinética , Modelos Biológicos , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo
10.
Cogn Behav Ther ; 33(2): 60-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15279311

RESUMEN

Effects of the September 11, 2001 terrorist attacks in the USA were investigated in 25 patients with obsessive compulsive disorder and 27 normal controls 4-6 months after the attacks. Participants completed a 15-item questionnaire to retrospectively assess changes in mood, cognition, behavior and somatic complaints since September 11, 2001. Overall, both patients with obsessive compulsive disorder and normal controls reported minor changes in mood, behavior and somatic complaints. However, normal controls reported severe to extreme initial impact, slightly more cognitive symptoms (uncertainty about the future, intrusive recollections and greater desire to be with loved ones) and a slightly greater degree of overall impact on emotion and behavior at 1, 2 and 3 months after September 11 than did patients with obsessive compulsive disorder. Results support previous research that has found a relatively minor lasting impact of September 11 on both clinical and normal populations. Differences in cognition and coping mechanisms between normal controls and patients with obsessive compulsive disorder are proposed.


Asunto(s)
Trastorno Obsesivo Compulsivo/psicología , Terrorismo/psicología , Adulto , Análisis de Varianza , Canadá/epidemiología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Trastorno Obsesivo Compulsivo/epidemiología , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...