Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 600(7888): 269-273, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34789878

RESUMEN

The brain is the seat of body weight homeostasis. However, our inability to control the increasing prevalence of obesity highlights a need to look beyond canonical feeding pathways to broaden our understanding of body weight control1-3. Here we used a reverse-translational approach to identify and anatomically, molecularly and functionally characterize a neural ensemble that promotes satiation. Unbiased, task-based functional magnetic resonance imaging revealed marked differences in cerebellar responses to food in people with a genetic disorder characterized by insatiable appetite. Transcriptomic analyses in mice revealed molecularly and topographically -distinct neurons in the anterior deep cerebellar nuclei (aDCN) that are activated by feeding or nutrient infusion in the gut. Selective activation of aDCN neurons substantially decreased food intake by reducing meal size without compensatory changes to metabolic rate. We found that aDCN activity terminates food intake by increasing striatal dopamine levels and attenuating the phasic dopamine response to subsequent food consumption. Our study defines a conserved satiation centre that may represent a novel therapeutic target for the management of excessive eating, and underscores the utility of a 'bedside-to-bench' approach for the identification of neural circuits that influence behaviour.


Asunto(s)
Mantenimiento del Peso Corporal/genética , Mantenimiento del Peso Corporal/fisiología , Cerebelo/fisiología , Alimentos , Biosíntesis de Proteínas , Genética Inversa , Respuesta de Saciedad/fisiología , Adulto , Animales , Regulación del Apetito/genética , Regulación del Apetito/fisiología , Núcleos Cerebelosos/citología , Núcleos Cerebelosos/fisiología , Cerebelo/citología , Señales (Psicología) , Dopamina/metabolismo , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Femenino , Homeostasis , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Neostriado/metabolismo , Neuronas/fisiología , Obesidad/genética , Filosofía , Adulto Joven
2.
Mol Psychiatry ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38503924

RESUMEN

Decades of psychosis research highlight the prevalence and the clinical significance of negative emotions, such as fear and anxiety. Translational evidence demonstrates the pivotal role of the amygdala in fear and anxiety. However, most of these approaches have used hypothesis-driven analyses with predefined regions of interest. A data-driven analysis may provide a complimentary, unbiased approach to identifying brain correlates of fear and anxiety. The aim of the current study was to identify the brain basis of fear and anxiety in early psychosis and controls using a data-driven approach. We analyzed data from the Human Connectome Project for Early Psychosis, a multi-site study of 125 people with psychosis and 58 controls with resting-state fMRI and clinical characterization. Multivariate pattern analysis of whole-connectome data was used to identify shared and psychosis-specific brain correlates of fear and anxiety using the NIH Toolbox Fear-Affect and Fear-Somatic Arousal scales. We then examined clinical correlations of Fear-Affect scores and connectivity patterns. Individuals with psychosis had higher levels of Fear-Affect scores than controls (p < 0.05). The data-driven analysis identified a cluster encompassing the amygdala and hippocampus where connectivity was correlated with Fear-Affect score (p < 0.005) in the entire sample. The strongest correlate of Fear-Affect was between this cluster and the anterior insula and stronger connectivity was associated with higher Fear-Affect scores (r = 0.31, p = 0.0003). The multivariate pattern analysis also identified a psychosis-specific correlate of Fear-Affect score between the amygdala/hippocampus cluster and a cluster in the ventromedial prefrontal cortex (VMPFC). Higher Fear-Affect scores were correlated with stronger amygdala/hippocampal-VMPFC connectivity in the early psychosis group (r = 0.33, p = 0.002), but not in controls (r = -0.15, p = 0.28). The current study provides evidence for the transdiagnostic role of the amygdala, hippocampus, and anterior insula in the neural basis of fear and anxiety and suggests a psychosis-specific relationship between fear and anxiety symptoms and amygdala/hippocampal-VMPFC connectivity. Our novel data-driven approach identifies novel, psychosis-specific treatment targets for fear and anxiety symptoms and provides complimentary evidence to decades of hypothesis-driven approaches examining the brain basis of threat processing.

3.
Mol Psychiatry ; 27(2): 1177-1183, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34697450

RESUMEN

The early stage of psychosis (ESP) is a critical period where effective intervention has the most favorable impact on outcomes. Thalamic connectivity abnormalities have been consistently found in psychosis, and are associated with clinical symptoms and cognitive deficits. However, most studies consider ESP patients as a homogeneous population and fail to take the duration of illness into account. In this study, we aimed to capture the progression of thalamic connectivity changes over the first five years of psychosis. Resting-state functional MRI scans were collected from 156 ESP patients (44 with longitudinal data) and 82 healthy controls (24 with longitudinal data). We first performed a case-control analysis comparing thalamic connectivity with 13 networks in the cortex and cerebellum. Next, we modelled the shape (flat, linear, curvilinear) of thalamic connectivity trajectories by comparing flexible non-linear versus linear models. We then tested the significance of the duration of illness and diagnosis in trajectories that changed over time. Connectivity changed over the ESP period between the thalamus and default mode network (DMN) and fronto-parietal network (FPN) nodes in both the cortex and cerebellum. Three models followed a curvilinear trajectory (early increase followed by a subsequent decrease), while thalamo-cerebellar FPN connectivity followed a linear trajectory of steady reductions over time, indicating different rates of change. Finally, diagnosis significantly predicted thalamic connectivity. Thalamo-cortical and thalamo-cerebellar connectivity change in a dynamic fashion during the ESP period. A better understanding of these changes may provide insights into the compensatory and progressive changes in functional connectivity in the early stages of illness.


Asunto(s)
Trastornos Psicóticos , Tálamo , Cerebelo , Humanos , Imagen por Resonancia Magnética , Vías Nerviosas
4.
Proc Natl Acad Sci U S A ; 116(18): 9050-9059, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30988201

RESUMEN

Converging evidence indicates that groups of patients with nominally distinct psychiatric diagnoses are not separated by sharp or discontinuous neurobiological boundaries. In healthy populations, individual differences in behavior are reflected in variability across the collective set of functional brain connections (functional connectome). These data suggest that the spectra of transdiagnostic symptom profiles observed in psychiatric patients may map onto detectable patterns of network function. To examine the manner through which neurobiological variation might underlie clinical presentation, we obtained fMRI data from over 1,000 individuals, including 210 diagnosed with a primary psychotic disorder or affective psychosis (bipolar disorder with psychosis and schizophrenia or schizoaffective disorder), 192 presenting with a primary affective disorder without psychosis (unipolar depression, bipolar disorder without psychosis), and 608 demographically matched healthy comparison participants recruited through a large-scale study of brain imaging and genetics. Here, we examine variation in functional connectomes across psychiatric diagnoses, finding striking evidence for disease connectomic "fingerprints" that are commonly disrupted across distinct forms of pathology and appear to scale as a function of illness severity. The presence of affective and psychotic illnesses was associated with graded disruptions in frontoparietal network connectivity (encompassing aspects of dorsolateral prefrontal, dorsomedial prefrontal, lateral parietal, and posterior temporal cortices). Conversely, other properties of network connectivity, including default network integrity, were preferentially disrupted in patients with psychotic illness, but not patients without psychotic symptoms. This work allows us to establish key biological and clinical features of the functional connectomes of severe mental disease.


Asunto(s)
Conectoma/métodos , Trastornos del Humor/fisiopatología , Trastornos Psicóticos/fisiopatología , Adulto , Trastorno Bipolar/fisiopatología , Encéfalo/fisiopatología , Trastorno Depresivo Mayor/fisiopatología , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Red Nerviosa/fisiopatología , Descanso/fisiología , Esquizofrenia/fisiopatología
5.
Mol Psychiatry ; 25(9): 2200, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30651603

RESUMEN

The original version of this article omitted the author "Roscoe O. Brady Jr." from the "Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, MA, USA" and the "Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA". This has been corrected in both the PDF and HTML versions of the article.

6.
Mol Psychiatry ; 25(9): 2119-2129, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-30443042

RESUMEN

Neuroimaging studies of psychotic disorders have demonstrated abnormalities in structural and functional connectivity involving widespread brain networks. However, these group-level observations have failed to yield any biomarkers that can provide confirmatory evidence of a patient's current symptoms, predict future symptoms, or predict a treatment response. Lack of precision in both neuroanatomical and clinical boundaries have likely contributed to the inability of even well-powered studies to resolve these key relationships. Here, we employed a novel approach to defining individual-specific functional connectivity in 158 patients diagnosed with schizophrenia (n = 49), schizoaffective disorder (n = 37), or bipolar disorder with psychosis (n = 72), and identified neuroimaging features that track psychotic symptoms in a dimension- or disorder-specific fashion. Using individually specified functional connectivity, we were able to estimate positive, negative, and manic symptoms that showed correlations ranging from r = 0.35 to r = 0.51 with the observed symptom scores. Comparing optimized estimation models among schizophrenia spectrum patients, positive and negative symptoms were associated with largely non-overlapping sets of cortical connections. Comparing between schizophrenia spectrum and bipolar disorder patients, the models for positive symptoms were largely non-overlapping between the two disorder classes. Finally, models derived using conventional region definition strategies performed at chance levels for most symptom domains. Individual-specific functional connectivity analyses revealed important new distinctions among cortical circuits responsible for the positive and negative symptoms, as well as key new information about how circuits underlying symptom expressions may vary depending on the underlying etiology and illness syndrome from which they manifest.


Asunto(s)
Trastorno Bipolar , Trastornos Psicóticos , Esquizofrenia , Biomarcadores , Humanos , Imagen por Resonancia Magnética , Trastornos Psicóticos/diagnóstico por imagen , Esquizofrenia/diagnóstico por imagen
7.
Immunity ; 33(2): 216-28, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20727792

RESUMEN

Natural Killer T (NKT) cells are lipid-reactive, CD1d-restricted T lymphocytes important in infection, cancer, and autoimmunity. In addition to foreign antigens, NKT cells react with endogenous self lipids. However, in the face of stimulating self antigen, it remains unclear how overstimulation of NKT cells is avoided. We hypothesized that constantly degraded endogenous antigen only accumulates upon inhibition of alpha-galactosidase A (alpha-Gal-A) in lysosomes. Here, we show that alpha-Gal-A deficiency caused vigorous activation of NKT cells. Moreover, microbes induced inhibition of alpha-Gal-A activity in antigen-presenting cells. This temporary enzyme block depended on Toll-like receptor (TLR) signaling and ultimately triggered lysosomal lipid accumulation. Thus, we present TLR-dependent negative regulation of alpha-Gal-A as a mechanistic link between pathogen recognition and self lipid antigen induction for NKT cells.


Asunto(s)
Autoantígenos/inmunología , Lípidos/inmunología , Lisosomas/inmunología , Células T Asesinas Naturales/inmunología , alfa-Galactosidasa/inmunología , Animales , Presentación de Antígeno , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Homeostasis , Activación de Linfocitos , Lisosomas/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Células T Asesinas Naturales/enzimología , Transducción de Señal , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , alfa-Galactosidasa/metabolismo
8.
J Clin Psychopharmacol ; 38(1): 80-85, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29232311

RESUMEN

PURPOSE: Rosenblat and McIntyre (Acta Psychiatr Scand. 2015;132: 180-191) propose that immune disorders are important mediators between bipolar disorders and medical comorbidities. Rosenblat et al (Bipolar Disord. 2016;18:89-101) present a meta-analysis showing that adjunctive anti-inflammatory agents could evoke moderate antidepressant responses in bipolar disorders. We propose using the anti-inflammatory drug colchicine to improve the long-term safety and efficacy of lithium treatment for bipolar disorders. METHODS: This report is based on searches of the PubMed and Web of Science databases. RESULTS: Bipolar disorders are associated with significant medical comorbidities such as hypertension, overweight/obesity, diabetes mellitus, metabolic syndrome, and arteriosclerosis, accompanied by enhanced release of pro-inflammatory markers during changes in mood state. During lithium therapy, granulocyte-colony stimulating factor, CD34+ hematopoietic stem/progenitor cells, and neutrophil elastase enter the circulation with activated neutrophils to promote the extravascular migration of activated neutrophils and enhance tissue inflammation. Concurrent treatment with lithium and low-dose colchicine could facilitate the responsiveness of bipolar patients to lithium by reducing leukocyte tissue emigration, the release of neutrophil elastase, and the release of leukocyte pro-inflammatory cytokines such as IL-1ß that are regulated by the NLRP3 inflammasome assembly complex. CONCLUSIONS: Concurrent therapy with lithium and low-dose colchicine could reduce complications involving leukocyte-mediated inflammatory states in bipolar patients and promote patient acceptance and responsiveness to lithium therapy.


Asunto(s)
Colchicina/administración & dosificación , Inflamación/prevención & control , Compuestos de Litio/efectos adversos , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Trastorno Bipolar/complicaciones , Trastorno Bipolar/tratamiento farmacológico , Colchicina/farmacología , Citocinas , Relación Dosis-Respuesta a Droga , Humanos , Inflamación/inducido químicamente , Compuestos de Litio/administración & dosificación , Aceptación de la Atención de Salud , Resultado del Tratamiento
9.
Proc Natl Acad Sci U S A ; 112(4): 1137-42, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25583479

RESUMEN

Gaucher disease is caused by mutations of the GBA1 gene, which encodes the lysosomal anchored gluococerebrosidase (GCase). GBA1 mutations commonly result in protein misfolding, abnormal chaperone recognition, and premature degradation, but are less likely to affect catalytic activity. In the present study, we demonstrate that the Hsp90/HOP/Cdc37 complex recruits Hsp27 after recognition of GCase mutants with subsequent targeting of GCase mutant peptides to degradation mechanisms such as VCP and the 26S proteasome. Inhibition of Hsp27 not only increased the quantity of enzyme but also enhanced GCase activity in fibroblasts derived from patients with Gaucher disease. These findings provide insight into a possible therapeutic strategy for protein misfolding diseases by correcting chaperone binding and altering subsequent downstream patterns of protein degradation.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteolisis , Deficiencias en la Proteostasis/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/genética , Chaperoninas/metabolismo , Fibroblastos/metabolismo , Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Proteínas de Choque Térmico HSP27/genética , Proteínas HSP90 de Choque Térmico/genética , Células HeLa , Proteínas de Choque Térmico , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Chaperonas Moleculares , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(7): 2680-5, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550296

RESUMEN

Use of megakaryocytes/platelets for transgene expression may take advantage of their rapid turnover and protective storage in platelets and reduce the risk of activating oncogenes in hematopoietic stem and progenitor cells (HSCs). Here, we show that human megakaryocytic cells could overexpress the lysosomal enzyme, α-l-iduronidase (IDUA), which is deficient in patients with mucopolysaccharidosis type I (MPS I). Upon megakaryocytic differentiation, the amount of released enzyme increased rapidly and steadily by 30-fold. Using a murine MPS I model, we demonstrated that megakaryocyte/platelets were capable of producing, packaging, and storing large amounts of IDUA with proper catalytic activity, lysosomal trafficking, and receptor-mediated uptake. IDUA can be released directly into extracellular space or within microparticles during megakaryocyte maturation or platelet activation, while retaining the capacity for cross-correction in patient's cells. Gene transfer into 1.7% of HSCs led to long-term normalization of plasma IDUA and preferential distribution of enzyme in liver and spleen with complete metabolic correction in MPS I mice. Detection of GFP (coexpressed with IDUA) in Kupffer cells and hepatocytes suggested liver delivery of platelet-derived IDUA possibly via the clearance pathway for senile platelets. These findings provide proof of concept that cells from megakaryocytic lineage and platelets are capable of generating and storing fully functional lysosomal enzymes and can also lead to efficient delivery of both the enzymes released into the circulation and those protected within platelets/microparticles. This study opens a door for use of the megakaryocytes/platelets as a depot for efficient production, delivery, and effective tissue distribution of lysosomal enzymes.


Asunto(s)
Plaquetas/enzimología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Iduronidasa/metabolismo , Lisosomas/enzimología , Mucopolisacaridosis I/enzimología , Animales , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas , Hepatocitos/metabolismo , Humanos , Iduronidasa/administración & dosificación , Iduronidasa/genética , Megacariocitos/citología , Ratones , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Transgenes/genética , Transgenes/fisiología
11.
Proc Natl Acad Sci U S A ; 111(1): 249-54, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24351928

RESUMEN

Gaucher disease is caused by mutations in the glucosidase, beta, acid gene that encodes glucocerebrosidase (GCase). Glucosidase, beta, acid mutations often cause protein misfolding and quantitative loss of GCase. In the present study, we found that celastrol, an herb derivative with known anticancer, anti-inflammatory, and antioxidant activity, significantly increased the quantity and catalytic activity of GCase. Celastrol interfered with the establishment of the heat-shock protein 90/Hsp90 cochaperone Cdc37/Hsp90-Hsp70-organizing protein chaperone complex with mutant GCase and reduced heat-shock protein 90-associated protein degradation. In addition, celastrol modulated the expression of molecular chaperones. Bcl2-associated athanogene 3 and heat shock 70kDa proteins 1A and 1B were significantly increased by celastrol. Furthermore, BAG family molecular chaperone regulator 3 assisted protein folding and maturation of mutant GCase. These findings provide insight into a therapeutic strategy for Gaucher disease and other human disorders that are associated with protein misfolding.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/metabolismo , Chaperonas Moleculares/química , Triterpenos/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis , Catálisis , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/metabolismo , Fibroblastos/metabolismo , Enfermedad de Gaucher/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosilceramidasa/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Humanos , Mutación , Triterpenos Pentacíclicos , Preparaciones de Plantas/farmacología , Unión Proteica , Desnaturalización Proteica , Pliegue de Proteína , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Proc Natl Acad Sci U S A ; 111(13): 4934-9, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24639522

RESUMEN

The inherited deficiency of the lysosomal glucocerebrosidase (GBA) due to mutations in the GBA gene results in Gaucher disease (GD). A vast majority of patients present with nonneuronopathic, type 1 GD (GD1). GBA deficiency causes the accumulation of two key sphingolipids, glucosylceramide (GL-1) and glucosylsphingosine (LysoGL-1), classically noted within the lysosomes of mononuclear phagocytes. How metabolites of GL-1 or LysoGL-1 produced by extralysosomal glucocerebrosidase GBA2 contribute to the GD1 pathophysiology is not known. We recently recapitulated hepatosplenomegaly, cytopenia, hypercytokinemia, and the bone-formation defect of human GD1 through conditional deletion of Gba in Mx1-Cre(+):GD1 mice. Here we show that the deletion of Gba2 significantly rescues the GD1 clinical phenotype, despite enhanced elevations in GL-1 and LysoGL-1. Most notably, the reduced bone volume and bone formation rate are normalized. These results suggest that metabolism of GL-1 or LysoGL-1 into downstream bioactive lipids is a major contributor to the bone-formation defect. Direct testing revealed a strong inhibition of osteoblast viability by nanomolar concentrations of sphingosine, but not of ceramide. These findings are consistent with toxicity of high circulating sphingosine levels in GD1 patients, which decline upon enzyme-replacement therapy; serum ceramide levels remain unchanged. Together, complementary results from mice and humans affected with GD1 not only pinpoint sphingosine as being an osteoblast toxin, but also set forth Gba2 as a viable therapeutic target for the development of inhibitors to ameliorate certain disabling consequences of GD1.


Asunto(s)
Enfermedad de Gaucher/genética , Enfermedad de Gaucher/terapia , Eliminación de Gen , beta-Glucosidasa/genética , Animales , Línea Celular , Enfermedad de Gaucher/enzimología , Humanos , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/patología , Fenotipo , Esfingolípidos/metabolismo , Esfingosina/metabolismo
13.
Mol Genet Metab ; 119(1-2): 144-50, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27471012

RESUMEN

Fabry disease is a glycosphingolipid storage disorder that is caused by a genetic deficiency of the enzyme alpha-galactosidase A (AGA, EC 3.2.1.22). It is a multisystem disease that affects the vascular, cardiac, renal, and nervous systems. One of the hallmarks of this disorder is neuropathic pain and sympathetic and parasympathetic nervous dysfunction. The exact mechanism by which changes in AGA activity result in change in neuronal function is not clear, partly due to of a lack of relevant model systems. In this study, we report the development of an in vitro model system to study neuronal dysfunction in Fabry disease by using short-hairpin RNA to create a stable knock-down of AGA in the human cholinergic neuronal cell line, LA-N-2. We show that gene-silenced cells show specifically reduced AGA activity and store globotriaosylceramide. In gene-silenced cells, release of the neurotransmitter acetylcholine is significantly reduced, demonstrating that this model may be used to study specific neuronal functions such as neurotransmitter release in Fabry disease.


Asunto(s)
Neuronas Colinérgicas/patología , Enfermedad de Fabry/genética , Neuralgia/metabolismo , alfa-Galactosidasa/genética , Neuronas Colinérgicas/metabolismo , Enfermedad de Fabry/metabolismo , Enfermedad de Fabry/patología , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Riñón/metabolismo , Riñón/patología , Neuralgia/genética , Neuralgia/patología , Sistema Nervioso Parasimpático/metabolismo , Sistema Nervioso Parasimpático/patología , ARN Interferente Pequeño/genética , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología , Trihexosilceramidas/metabolismo , alfa-Galactosidasa/biosíntesis
14.
Proc Natl Acad Sci U S A ; 110(3): 966-71, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23277556

RESUMEN

Gaucher disease is caused by mutations of the GBA gene that encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA mutations often result in protein misfolding and premature degradation, but usually exert less effect on catalytic activity. In this study, we identified the molecular mechanism by which histone deacetylase inhibitors increase the quantity and activity of GCase. Specifically, these inhibitors limit the deacetylation of heat shock protein 90, resulting in less recognition of the mutant peptide and GCase degradation. These findings provide insight into a possible therapeutic strategy for Gaucher disease and other genetic disorders by modifying molecular chaperone and protein degradation pathways.


Asunto(s)
Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/enzimología , Glucosilceramidasa/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Chaperonas Moleculares/metabolismo , Acetilación , Degradación Asociada con el Retículo Endoplásmico , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/genética , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Modelos Biológicos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(8): 2999-3004, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382178

RESUMEN

To realize the potential of large molecular weight substances to treat neurological disorders, novel approaches are required to surmount the blood-brain barrier (BBB). We investigated whether fusion of a receptor-binding peptide from apolipoprotein E (apoE) with a potentially therapeutic protein can bind to LDL receptors on the BBB and be transcytosed into the CNS. A lysosomal enzyme, α-L-iduronidase (IDUA), was used for biological and therapeutic evaluation in a mouse model of mucopolysaccharidosis (MPS) type I, one of the most common lysosomal storage disorders with CNS deficits. We identified two fusion candidates, IDUAe1 and IDUAe2, by in vitro screening, that exhibited desirable receptor-mediated binding, endocytosis, and transendothelial transport as well as appropriate lysosomal enzyme trafficking and biological function. Robust peripheral IDUAe1 or IDUAe2 generated by transient hepatic expression led to elevated enzyme levels in capillary-depleted, enzyme-deficient brain tissues and protein delivery into nonendothelium perivascular cells, neurons, and astrocytes within 2 d of treatment. Moreover, 5 mo after long-term delivery of moderate levels of IDUAe1 derived from maturing red blood cells, 2% to 3% of normal brain IDUA activities were obtained in MPS I mice, and IDUAe1 protein was detected in neurons and astrocytes throughout the brain. The therapeutic potential was demonstrated by normalization of brain glycosaminoglycan and ß-hexosaminidase in MPS I mice 5 mo after moderate yet sustained delivery of IDUAe1. These findings provide a noninvasive and BBB-targeted procedure for the delivery of large-molecule therapeutic agents to treat neurological lysosomal storage disorders and potentially other diseases that involve the brain.


Asunto(s)
Apolipoproteínas E/metabolismo , Barrera Hematoencefálica , Lisosomas/enzimología , Ingeniería de Proteínas , Receptores de LDL/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Ratones , Mucopolisacaridosis I/enzimología , Mucopolisacaridosis I/metabolismo , Transcitosis
16.
Proc Natl Acad Sci U S A ; 110(26): 10747-52, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23754423

RESUMEN

Acute traumatic brain injury (TBI) is associated with long-term cognitive and behavioral dysfunction. In vivo studies have shown histone deacetylase inhibitors (HDACis) to be neuroprotective following TBI in rodent models. HDACis are intriguing candidates because they are capable of provoking widespread genetic changes and modulation of protein function. By using known HDACis and a unique small-molecule pan-HDACi (LB-205), we investigated the effects and mechanisms associated with HDACi-induced neuroprotection following CNS injury in an astrocyte scratch assay in vitro and a rat TBI model in vivo. We demonstrate the preservation of sufficient expression of nerve growth factor (NGF) and activation of the neurotrophic tyrosine kinase receptor type 1 (TrkA) pathway following HDACi treatment to be crucial in stimulating the survival of CNS cells after TBI. HDACi treatment up-regulated the expression of NGF, phospho-TrkA, phospho-protein kinase B (p-AKT), NF-κB, and B-cell lymphoma 2 (Bcl-2) cell survival factors while down-regulating the expression of p75 neurotrophin receptor (NTR), phospho-JNK, and Bcl-2-associated X protein apoptosis factors. HDACi treatment also increased the expression of the stem cell biomarker nestin, and decreased the expression of reactive astrocyte biomarker GFAP within damaged tissue following TBI. These findings provide further insight into the mechanisms by which HDACi treatment after TBI is neuroprotective and support the continued study of HDACis following acute TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Factores de Crecimiento Nervioso/fisiología , Fármacos Neuroprotectores/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Factores de Crecimiento Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley
17.
Mol Ther ; 22(12): 2028-2037, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25088464

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a progressive lysosomal storage disorder with systemic and central nervous system (CNS) involvement due to deficiency of α-L-iduronidase (IDUA). We previously identified a receptor-binding peptide from apolipoprotein E (e) that facilitated a widespread delivery of IDUAe fusion protein into CNS. In this study, we evaluated the long-term CNS biodistribution, dose-correlation, and therapeutic benefits of IDUAe after systemic, sustained delivery via hematopoietic stem cell (HSC)-mediated gene therapy with expression restricted to erythroid/megakaryocyte lineages. Compared to the highest dosage group treated by nontargeted control IDUAc (165 U/ml), physiological levels of IDUAe in the circulation (12 U/ml) led to better CNS benefits in MPS I mice as demonstrated in glycosaminoglycan accumulation, histopathology analysis, and neurological behavior. Long-term brain metabolic correction and normalization of exploratory behavior deficits in MPS I mice were observed by peripheral enzyme therapy with physiological levels of IDUAe derived from clinically attainable levels of HSC transduction efficiency (0.1). Importantly, these levels of IDUAe proved to be more beneficial on correction of cerebrum pathology and behavioral deficits in MPS I mice than wild-type HSCs fully engrafted in MPS I chimeras. These results provide compelling evidence for CNS efficacy of IDUAe and its prospective translation to clinical application.


Asunto(s)
Encéfalo/patología , Células Madre Hematopoyéticas/citología , Iduronidasa/genética , Iduronidasa/farmacocinética , Mucopolisacaridosis I/terapia , Receptores de Péptidos/metabolismo , Animales , Apolipoproteínas E/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/enzimología , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Humanos , Iduronidasa/uso terapéutico , Ratones , Mucopolisacaridosis I/enzimología , Mucopolisacaridosis I/patología , Receptores de Péptidos/genética , Proteínas Recombinantes , Distribución Tisular
18.
Proc Natl Acad Sci U S A ; 109(18): 6963-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22505738

RESUMEN

Astrocytes are the most abundant cell of the CNS and demonstrate contact inhibition in which a nonproliferative, nonmotile cellular state is achieved once stable intercellular contacts are formed between mature cells. Cellular injury disrupts these intercellular contacts, causing a loss of contact inhibition and the rapid initiation of healing. Dysregulation of the molecular pathways involved in this process is thought to lead to an aggressive cellular state associated with neoplasia. We investigated whether a comparable correlation exists between the response of astrocytes to injury and the malignant phenotype of astrocytomas. We discovered that the loss of contact inhibition plays a critical role in the initiation and regulation of reactive astrocytes in the healing of wounds. In particular, injury of the astrocytes interrupts and destabilizes the cadherin-catenin complexes at the cell membrane leading to nuclear translocation of ß-catenin and characteristic changes associated with the activation of astrocytes. Similar signaling pathways are found to be active--but dysregulated--in astrocytomas. Inhibition of ß-catenin signaling diminished both the response of astrocytes to injury and induction of the malignant phenotype of astrocytomas. The findings shed light on a unique mechanism associated with the pathogenesis of astrocytomas and provide a model for the loss of contact inhibition that may broadly apply to understanding the mechanisms of tissue repair and tumorigenesis in the brain.


Asunto(s)
Astrocitos/metabolismo , Astrocitoma/etiología , Astrocitoma/metabolismo , beta Catenina/metabolismo , Animales , Astrocitoma/patología , Proliferación Celular , Transformación Celular Neoplásica , Células Cultivadas , Técnicas de Silenciamiento del Gen , Ratones , Modelos Neurológicos , Fenotipo , ARN Interferente Pequeño , Transducción de Señal , Células Tumorales Cultivadas , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
19.
Proc Natl Acad Sci U S A ; 108(52): 21200-5, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160715

RESUMEN

Gaucher disease (GD) is caused by a spectrum of genetic mutations within the gene encoding the lysosomal enzyme glucocerebrosidase (GCase). These mutations often lead to misfolded proteins that are recognized by the unfolded protein response system and are degraded through the ubiquitin-proteasome pathway. Modulating this pathway with histone deacetylase inhibitors (HDACis) has been shown to improve protein stability in other disease settings. To identify the mechanisms involved in the regulation of GCase and determine the effects of HDACis on protein stability, we investigated the most prevalent mutations for nonneuronopathic (N370S) and neuronopathic (L444P) GD in cultured fibroblasts derived from GD patients and HeLa cells transfected with these mutations. The half-lives of mutant GCase proteins correspond to decreases in protein levels and enzymatic activity. GCase was found to bind to Hsp70, which directed the protein to TCP1 for proper folding, and to Hsp90, which directed the protein to the ubiquitin-proteasome pathway. Using a known HDACi (SAHA) and a unique small-molecule HDACi (LB-205), GCase levels increased rescuing enzymatic activity in mutant cells. The increase in the quantity of protein can be attributed to increases in protein half-life that correspond primarily with a decrease in degradation rather than an increase in chaperoned folding. HDACis reduce binding to Hsp90 and prevent subsequent ubiquitination and proteasomal degradation without affecting binding to Hsp70 or TCP1. These findings provide insight into the pathogenesis of GD and indicate a potent therapeutic potential of HDAC inhibitors for the treatment of GD and other human protein misfolding disorders.


Asunto(s)
Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Inhibidores de Histona Desacetilasas/farmacología , Mutación/genética , Estabilidad Proteica/efectos de los fármacos , Western Blotting , Clonación Molecular , Fibroblastos , Enfermedad de Gaucher/tratamiento farmacológico , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Mutagénesis Sitio-Dirigida , Pliegue de Proteína , Proteolisis/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 108(12): 4980-5, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21383154

RESUMEN

Neurofibromatosis type 2 (NF2) is a multiple neoplasia syndrome and is caused by a mutation of the NF2 tumor suppressor gene that encodes for the tumor suppressor protein merlin. Biallelic NF2 gene inactivation results in the development of central nervous system tumors, including schwannomas, meningiomas, ependymomas, and astrocytomas. Although a wide variety of missense germline mutations in the coding sequences of the NF2 gene can cause loss of merlin function, the mechanism of this functional loss is unknown. To gain insight into the mechanisms underlying loss of merlin function in NF2, we investigated mutated merlin homeostasis and function in NF2-associated tumors and cell lines. Quantitative protein and RT-PCR analysis revealed that whereas merlin protein expression was significantly reduced in NF2-associated tumors, mRNA expression levels were unchanged. Transfection of genetic constructs of common NF2 missense mutations into NF2 gene-deficient meningioma cell lines revealed that merlin loss of function is due to a reduction in mutant protein half-life and increased protein degradation. Transfection analysis also demonstrated that recovery of tumor suppressor protein function is possible, indicating that these mutants maintain intrinsic functional capacity. Further, increased expression of mutant protein is possible after treatment with specific proteostasis regulators, implicating protein quality control systems in the degradative fate of mutant tumor suppressor proteins. These findings provide direct insight into protein function and tumorigenesis in NF2 and indicate a unique treatment paradigm for this disorder.


Asunto(s)
Neoplasias del Sistema Nervioso Central/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes de la Neurofibromatosis 2 , Mutación Missense , Neurofibromatosis 2/metabolismo , Neurofibromina 2/biosíntesis , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/genética , Silenciador del Gen , Humanos , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA