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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 147(3): 615-28, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036569

RESUMEN

Assemblies of ß-amyloid (Aß) peptides are pathological mediators of Alzheimer's Disease (AD) and are produced by the sequential cleavages of amyloid precursor protein (APP) by ß-secretase (BACE1) and γ-secretase. The generation of Aß is coupled to neuronal activity, but the molecular basis is unknown. Here, we report that the immediate early gene Arc is required for activity-dependent generation of Aß. Arc is a postsynaptic protein that recruits endophilin2/3 and dynamin to early/recycling endosomes that traffic AMPA receptors to reduce synaptic strength in both hebbian and non-hebbian forms of plasticity. The Arc-endosome also traffics APP and BACE1, and Arc physically associates with presenilin1 (PS1) to regulate γ-secretase trafficking and confer activity dependence. Genetic deletion of Arc reduces Aß load in a transgenic mouse model of AD. In concert with the finding that patients with AD can express anomalously high levels of Arc, we hypothesize that Arc participates in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endosomas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas , Animales , Membrana Celular/metabolismo , Humanos , Ratones , Ratones Noqueados
3.
Nature ; 537(7618): 50-6, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27582220

RESUMEN

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/efectos de los fármacos , Amiloide/metabolismo , Péptidos beta-Amiloides/química , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ensayos Clínicos Fase III como Asunto , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Modelos Biológicos , Placa Amiloide/patología , Agregación Patológica de Proteínas/tratamiento farmacológico , Solubilidad
4.
Proc Natl Acad Sci U S A ; 113(17): 4777-82, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27078105

RESUMEN

Dimethyl fumarate (DMF) (BG-12, Tecfidera) is a fumaric acid ester (FAE) that was advanced as a multiple sclerosis (MS) therapy largely for potential neuroprotection as it was recognized that FAEs are capable of activating the antioxidative transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway. However, DMF treatment in randomized controlled MS trials was associated with marked reductions in relapse rate and development of active brain MRI lesions, measures considered to reflect CNS inflammation. Here, we investigated the antiinflammatory contribution of Nrf2 in DMF treatment of the MS model, experimental autoimmune encephalomyelitis (EAE). C57BL/6 wild-type (WT) and Nrf2-deficient (Nrf2(-/-)) mice were immunized with myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 (p35-55) for EAE induction and treated with oral DMF or vehicle daily. DMF protected WT and Nrf2(-/-) mice equally well from development of clinical and histologic EAE. The beneficial effect of DMF treatment in Nrf2(-/-) and WT mice was accompanied by reduced frequencies of IFN-γ and IL-17-producing CD4(+) cells and induction of antiinflammatory M2 (type II) monocytes. DMF also modulated B-cell MHC II expression and reduced the incidence of clinical disease in a B-cell-dependent model of spontaneous CNS autoimmunity. Our observations that oral DMF treatment promoted immune modulation and provided equal clinical benefit in acute EAE in Nrf2(-/-) and WT mice, suggest that the antiinflammatory activity of DMF in treatment of MS patients may occur through alternative pathways, independent of Nrf2.


Asunto(s)
Inmunidad Adaptativa/inmunología , Dimetilfumarato/administración & dosificación , Inmunidad Innata/inmunología , Inmunomodulación/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Bazo/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Administración Oral , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/administración & dosificación , Inmunomodulación/efectos de los fármacos , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/efectos de los fármacos
5.
J Biol Chem ; 292(43): 17963-17974, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28860188

RESUMEN

Aberrant activation of matrix metalloproteinases (MMPs) is a common feature of pathological cascades observed in diverse disorders, such as cancer, fibrosis, immune dysregulation, and neurodegenerative diseases. MMP-9, in particular, is highly dynamically regulated in several pathological processes. Development of MMP inhibitors has therefore been an attractive strategy for therapeutic intervention. However, a long history of failed clinical trials has demonstrated that broad-spectrum MMP inhibitors have limited clinical utility, which has spurred the development of inhibitors selective for individual MMPs. Attaining selectivity has been technically challenging because of sequence and structural conservation across the various MMPs. Here, through a biochemical and structural screening paradigm, we have identified JNJ0966, a highly selective compound that inhibited activation of MMP-9 zymogen and subsequent generation of catalytically active enzyme. JNJ0966 had no effect on MMP-1, MMP-2, MMP-3, MMP-9, or MMP-14 catalytic activity and did not inhibit activation of the highly related MMP-2 zymogen. The molecular basis for this activity was characterized as an interaction of JNJ0966 with a structural pocket in proximity to the MMP-9 zymogen cleavage site near Arg-106, which is distinct from the catalytic domain. JNJ0966 was efficacious in reducing disease severity in a mouse experimental autoimmune encephalomyelitis model, demonstrating the viability of this therapeutic approach. This discovery reveals an unprecedented pharmacological approach to MMP inhibition, providing an opportunity to improve selectivity of future clinical drug candidates. Targeting zymogen activation in this manner may also allow for pharmaceutical exploration of other enzymes previously viewed as intractable drug targets.


Asunto(s)
Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/química , Metaloproteinasa 9 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/química , Regulación Alostérica , Animales , Células COS , Dominio Catalítico , Chlorocebus aethiops , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Dominios Proteicos
6.
Acta Neuropathol ; 130(2): 279-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25920452

RESUMEN

Dimethyl fumarate (DMF), recently approved as an oral immunomodulatory treatment for relapsing-remitting multiple sclerosis (MS), metabolizes to monomethyl fumarate (MMF) which crosses the blood-brain barrier and has demonstrated neuroprotective effects in experimental studies. We postulated that MMF exerts neuroprotective effects through modulation of microglia activation, a critical component of the neuroinflammatory cascade that occurs in neurodegenerative diseases such as MS. To ascertain our hypothesis and define the mechanistic pathways involved in the modulating effect of fumarates, we used real-time PCR and biochemical assays to assess changes in the molecular and functional phenotype of microglia, quantitative Western blotting to monitor activation of postulated pathway components, and ex vivo whole-cell patch clamp recording of excitatory post-synaptic currents in corticostriatal slices from mice with experimental autoimmune encephalomyelitis (EAE), a model for MS, to study synaptic transmission. We show that exposure to MMF switches the molecular and functional phenotype of activated microglia from classically activated, pro-inflammatory type to alternatively activated, neuroprotective one, through activation of the hydroxycarboxylic acid receptor 2 (HCAR2). We validate a downstream pathway mediated through the AMPK-Sirt1 axis resulting in deacetylation, and thereby inhibition, of NF-κB and, consequently, of secretion of pro-inflammatory molecules. We demonstrate through ex vivo monitoring of spontaneous glutamate-mediated excitatory post-synaptic currents of single neurons in corticostriatal slices from EAE mice that the neuroprotective effect of DMF was exerted on neurons at pre-synaptic terminals by modulating glutamate release. By exposing control slices to untreated and MMF-treated activated microglia, we confirm the modulating effect of MMF on microglia function and, thereby, its indirect neuroprotective effect at post-synaptic level. These findings, whereby DMF-induced activation of a new HCAR2-dependent pathway on microglia leads to the modulation of neuroinflammation and restores synaptic alterations occurring in EAE, represent a possible novel mechanism of action for DMF in MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Fumaratos/farmacología , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Sinapsis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Línea Celular , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/fisiopatología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Ácido Glutámico/metabolismo , Ratones Endogámicos C57BL , Microglía/fisiología , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Sinapsis/fisiología , Técnicas de Cultivo de Tejidos
7.
Eur J Neurosci ; 39(7): 1225-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24713001

RESUMEN

Amyloid beta (Aß), a key component in the pathophysiology of Alzheimer's disease, is thought to target excitatory synapses early in the disease. However, the mechanism by which Aß weakens synapses is not well understood. Here we showed that the PDZ domain protein, protein interacting with C kinase 1 (PICK1), was required for Aß to weaken synapses. In mice lacking PICK1, elevations of Aß failed to depress synaptic transmission in cultured brain slices. In dissociated cultured neurons, Aß failed to reduce surface α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit 2, a subunit of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors that binds with PICK1 through a PDZ ligand-domain interaction. Lastly, a novel small molecule (BIO922) discovered through structure-based drug design that targets the specific interactions between GluA2 and PICK1 blocked the effects of Aß on synapses and surface receptors. We concluded that GluA2-PICK1 interactions are a key component of the effects of Aß on synapses.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Proteínas Portadoras/metabolismo , Potenciales Postsinápticos Excitadores , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/toxicidad , Sinapsis/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Células Cultivadas , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/fisiología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Nucleares/genética , Unión Proteica , Ratas , Receptores AMPA/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
9.
SLAS Technol ; 28(2): 63-69, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36455858

RESUMEN

The development of phenotypic assays with appropriate analyses is an important step in the drug discovery process. Assays using induced pluripotent stem cell (iPSC)-derived human neurons are emerging as powerful tools for drug discovery in neurological disease. We have previously shown that longitudinal single cell tracking enabled the quantification of survival and death of neurons after overexpression of α-synuclein with a familial Parkinson's disease mutation (A53T). The reliance of this method on manual counting, however, rendered the process labor intensive, time consuming and error prone. To overcome these hurdles, we have developed automated detection algorithms for neurons using the BioStation CT live imaging system and CL-Quant software. In the current study, we use these algorithms to successfully measure the risk of neuronal death caused by overexpression of α-synuclein (A53T) with similar accuracy and improved consistency as compared to manual counting. This novel method also provides additional key readouts of neuronal fitness including total neurite length and the number of neurite nodes projecting from the cell body. Finally, the algorithm reveals the neuroprotective effects of brain-derived neurotrophic factor (BDNF) treatment in neurons overexpressing α-synuclein (A53T). These data show that an automated algorithm improves the consistency and considerably shortens the analysis time of assessing neuronal health, making this method advantageous for small molecule screening for inhibitors of synucleinopathy and other neurodegenerative diseases.


Asunto(s)
Sinucleinopatías , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Sinucleinopatías/metabolismo , Rastreo Celular , Neuronas/metabolismo , Algoritmos
10.
J Pharmacol Exp Ther ; 341(1): 274-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22267202

RESUMEN

Oxidative stress is central to the pathology of several neurodegenerative diseases, including multiple sclerosis, and therapeutics designed to enhance antioxidant potential could have clinical value. The objective of this study was to characterize the potential direct neuroprotective effects of dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) on cellular resistance to oxidative damage in primary cultures of central nervous system (CNS) cells and further explore the dependence and function of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in this process. Treatment of animals or primary cultures of CNS cells with DMF or MMF resulted in increased nuclear levels of active Nrf2, with subsequent up-regulation of canonical antioxidant target genes. DMF-dependent up-regulation of antioxidant genes in vivo was lost in mice lacking Nrf2 [Nrf2(-/-)]. DMF or MMF treatment increased cellular redox potential, glutathione, ATP levels, and mitochondrial membrane potential in a concentration-dependent manner. Treating astrocytes or neurons with DMF or MMF also significantly improved cell viability after toxic oxidative challenge in a concentration-dependent manner. This effect on viability was lost in cells that had eliminated or reduced Nrf2. These data suggest that DMF and MMF are cytoprotective for neurons and astrocytes against oxidative stress-induced cellular injury and loss, potentially via up-regulation of an Nrf2-dependent antioxidant response. These data also suggest DMF and MMF may function through improving mitochondrial function. The clinical utility of DMF in multiple sclerosis is being explored through phase III trials with BG-12, which is an oral therapeutic containing DMF as the active ingredient.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Citoprotección/genética , Fumaratos/farmacología , Factor 2 Relacionado con NF-E2/fisiología , Neuronas/metabolismo , Estrés Oxidativo/genética , Transducción de Señal/genética , Animales , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Brain ; 134(Pt 3): 678-92, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21354971

RESUMEN

Inflammation and oxidative stress are thought to promote tissue damage in multiple sclerosis. Thus, novel therapeutics enhancing cellular resistance to free radicals could prove useful for multiple sclerosis treatment. BG00012 is an oral formulation of dimethylfumarate. In a phase II multiple sclerosis trial, BG00012 demonstrated beneficial effects on relapse rate and magnetic resonance imaging markers indicative of inflammation as well as axonal destruction. First we have studied effects of dimethylfumarate on the disease course, central nervous system, tissue integrity and the molecular mechanism of action in an animal model of chronic multiple sclerosis: myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis in C57BL/6 mice. In the chronic phase of experimental autoimmune encephalomyelitis, preventive or therapeutic application of dimethylfumarate ameliorated the disease course and improved preservation of myelin, axons and neurons. In vitro, the application of fumarates increased murine neuronal survival and protected human or rodent astrocytes against oxidative stress. Application of dimethylfumarate led to stabilization of the transcription factor nuclear factor (erythroid-derived 2)-related factor 2, activation of nuclear factor (erythroid-derived 2)-related factor 2-dependent transcriptional activity and accumulation of NADP(H) quinoline oxidoreductase-1 as a prototypical target gene. Furthermore, the immediate metabolite of dimethylfumarate, monomethylfumarate, leads to direct modification of the inhibitor of nuclear factor (erythroid-derived 2)-related factor 2, Kelch-like ECH-associated protein 1, at cysteine residue 151. In turn, increased levels of nuclear factor (erythroid-derived 2)-related factor 2 and reduced protein nitrosylation were detected in the central nervous sytem of dimethylfumarate-treated mice. Nuclear factor (erythroid-derived 2)-related factor 2 was also upregulated in the spinal cord of autopsy specimens from untreated patients with multiple sclerosis. In dimethylfumarate-treated mice suffering from experimental autoimmune encephalomyelitis, increased immunoreactivity for nuclear factor (erythroid-derived 2)-related factor 2 was detected by confocal microscopy in neurons of the motor cortex and the brainstem as well as in oligodendrocytes and astrocytes. In mice deficient for nuclear factor (erythroid-derived 2)-related factor 2 on the same genetic background, the dimethylfumarate mediated beneficial effects on clinical course, axon preservation and astrocyte activation were almost completely abolished thus proving the functional relevance of this transcription factor for the neuroprotective mechanism of action. We conclude that the ability of dimethylfumarate to activate nuclear factor (erythroid-derived 2)-related factor 2 may offer a novel cytoprotective modality that further augments the natural antioxidant responses in multiple sclerosis tissue and is not yet targeted by other multiple sclerosis therapies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Fumaratos/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Aldehído Reductasa/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Antioxidantes/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Axones/metabolismo , Axones/patología , Complejo CD3/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Encefalomielitis Autoinmune Experimental/etiología , Femenino , Fumaratos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/efectos adversos , Proteínas Fluorescentes Verdes/genética , Humanos , Peróxido de Hidrógeno/farmacología , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Proteínas de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Fármacos Neuroprotectores/farmacología , Proteínas Nogo , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/efectos adversos , ARN Interferente Pequeño/farmacología , Sueño/fisiología , Médula Espinal/citología , Estadísticas no Paramétricas , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo , Transfección
12.
Mol Neurobiol ; 59(4): 2171-2189, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35060064

RESUMEN

Stearoyl-CoA desaturase (SCD) is a potential therapeutic target for Parkinson's and related neurodegenerative diseases. SCD inhibition ameliorates neuronal toxicity caused by aberrant α-synuclein, a lipid-binding protein implicated in Parkinson's disease. Its inhibition depletes monounsaturated fatty acids, which may modulate α-synuclein conformations and membrane interactions. Herein, we characterize the pharmacokinetic and pharmacodynamic properties of YTX-7739, a clinical-stage SCD inhibitor. Administration of YTX-7739 to rats and monkeys for 15 days caused a dose-dependent increase in YTX-7739 concentrations that were well-tolerated and associated with concentration-dependent reductions in the fatty acid desaturation index (FADI), the ratio of monounsaturated to saturated fatty acids. An approximate 50% maximal reduction in the carbon-16 desaturation index was observed in the brain, with comparable responses in the plasma and skin. A study with a diet supplemented in SCD products indicates that changes in brain C16 desaturation were due to local SCD inhibition, rather than to changes in systemic fatty acids that reach the brain. Assessment of pharmacodynamic response onset and reversibility kinetics indicated that approximately 7 days of dosing were required to achieve maximal responses, which persisted for at least 2 days after cessation of dosing. YTX-7739 thus achieved sufficient concentrations in the brain to inhibit SCD and produce pharmacodynamic responses that were well-tolerated in rats and monkeys. These results provide a framework for evaluating YTX-7739 pharmacology clinically as a disease-modifying therapy to treat synucleinopathies.


Asunto(s)
Enfermedad de Parkinson , Estearoil-CoA Desaturasa , Animales , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Metabolismo de los Lípidos/fisiología , Ratas , Estearoil-CoA Desaturasa/metabolismo , alfa-Sinucleína/metabolismo
13.
PLoS One ; 17(12): e0277532, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36454869

RESUMEN

There are currently no preventive or disease-modifying therapies for Parkinson's Disease (PD). Failures in clinical trials necessitate a re-evaluation of existing pre-clinical models in order to adopt systems that better recapitulate underlying disease mechanisms and better predict clinical outcomes. In recent years, models utilizing patient-derived induced pluripotent stem cells (iPSC) have emerged as attractive models to recapitulate disease-relevant neuropathology in vitro without exogenous overexpression of disease-related pathologic proteins. Here, we utilized iPSC derived from patients with early-onset PD and dementia phenotypes that harbored either a point mutation (A53T) or multiplication at the α-synuclein/SNCA gene locus. We generated a three-dimensional (3D) cortical neurosphere culture model to better mimic the tissue microenvironment of the brain. We extensively characterized the differentiation process using quantitative PCR, Western immunoblotting and immunofluorescence staining. Differentiated and aged neurospheres revealed alterations in fatty acid profiles and elevated total and pathogenic phospho-α-synuclein levels in both A53T and the triplication lines compared to their isogenic control lines. Furthermore, treatment of the neurospheres with a small molecule inhibitor of stearoyl CoA desaturase (SCD) attenuated the protein accumulation and aberrant fatty acid profile phenotypes. Our findings suggest that the 3D cortical neurosphere model is a useful tool to interrogate targets for PD and amenable to test small molecule therapeutics.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/genética , Enfermedad de Parkinson/genética , Organoides , Ácidos Grasos
14.
Neurotherapeutics ; 19(3): 1018-1036, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35445353

RESUMEN

Increasing evidence has shown that Parkinson's disease (PD) impairs midbrain dopaminergic, cortical and other neuronal subtypes in large part due to the build-up of lipid- and vesicle-rich α-synuclein (αSyn) cytotoxic inclusions. We previously identified stearoyl-CoA desaturase (SCD) as a potential therapeutic target for synucleinopathies. A brain-penetrant SCD inhibitor, YTX-7739, was developed and has entered Phase 1 clinical trials. Here, we report the efficacy of YTX-7739 in reversing pathological αSyn phenotypes in various in vitro and in vivo PD models. In cell-based assays, YTX-7739 decreased αSyn-mediated neuronal death, reversed the abnormal membrane interaction of amplified E46K ("3K") αSyn, and prevented pathological phenotypes in A53T and αSyn triplication patient-derived neurospheres, including dysregulated fatty acid profiles and pS129 αSyn accumulation. In 3K PD-like mice, YTX-7739 crossed the blood-brain barrier, decreased unsaturated fatty acids, and prevented progressive motor deficits. Both YTX-7739 treatment and decreasing SCD activity through deletion of one copy of the SCD1 gene (SKO) restored the physiological αSyn tetramer-to-monomer ratio, dopaminergic integrity, and neuronal survival in 3K αSyn mice. YTX-7739 efficiently reduced pS129 + and PK-resistant αSyn in both human wild-type αSyn and 3K mutant mice similar to the level of 3K-SKO. Together, these data provide further validation of SCD as a PD therapeutic target and YTX-7739 as a clinical candidate for treating human α-synucleinopathies.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Encéfalo/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
Bioorg Med Chem Lett ; 21(21): 6485-90, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21925881

RESUMEN

The discovery of potent N-hydroxyl caprolactam matrix metalloproteinase (MMP) inhibitors (6) based on the natural product Cobactin-T (2) is described. The synthetic method, which utilizes the ring closing metathesis reaction, is compatible to provide complementary (R) and (S) enantiomers. These compounds tested against MMP-2 and 9, show that the R stereochemistry (i.e., 16), which is opposite for that found in the natural product Cobactin-T is >1000-fold more active with IC(50) values of 0.2-0.6nM against both enzymes. The variation in the incorporation of the sulfonamide enzyme recognition element (Ar(2)XAr(1)SO(2)N(R(1)), 6), along with alterations in the RCM/double bond chemistry (R(2)) provided a series of sub nanomolar MMP inhibitors. For example, compounds 16 and 34 were found to be the most potent with IC(50) values against MMP-2 and MMP-9 found to be between 0.2 and 0.6nM with 34 being the most potent compound discovered (MMP-2 IC(50)=0.39nM and MMP-9 IC(50)=0.22nM). Compounds 16 and 34 showed acceptable drug-like properties in vivo with compound 34 showing oral bioavailability.


Asunto(s)
Azepinas/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/farmacología , Azepinas/farmacocinética , Disponibilidad Biológica , Ciclización , Descubrimiento de Drogas , Concentración 50 Inhibidora , Inhibidores de Proteasas/farmacocinética , Estereoisomerismo
16.
Bioorg Med Chem Lett ; 21(24): 7277-80, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22061640

RESUMEN

Starting from literature examples of nonsteroidal anti-inflammatory drugs (NSAIDs)-type carboxylic acid γ-secretase modulators (GSMs) and using a scaffold design approach, we identified 4-aminomethylphenylacetic acid 4 with a desirable γ-secretase modulation profile. Scaffold optimization led to the discovery of a novel chemical series, represented by 6b, having improved brain penetration. Further SAR studies provided analog 6q that exhibited a good pharmacological profile. Oral administration of 6q significantly reduced brain Aß42 levels in mice and rats.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/química , Inhibidores Enzimáticos/química , Fenilacetatos/química , Piperidinas/química , Administración Oral , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/farmacocinética , Encéfalo/metabolismo , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Ratones , Fragmentos de Péptidos/metabolismo , Fenilacetatos/síntesis química , Fenilacetatos/farmacocinética , Piperidinas/síntesis química , Piperidinas/farmacocinética , Ratas
17.
ACS Med Chem Lett ; 12(3): 351-355, 2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33738061

RESUMEN

S1P5 is one of the five sphingosine-1-phosphate (S1P) receptors which play important roles in immune and CNS cell homeostasis, growth, and differentiation. Little is known about the effect of modulation of S1P5 due to the lack of S1P5 specific modulators with suitable druglike properties. Here we describe the discovery and optimization of a novel series of potent selective S1P5 antagonists and the identification of an orally active brain-penetrant tool compound 15.

18.
Artículo en Inglés | MEDLINE | ID: mdl-33468560

RESUMEN

OBJECTIVE: To test the hypothesis that dimethyl fumarate (DMF, Tecfidera) elicits different biological changes from DMF combined with monoethyl fumarate (MEF) (Fumaderm, a psoriasis therapy), we investigated DMF and MEF in rodents and cynomolgus monkeys. Possible translatability of findings was explored with lymphocyte counts from a retrospective cohort of patients with MS. METHODS: In rodents, we evaluated pharmacokinetic and pharmacodynamic effects induced by DMF and MEF monotherapies or in combination (DMF/MEF). Clinical implications were investigated in a retrospective, observational analysis of patients with MS treated with DMF/MEF (n = 36). RESULTS: In rodents and cynomolgus monkeys, monomethyl fumarate (MMF, the primary metabolite of DMF) exhibited higher brain penetration, whereas MEF was preferentially partitioned into the kidney. In mice, transcriptional profiling for DMF and MEF alone identified both common and distinct pharmacodynamic responses, with almost no overlap between DMF- and MEF-induced differentially expressed gene profiles in immune tissues. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated oxidative stress response pathway was exclusively regulated by DMF, whereas apoptosis pathways were activated by MEF. DMF/MEF treatment demonstrated that DMF and MEF functionally interact to modify DMF- and MEF-specific responses in unpredictable ways. In patients with MS, DMF/MEF treatment led to early and pronounced suppression of lymphocytes, predominantly CD8+ T cells. In a multivariate regression analysis, the absolute lymphocyte count (ALC) was associated with age at therapy start, baseline ALC, and DMF/MEF dosage but not with previous immunosuppressive medication and sex. Furthermore, the ALC increased in a small cohort of patients with MS (n = 6/7) after switching from DMF/MEF to DMF monotherapy. CONCLUSIONS: Fumaric acid esters exhibit different biodistribution and may elicit different biological responses; furthermore, pharmacodynamic effects of combinations differ unpredictably from monotherapy. The strong potential to induce lymphopenia in patients with MS may be a result of activation of apoptosis pathways by MEF compared with DMF.


Asunto(s)
Dimetilfumarato/química , Dimetilfumarato/farmacología , Fumaratos/química , Fumaratos/farmacología , Esclerosis Múltiple/tratamiento farmacológico , Animales , Estudios Transversales , Dimetilfumarato/uso terapéutico , Femenino , Fumaratos/uso terapéutico , Perfilación de la Expresión Génica/métodos , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/sangre , Esclerosis Múltiple/genética , Ratas , Ratas Sprague-Dawley , Estudios Retrospectivos
19.
Bioorg Med Chem Lett ; 20(7): 2264-9, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20188554

RESUMEN

Modifying FTY720, an immunosuppressant modulator, led to a new series of well phosphorylated tetralin analogs as potent S1P1 receptor agonists. The stereochemistry effect of tetralin ring was probed, and (-)-(R)-2-amino-2-((S)-6-octyl-1,2,3,4-tetrahydronaphthalen-2-yl)propan-1-ol was identified as a good SphK2 substrate and potent S1P1 agonist with good oral bioavailability.


Asunto(s)
Inmunosupresores/farmacología , Profármacos/farmacología , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/metabolismo , Tetrahidronaftalenos/farmacología , Administración Oral , Animales , Cristalografía por Rayos X , Inmunosupresores/química , Inmunosupresores/metabolismo , Inmunosupresores/farmacocinética , Linfopenia/inducido químicamente , Ratones , Modelos Moleculares , Esclerosis Múltiple/tratamiento farmacológico , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Relación Estructura-Actividad , Tetrahidronaftalenos/química , Tetrahidronaftalenos/metabolismo , Tetrahidronaftalenos/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA