Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
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
2.
Methods Mol Biol ; 2049: 419-444, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31602625

RESUMEN

Neurodegenerative diseases (ND) represent a growing, global health crisis, one that lacks any disease-modifying therapeutic strategy. This critical need for new therapies must be met with an exhaustive approach to exploit all tools available. A yeast (Saccharomyces cerevisiae) model of α-synuclein toxicity-the protein causally linked to Parkinson's disease and other synucleinopathies-offers a powerful approach that takes advantage of the unique offerings of this system: tractable genetics, robust high-throughput screening strategies, unparalleled data repositories, powerful computational tools, and extensive evolutionary conservation of fundamental biological pathways. These attributes have enabled genetic and small molecule screens that have revealed toxic phenotypes and drug targets that translate directly to patient-derived iPSC neurons. Extending these insights, recent advances in genetic network analyses have generated the first "humanized" α-synuclein network, which has identified druggable proteins and led to validation of the toxic phenotypes in patient-derived cells. Unbiased phenotypic small molecule screens can identify compounds targeting critical proteins within α-synuclein networks. While identification of direct drug targets for phenotypic screen hits represents a bottleneck, high-throughput chemical genetic methods provide a means to uncover cellular targets and pathways for large numbers of compounds in parallel. Taken together, the yeast α-synuclein model and associated tools can reveal insights into underlying cellular pathologies, lead molecules and their cognate targets, and strategies to translate mechanisms of toxicity and cytoprotection into complex neuronal systems.


Asunto(s)
Saccharomyces cerevisiae/metabolismo , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Redes Reguladoras de Genes , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Sinucleinopatías/tratamiento farmacológico
3.
ACS Chem Neurosci ; 8(9): 2039-2055, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28628299

RESUMEN

The lack of therapies for neurodegenerative diseases arises from our incomplete understanding of their underlying cellular toxicities and the limited number of predictive model systems. It is critical that we develop approaches to identify novel targets and lead compounds. Here, a phenotypic screen of yeast proteinopathy models identified dihydropyrimidine-thiones (DHPM-thiones) that selectively rescued the toxicity caused by ß-amyloid (Aß), the peptide implicated in Alzheimer's disease. Rescue of Aß toxicity by DHPM-thiones occurred through a metal-dependent mechanism of action. The bioactivity was distinct, however, from that of the 8-hydroxyquinoline clioquinol (CQ). These structurally dissimilar compounds strongly synergized at concentrations otherwise not competent to reduce toxicity. Cotreatment ameliorated Aß toxicity by reducing Aß levels and restoring functional vesicle trafficking. Notably, these low doses significantly reduced deleterious off-target effects caused by CQ on mitochondria at higher concentrations. Both single and combinatorial treatments also reduced death of neurons expressing Aß in a nematode, indicating that DHPM-thiones target a conserved protective mechanism. Furthermore, this conserved activity suggests that expression of the Aß peptide causes similar cellular pathologies from yeast to neurons. Our identification of a new cytoprotective scaffold that requires metal-binding underscores the critical role of metal phenomenology in mediating Aß toxicity. Additionally, our findings demonstrate the valuable potential of synergistic compounds to enhance on-target activities, while mitigating deleterious off-target effects. The identification and prosecution of synergistic compounds could prove useful for developing AD therapeutics where combination therapies may be required to antagonize diverse pathologies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Clioquinol/farmacología , Metales/metabolismo , Fármacos Neuroprotectores/farmacología , Tionas/farmacología , Péptidos beta-Amiloides/toxicidad , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Clioquinol/toxicidad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Iones/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Tionas/toxicidad , Levaduras
4.
Science ; 342(6161): 979-83, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24158909

RESUMEN

α-Synuclein (α-syn) is a small lipid-binding protein implicated in several neurodegenerative diseases, including Parkinson's disease, whose pathobiology is conserved from yeast to man. There are no therapies targeting these underlying cellular pathologies, or indeed those of any major neurodegenerative disease. Using unbiased phenotypic screens as an alternative to target-based approaches, we discovered an N-aryl benzimidazole (NAB) that strongly and selectively protected diverse cell types from α-syn toxicity. Three chemical genetic screens in wild-type yeast cells established that NAB promoted endosomal transport events dependent on the E3 ubiquitin ligase Rsp5/Nedd4. These same steps were perturbed by α-syn itself. Thus, NAB identifies a druggable node in the biology of α-syn that can correct multiple aspects of its underlying pathology, including dysfunctional endosomal and endoplasmic reticulum-to-Golgi vesicle trafficking.


Asunto(s)
Bencimidazoles/farmacología , Citoprotección , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Redes Reguladoras de Genes/efectos de los fármacos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas de Saccharomyces cerevisiae/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitina-Proteína Ligasas/genética , alfa-Sinucleína/metabolismo , Animales , Bencimidazoles/química , Caenorhabditis elegans , Células Cultivadas , Evaluación Preclínica de Medicamentos , Ubiquitina-Proteína Ligasas Nedd4 , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Ratas , Saccharomyces cerevisiae/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
5.
Drug Discov Today Technol ; 10(1): e121-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24050240

RESUMEN

Parkinson's disease (PD) is a devastating neurodegenerative disease that affects over one million patients in the US. Yet, no disease modifying drugs exist, only those that temporarily alleviate symptoms. Because of its poorly defined and highly complex disease etiology, it is essential to embrace unbiased and innovative approaches for identifying new chemical entities that target the underlying toxicities associated with PD. Traditional target-based drug discovery paradigm can suffer from a bias toward a small number of potential targets. Phenotypic screening of both genetic and pharmacological PD models offers an alternative approach to discover compounds that target the initiating causes and effectors of cellular toxicity. The relative paucity of reported phenotypic screens illustrates the intrinsic difficulty in establishing model systems that are both biologically meaningful and adaptable to high-throughput screening. Parallel advances in PD models and in vivo screening technologies will help create opportunities for identifying new therapeutic leads with unanticipated, breakthrough mechanisms of action.


Asunto(s)
Evaluación Preclínica de Medicamentos , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos , Fenotipo , Levaduras/genética , alfa-Sinucleína/genética
6.
Dis Model Mech ; 3(3-4): 194-208, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20038714

RESUMEN

alpha-Synuclein (alpha-syn) is a small lipid-binding protein involved in vesicle trafficking whose function is poorly characterized. It is of great interest to human biology and medicine because alpha-syn dysfunction is associated with several neurodegenerative disorders, including Parkinson's disease (PD). We previously created a yeast model of alpha-syn pathobiology, which established vesicle trafficking as a process that is particularly sensitive to alpha-syn expression. We also uncovered a core group of proteins with diverse activities related to alpha-syn toxicity that is conserved from yeast to mammalian neurons. Here, we report that a yeast strain expressing a somewhat higher level of alpha-syn also exhibits strong defects in mitochondrial function. Unlike our previous strain, genetic suppression of endoplasmic reticulum (ER)-to-Golgi trafficking alone does not suppress alpha-syn toxicity in this strain. In an effort to identify individual compounds that could simultaneously rescue these apparently disparate pathological effects of alpha-syn, we screened a library of 115,000 compounds. We identified a class of small molecules that reduced alpha-syn toxicity at micromolar concentrations in this higher toxicity strain. These compounds reduced the formation of alpha-syn foci, re-established ER-to-Golgi trafficking and ameliorated alpha-syn-mediated damage to mitochondria. They also corrected the toxicity of alpha-syn in nematode neurons and in primary rat neuronal midbrain cultures. Remarkably, the compounds also protected neurons against rotenone-induced toxicity, which has been used to model the mitochondrial defects associated with PD in humans. That single compounds are capable of rescuing the diverse toxicities of alpha-syn in yeast and neurons suggests that they are acting on deeply rooted biological processes that connect these toxicities and have been conserved for a billion years of eukaryotic evolution. Thus, it seems possible to develop novel therapeutic strategies to simultaneously target the multiple pathological features of PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Mitocondrias/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Animales , Antiparkinsonianos/farmacología , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Evaluación Preclínica de Medicamentos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/ultraestructura , Perfilación de la Expresión Génica , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/patología , Transporte de Proteínas/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Rotenona/toxicidad , Saccharomyces cerevisiae/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Relación Estructura-Actividad , alfa-Sinucleína/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA