Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Dis Model Mech ; 15(6)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35419585

RESUMEN

Heterozygous mutations in the GBA1 gene - encoding lysosomal glucocerebrosidase (GCase) - are the most common genetic risk factors for Parkinson's disease (PD). Experimental evidence suggests a correlation between decreased GCase activity and accumulation of alpha-synuclein (aSyn). To enable a better understanding of the relationship between aSyn and GCase activity, we developed and characterized two mouse models that investigate aSyn pathology in the context of reduced GCase activity. The first model used constitutive overexpression of wild-type human aSyn in the context of the homozygous GCase activity-reducing D409V mutant form of GBA1. Although increased aSyn pathology and grip strength reductions were observed in this model, the nigrostriatal system remained largely intact. The second model involved injection of aSyn preformed fibrils (PFFs) into the striatum of the homozygous GBA1 D409V knock-in mouse model. The GBA1 D409V mutation did not exacerbate the pathology induced by aSyn PFF injection. This study sheds light on the relationship between aSyn and GCase in mouse models, highlighting the impact of model design on the ability to model a relationship between these proteins in PD-related pathology.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ratones , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
2.
Per Med ; 14(1): 13-15, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-29749821

RESUMEN

This interview was conducted by Lauren Pulling, Editor of Neurology Central. The full interview can be accessed on Neurology Central as part of the NCTalks podcast series which is available at: www.neurology-central.com/2016/11/28/nctalks-mark-frasier-precision-medicine-parkinsons-disease Mark Frasier co-manages a team of research professionals who stay closely linked to the Parkinson's research community in order to develop an aggressive and innovative agenda for accelerating research and drug development for Parkinson's disease. He also supports the foundation's priority interest in developing biomarkers for Parkinson's disease that will accelerate clinical trials of new drugs. He earned an undergraduate degree in biochemistry from the University of Dayton and a PhD in pharmacology from Loyola University Chicago. He completed his postdoctoral work in the Neuroscience Discovery Research Group at Eli Lilly, Inc., in Indianapolis, Indiana, where he worked on drug discovery research in Parkinson's and Alzheimer's disease. He joined the Michael J. Fox Foundation in 2006 and is now Senior Vice President of Research Programs.


Asunto(s)
Enfermedad de Parkinson/genética , Trastornos Parkinsonianos/genética , Biomarcadores , Descubrimiento de Drogas , Humanos , Neurología , Neurociencias , Enfermedad de Parkinson/terapia , Trastornos Parkinsonianos/terapia , Medicina de Precisión , Investigación
3.
4.
Neurobiol Dis ; 70: 190-203, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24969022

RESUMEN

Recessively inherited loss-of-function mutations in the PTEN-induced putative kinase 1(Pink1), DJ-1 (Park7) and Parkin (Park2) genes are linked to familial cases of early-onset Parkinson's disease (PD). As part of its strategy to provide more tools for the research community, The Michael J. Fox Foundation for Parkinson's Research (MJFF) funded the generation of novel rat models with targeted disruption ofPink1, DJ-1 or Parkin genes and determined if the loss of these proteins would result in a progressive PD-like phenotype. Pathological, neurochemical and behavioral outcome measures were collected at 4, 6 and 8months of age in homozygous KO rats and compared to wild-type (WT) rats. Both Pink1 and DJ-1 KO rats showed progressive nigral neurodegeneration with about 50% dopaminergic cell loss observed at 8 months of age. ThePink1 KO and DJ-1 KO rats also showed a two to three fold increase in striatal dopamine and serotonin content at 8 months of age. Both Pink1 KO and DJ-1 KO rats exhibited significant motor deficits starting at 4months of age. However, Parkin KO rats displayed normal behaviors with no neurochemical or pathological changes. These results demonstrate that inactivation of the Pink1 or DJ-1 genes in the rat produces progressive neurodegeneration and early behavioral deficits, suggesting that these recessive genes may be essential for the survival of dopaminergic neurons in the substantia nigra (SN). These MJFF-generated novel rat models will assist the research community to elucidate the mechanisms by which these recessive genes produce PD pathology and potentially aid in therapeutic development.


Asunto(s)
Proteínas Asociadas a Microtúbulos/deficiencia , Trastornos Parkinsonianos/fisiopatología , Fenotipo , Proteínas Quinasas/deficiencia , Ubiquitina-Proteína Ligasas/deficiencia , Envejecimiento , Animales , Animales Modificados Genéticamente , Encéfalo/patología , Encéfalo/fisiopatología , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/fisiología , Técnicas de Inactivación de Genes , Genes Recesivos , Masculino , Proteínas Asociadas a Microtúbulos/genética , Actividad Motora/fisiología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Proteína Desglicasa DJ-1 , Proteínas Quinasas/genética , Ratas Long-Evans , Serotonina/metabolismo , Ubiquitina-Proteína Ligasas/genética
5.
J Parkinsons Dis ; 3(4): 565-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24192754

RESUMEN

The development of an α-synuclein imaging agent could be transformative for Parkinson's disease research and drug development. The ability to image α-synuclein in the brain would enable tracking of the degree and location of pathology over time and monitoring of therapies aimed at reducing α-synuclein levels. The Michael J. Fox Foundation has assembled a consortium of researchers to develop an α-synuclein radiotracer for use in positron emission tomography (PET) imaging studies. While this poses a number of challenges they should not be insurmountable and lessons learned from the development of tau radiotracers should provide valuable insights.


Asunto(s)
Encéfalo/diagnóstico por imagen , Enfermedad de Parkinson/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , alfa-Sinucleína/metabolismo , Biomarcadores , Investigación Biomédica , Encéfalo/patología , Progresión de la Enfermedad , Diagnóstico Precoz , Humanos , Enfermedad de Parkinson/patología , Radiofármacos , alfa-Sinucleína/química
6.
PLoS One ; 8(11): e80705, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244710

RESUMEN

The objective of this study was to evaluate the pathology time course of the LRRK2 knockout rat model of Parkinson's disease at 1-, 2-, 4-, 8-, 12-, and 16-months of age. The evaluation consisted of histopathology and ultrastructure examination of selected organs, including the kidneys, lungs, spleen, heart, and liver, as well as hematology, serum, and urine analysis. The LRRK2 knockout rat, starting at 2-months of age, displayed abnormal kidney staining patterns and/or morphologic changes that were associated with higher serum phosphorous, creatinine, cholesterol, and sorbitol dehydrogenase, and lower serum sodium and chloride compared to the LRRK2 wild-type rat. Urinalysis indicated pronounced changes in LRRK2 knockout rats in urine specific gravity, total volume, urine potassium, creatinine, sodium, and chloride that started as early as 1- to 2-months of age. Electron microscopy of 16-month old LRRK2 knockout rats displayed an abnormal kidney, lung, and liver phenotype. In contrast, there were equivocal or no differences in the heart and spleen of LRRK2 wild-type and knockout rats. These findings partially replicate data from a recent study in 4-month old LRRK2 knockout rats and expand the analysis to demonstrate that the renal and possibly lung and liver abnormalities progress with age. The characterization of LRRK2 knockout rats may prove to be extremely valuable in understanding potential safety liabilities of LRRK2 kinase inhibitor therapeutics for treating Parkinson's disease.


Asunto(s)
Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Riñón/metabolismo , Riñón/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Masculino , Fenotipo , Ratas , Ratas Mutantes , Bazo/metabolismo , Bazo/patología
7.
Dis Model Mech ; 6(6): 1316-24, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24046356

RESUMEN

Progress in Parkinson's disease (PD) research and therapeutic development is hindered by many challenges, including a need for robust preclinical animal models. Limited availability of these tools is due to technical hurdles, patent issues, licensing restrictions and the high costs associated with generating and distributing these animal models. Furthermore, the lack of standardization of phenotypic characterization and use of varying methodologies has made it difficult to compare outcome measures across laboratories. In response, The Michael J. Fox Foundation for Parkinson's Research (MJFF) is directly sponsoring the generation, characterization and distribution of preclinical rodent models, enabling increased access to these crucial tools in order to accelerate PD research. To date, MJFF has initiated and funded the generation of 30 different models, which include transgenic or knockout models of PD-relevant genes such as Park1 (also known as Park4 and SNCA), Park8 (LRRK2), Park7 (DJ-1), Park6 (PINK1), Park2 (Parkin), VPS35, EiF4G1 and GBA. The phenotypic characterization of these animals is performed in a uniform and streamlined manner at independent contract research organizations. Finally, MJFF created a central repository at The Jackson Laboratory (JAX) that houses both non-MJFF and MJFF-generated preclinical animal models. Funding from MJFF, which subsidizes the costs involved in transfer, rederivation and colony expansion, has directly resulted in over 2500 rodents being distributed to the PD community for research use.


Asunto(s)
Investigación Biomédica , Modelos Animales , Enfermedad de Parkinson , Animales , Animales Modificados Genéticamente , Humanos , Enfermedad de Parkinson/genética , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...