Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell Tissue Res ; 369(3): 445-454, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28466093

RESUMEN

Myeloperoxidase (MPO) is a key enzyme in inflammatory and degenerative processes, although conflicting reports have been presented concerning its expression in the brain. We studied the cellular localization of MPO and compared numbers of MPO cells in various brain regions between neurologically healthy individuals and patients with Parkinson's disease (PD) or Alzheimer's disease (AD; n = 10-25). We also investigated two rodent PD models. MPO immunoreactivity (ir) was detected in monocytes, perivascular macrophages and amoeboid microglia in the human brain parenchyma, whereas no co-localization with glial fibrillary acidic protein (GFAP) ir was observed. In the midbrain, caudate and putamen, we found a significant increase of MPO-immunoreactive cells in PD compared with control brains, whereas in the cerebellum, no difference was apparent. MPO ir was detected neither in neurons nor in occasional small beta-amyloid-immunoreactive plaques in PD or control cases. In the frontal cortex of AD patients, we found significantly more MPO-immunoreactive cells compared with control cases, together with intense MPO ir in extracellular plaques. In the hippocampus of several AD cases, MPO-like ir was observed in some pyramidal neurons. Neither rapid dopamine depletion in the rat PD model, nor slow degeneration of dopamine neurons in MitoPark mice induced the expression of MPO ir in any brain region. MPO mRNA was not detectable with radioactive in situ hybridization in any human or rodent brain area, although myeloid cells from bone marrow displayed clear MPO signals. Our results indicate significant increases of MPO-immunoreactive cells in brain regions affected by neurodegeneration in PD and AD, supporting investigations of MPO inhibitors in novel treatment strategies.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/enzimología , Encéfalo/patología , Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , Peroxidasa/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/enzimología , Enfermedad de Parkinson/enzimología , Ratas Sprague-Dawley
2.
Proc Natl Acad Sci U S A ; 110(19): 7916-21, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23620518

RESUMEN

It is well-established that subcompartments of endoplasmic reticulum (ER) are in physical contact with the mitochondria. These lipid raft-like regions of ER are referred to as mitochondria-associated ER membranes (MAMs), and they play an important role in, for example, lipid synthesis, calcium homeostasis, and apoptotic signaling. Perturbation of MAM function has previously been suggested in Alzheimer's disease (AD) as shown in fibroblasts from AD patients and a neuroblastoma cell line containing familial presenilin-2 AD mutation. The effect of AD pathogenesis on the ER-mitochondria interplay in the brain has so far remained unknown. Here, we studied ER-mitochondria contacts in human AD brain and related AD mouse and neuronal cell models. We found uniform distribution of MAM in neurons. Phosphofurin acidic cluster sorting protein-2 and σ1 receptor, two MAM-associated proteins, were shown to be essential for neuronal survival, because siRNA knockdown resulted in degeneration. Up-regulated MAM-associated proteins were found in the AD brain and amyloid precursor protein (APP)Swe/Lon mouse model, in which up-regulation was observed before the appearance of plaques. By studying an ER-mitochondria bridging complex, inositol-1,4,5-triphosphate receptor-voltage-dependent anion channel, we revealed that nanomolar concentrations of amyloid ß-peptide increased inositol-1,4,5-triphosphate receptor and voltage-dependent anion channel protein expression and elevated the number of ER-mitochondria contact points and mitochondrial calcium concentrations. Our data suggest an important role of ER-mitochondria contacts and cross-talk in AD pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Receptor Cross-Talk , Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Células CHO , Calcio/metabolismo , Línea Celular Tumoral , Cricetinae , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Hipocampo/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato , Microdominios de Membrana/metabolismo , Ratones , Mutación , Neuronas/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores sigma/metabolismo , Fracciones Subcelulares/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Receptor Sigma-1
3.
Biochim Biophys Acta ; 1832(12): 2352-67, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24075941

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene represent the most common genetic cause of Parkinson's disease (PD). However, LRRK2 function and molecular mechanisms causing the parkinsonian phenotype remain widely unknown. Most of LRRK2 knockdown and overexpression models strengthen the relevance of LRRK2 in regulating neurite outgrowth. We have recently identified ARHGEF7 as the first guanine nucleotide exchange factor (GEF) of LRRK2. This GEF is influencing neurite outgrowth through regulation of actin polymerization. Here, we examined the expression profile of neuroblastoma cells with reduced LRRK2 and ARHGEF7 levels to identify additional partners of LRRK2 in this process. Tropomyosins (TPMs), and in particular TPM4, were the most interesting candidates next to other actin cytoskeleton regulating transcripts in this dataset. Subsequently, enhanced neurite branching was shown using primary hippocampal neurons of LRRK2 knockdown animals. Furthermore, we observed an enhanced number of growth cones per neuron and a mislocalization and dysregulation of ARHGEF7 and TPM4 in these neuronal compartments. Our results reveal a fascinating connection between the neurite outgrowth phenotype of LRRK2 models and the regulation of actin polymerization directing further investigations of LRRK2-related pathogenesis.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Conos de Crecimiento/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Tropomiosina/metabolismo , Animales , Biomarcadores/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Técnicas para Inmunoenzimas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Noqueados , Células 3T3 NIH , Neuritas/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/citología , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/genética , Tropomiosina/genética
4.
Neurobiol Dis ; 71: 345-58, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25174890

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset, autosomal dominant Parkinson's disease (PD). The clinical and neurochemical features of LRRK2-linked PD are similar to idiopathic disease although neuropathology is somewhat heterogeneous. Dominant mutations in LRRK2 precipitate neurodegeneration through a toxic gain-of-function mechanism which can be modeled in transgenic mice overexpressing human LRRK2 variants. A number of LRRK2 transgenic mouse models have been developed that display abnormalities in dopaminergic neurotransmission and alterations in tau metabolism yet without consistently inducing dopaminergic neurodegeneration. To directly explore the impact of mutant LRRK2 on the nigrostriatal dopaminergic pathway, we developed conditional transgenic mice that selectively express human R1441C LRRK2 in dopaminergic neurons from the endogenous murine ROSA26 promoter. The expression of R1441C LRRK2 does not induce the degeneration of substantia nigra dopaminergic neurons or striatal dopamine deficits in mice up to 2years of age, and fails to precipitate abnormal protein inclusions containing alpha-synuclein, tau, ubiquitin or autophagy markers (LC3 and p62). Furthermore, mice expressing R1441C LRRK2 exhibit normal motor activity and olfactory function with increasing age. Intriguingly, the expression of R1441C LRRK2 induces age-dependent abnormalities of the nuclear envelope in nigral dopaminergic neurons including reduced nuclear circularity and increased invaginations of the nuclear envelope. In addition, R1441C LRRK2 mice display increased neurite complexity of cultured midbrain dopaminergic neurons. Collectively, these novel R1441C LRRK2 conditional transgenic mice reveal altered dopaminergic neuronal morphology with advancing age, and provide a useful tool for exploring the pathogenic mechanisms underlying the R1441C LRRK2 mutation in PD.


Asunto(s)
Nucléolo Celular/patología , Neuronas Dopaminérgicas/ultraestructura , Mesencéfalo/citología , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Arginina/genética , Células Cultivadas , Cisteína/genética , Neuronas Dopaminérgicas/patología , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Transgénicos
5.
Hum Mol Genet ; 21(11): 2420-31, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22357653

RESUMEN

Mutations in the genes encoding LRRK2 and α-synuclein cause autosomal dominant forms of familial Parkinson's disease (PD). Fibrillar forms of α-synuclein are a major component of Lewy bodies, the intracytoplasmic proteinaceous inclusions that are a pathological hallmark of idiopathic and certain familial forms of PD. LRRK2 mutations cause late-onset familial PD with a clinical, neurochemical and, for the most part, neuropathological phenotype that is indistinguishable from idiopathic PD. Importantly, α-synuclein-positive Lewy bodies are the most common pathology identified in the brains of PD subjects harboring LRRK2 mutations. These observations may suggest that LRRK2 functions in a common pathway with α-synuclein to regulate its aggregation. To explore the potential pathophysiological interaction between LRRK2 and α-synuclein in vivo, we modulated LRRK2 expression in a well-established human A53T α-synuclein transgenic mouse model with transgene expression driven by the hindbrain-selective prion protein promoter. Deletion of LRRK2 or overexpression of human G2019S-LRRK2 has minimal impact on the lethal neurodegenerative phenotype that develops in A53T α-synuclein transgenic mice, including premature lethality, pre-symptomatic behavioral deficits and human α-synuclein or glial neuropathology. We also find that endogenous or human LRRK2 and A53T α-synuclein do not interact together to influence the number of nigrostriatal dopaminergic neurons. Taken together, our data suggest that α-synuclein-related pathology, which occurs predominantly in the hindbrain of this A53T α-synuclein mouse model, occurs largely independently from LRRK2 expression. These observations fail to provide support for a pathophysiological interaction of LRRK2 and α-synuclein in vivo, at least within neurons of the mouse hindbrain.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , alfa-Sinucleína/genética , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/metabolismo
6.
FASEB J ; 25(4): 1345-52, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21163861

RESUMEN

The serine-protease OMI/HTRA2, required for several cellular processes, including mitochondrial function, autophagy, chaperone activity, and apoptosis, has been implicated in the pathogenesis of both Alzheimer's disease (AD) and Parkinson's disease (PD). Western blot quantification of OMI/HTRA2 in frontal cortex of patients with AD (n=10) and control subjects (n=10) in two separate materials indicated reduced processed (active, 35 kDa) OMI/HTRA2 levels, whereas unprocessed (50 kDa) enzyme levels were not significantly different between the groups. Interestingly, the specific protease activity of OMI/HTRA2 was found to be significantly increased in patients with AD (n=10) compared to matched control subjects (n=10) in frontal cortex in two separate materials. Comparison of OMI/HTRA2 mRNA levels in frontal cortex and hippocampus, two brain areas particularly affected by AD, indicated similar levels in patients with AD (n=10) and matched control subjects (n=10). In addition, we analyzed the occurrence of the OMI/HTRA2 variants A141S and G399S in Swedish case-control materials for AD and PD and found a weak association of A141S with AD, but not with PD. In conclusion, our genetic, histological, and biochemical findings give further support to an involvement of OMI/HTRA2 in the pathology of AD; however, further studies are needed to clarify the role of this gene in neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas Mitocondriales/genética , Serina Endopeptidasas/genética , Anciano , Estudios de Casos y Controles , Corteza Cerebral/enzimología , Frecuencia de los Genes , Serina Peptidasa A2 que Requiere Temperaturas Altas , Hipocampo/enzimología , Humanos , Persona de Mediana Edad , Proteínas Mitocondriales/metabolismo , Mutación Missense , Enfermedad de Parkinson/genética , Serina Endopeptidasas/metabolismo
7.
FASEB J ; 25(4): 1333-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21233488

RESUMEN

Parkinson's disease (PD) involves progressive loss of nigrostriatal dopamine (DA) neurons over an extended period of time. Mitochondrial damage may lead to PD, and neurotoxins affecting mitochondria are widely used to produce degeneration of the nigrostriatal circuitry. Deletion of the mitochondrial transcription factor A gene (Tfam) in C57BL6 mouse DA neurons leads to a slowly progressing parkinsonian phenotype in which motor impairment is first observed at ~12 wk of age. L-DOPA treatment improves motor dysfunction in these "MitoPark" mice, but this declines when DA neuron loss is more complete. To investigate early neurobiological events potentially contributing to PD, we compared the neurochemical and electrophysiological properties of the nigrostriatal circuit in behaviorally asymptomatic 6- to 8-wk-old MitoPark mice and age-matched control littermates. Release, but not uptake of DA, was impaired in MitoPark mouse striatal brain slices, and nigral DA neurons lacked characteristic pacemaker activity compared with control mice. Also, hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channel function was reduced in MitoPark DA neurons, although HCN messenger RNA was unchanged. This study demonstrates altered nigrostriatal function that precedes behavioral parkinsonian symptoms in this genetic PD model. A full understanding of these presymptomatic cellular properties may lead to more effective early treatments of PD.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Proteínas de Unión al ADN/genética , Proteínas Mitocondriales/genética , Neuronas/fisiología , Enfermedad de Parkinson/fisiopatología , Factores de Transcripción/genética , Animales , Cuerpo Estriado , Modelos Animales de Enfermedad , Dopamina/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Sustancia Negra
8.
Neurosci Lett ; 784: 136767, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35779693

RESUMEN

INTRODUCTION: Genetic variants in the Beta-glucocerebrosidase gene (GBA1) is a known risk factor for Parkinson's disease. The GBA1 mutations L444P, N370S and many other have been shown to associate with the disease in populations with diverse background. Some GBA1 polymorphisms have a less pronounced effect, and their pathogenicity has been debated. We have previously found associations with L444P, N370S and E326K and Parkinson's disease in Sweden. METHOD: In this study we used pyrosequencing to genotype the T369M variant in a large Swedish cohort consisting of 1,131 patients with idiopathic Parkinson's disease, and 1,594 control subjects to evaluate the possibility of this variant conferring an increased risk for Parkinson's disease. RESULTS: The minor allele frequency was 2.15% in patients and 1.76% in controls. Statistical analysis showed that there was no significant difference in allele frequency between patients and control subjects, p-value 0.37, Odds Ratio 1.23 with a 95% confidence interval of 0.82-1.83. CONCLUSION: Our results suggest that T369M is not a risk factor for Parkinson's disease in the Swedish population.


Asunto(s)
Glucosilceramidasa , Enfermedad de Parkinson , Glucosilceramidasa/genética , Humanos , Mutación , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/genética , Factores de Riesgo , Suecia/epidemiología
9.
Neurobiol Aging ; 45: 212.e5-212.e11, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27255555

RESUMEN

Several genetic studies have demonstrated an association between mutations in glucocerebrosidase (GBA), originally implicated in Gaucher's disease, and an increased risk of Parkinson's disease (PD). We have investigated the possible involvement of genetic GBA variations in PD in the Swedish population. Three GBA variants, E326K, N370S, and L444P were screened in the largest Swedish Parkinson cohort reported to date; 1625 cases and 2025 control individuals. We found a significant association with high effect size of the rare variant L444P with PD (odds ratio 8.17; 95% confidence interval: 2.51-26.23; p-value = 0.0020) and a significant association of the common variant E326K (odds ratio 1.60; 95% confidence interval: 1.16-2.22; p-value = 0.026). The rare variant N370S showed a trend for association. Most L444P carriers (68%) were found to reside in northern Sweden, which is consistent with a higher prevalence of Gaucher's disease in this part of the country. Our findings support the role of GBA mutations as risk factors for PD and point to lysosomal dysfunction as a mechanism contributing to PD etiology.


Asunto(s)
Estudios de Asociación Genética , Glucosilceramidasa/genética , Mutación , Enfermedad de Parkinson/genética , Anciano , Femenino , Humanos , Masculino , Suecia
10.
Behav Brain Res ; 227(1): 252-7, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22079585

RESUMEN

Alcohol dehydrogenases (ADH) catalyze the reversible metabolism of many types of alcohols and aldehydes to prevent the possible toxic accumulation of these compounds. ADHs are of interest in Parkinson's disease (PD) since these compounds can be harmful to dopamine (DA) neurons. Genetic variants in ADH1C and ADH4 have been found to associate with PD and lack of Adh4 gene activity in a mouse model has recently been reported to induce changes in the DA system. Adh1 knockout (Adh1-/-) and Adh1/4 double knockout (Adh1/4-/-) mice were investigated for possible changes in DA system related activity, biochemical parameters and olfactory function compared to wild-type (WT) mice. Locomotor activity was tested at ∼7 (adult) and >15 months of age to mimic the late onset of PD. Adh1-/- and Adh1/4-/- mice displayed a significantly higher spontaneous locomotor activity than WT littermates. Both apomorphine and d-amphetamine increased total distance activity in Adh1-/- mice at both age intervals and in Adh1/4-/- mice at 7 months of age compared to WT mice. No significant changes were found regarding olfactory function, however biochemical data showed decreased 3,4-dihydroxyphenylacetic acid (DOPAC)/DA ratios in the olfactory bulb and decreased homovanillic acid (HVA)/DA ratios in the olfactory bulb, frontal cortex and striatum of Adh1/4-/- mice compared to WT mice. Our results suggest that lack of Adh1 alone or Adh1 and Adh4 together lead to changes in DA system related behavior, and that these knockout mice might be possible rodent models to study presymptomatic PD.


Asunto(s)
Alcohol Deshidrogenasa/deficiencia , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología , Ácido 3,4-Dihidroxifenilacético/metabolismo , Análisis de Varianza , Animales , Apomorfina/farmacología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Técnicas Electroquímicas , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Femenino , Genotipo , Ácido Homovanílico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Trastornos del Olfato/etiología , Trastornos del Olfato/genética , Enfermedad de Parkinson/tratamiento farmacológico , Olfato/efectos de los fármacos , Olfato/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA