Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
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.
RNA Biol ; 13(1): 15-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26669816

RESUMEN

Adenosine deaminases bind double stranded RNA and convert adenosine to inosine. Editing creates multiple isoforms of neurotransmitter receptors, such as with Gria2. Adar2 KO mice die of seizures shortly after birth, but if the Gria2 Q/R editing site is mutated to mimic the edited version then the animals are viable. We performed RNA-Seq on frontal cortices of Adar2(-/-) Gria2(R/R) mice and littermates. We found 56 editing sites with significantly diminished editing levels in Adar2 deficient animals with the majority in coding regions. Only two genes and 3 exons showed statistically significant differences in expression levels. This work illustrates that ADAR2 is important in site-specific changes of protein coding sequences but has relatively modest effects on gene expression and splicing in the adult mouse frontal cortex.


Asunto(s)
Adenosina Desaminasa/metabolismo , Expresión Génica , Empalme del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ARN/métodos , Adenosina Desaminasa/genética , Animales , Lóbulo Frontal/metabolismo , Técnicas de Inactivación de Genes , Ratones , Mutación , Edición de ARN , Proteínas de Unión al ARN/genética
3.
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
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.
Proc Natl Acad Sci U S A ; 107(1): 389-94, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20018672

RESUMEN

The "One neuron-one neurotransmitter" concept has been challenged frequently during the last three decades, and the coexistence of neurotransmitters in individual neurons is now regarded as a common phenomenon. The functional significance of neurotransmitter coexistence is, however, less well understood. Several studies have shown that a subpopulation of dopamine (DA) neurons in the ventral tegmental area (VTA) expresses the vesicular glutamate transporter 2 (VGLUT2) and has been suggested to use glutamate as a cotransmitter. The VTA dopamine neurons project to limbic structures including the nucleus accumbens, and are involved in mediating the motivational and locomotor activating effects of psychostimulants. To determine the functional role of glutamate cotransmission by these neurons, we deleted VGLUT2 in DA neurons by using a conditional gene-targeting approach in mice. A DAT-Cre/Vglut2Lox mouse line (Vglut2(f/f;DAT-Cre) mice) was produced and analyzed by in vivo amperometry as well as by several behavioral paradigms. Although basal motor function was normal in the Vglut2(f/f;DAT-Cre) mice, their risk-taking behavior was altered. Interestingly, in both home-cage and novel environments, the gene targeted mice showed a greatly blunted locomotor response to the psychostimulant amphetamine, which acts via the midbrain DA system. Our results show that VGLUT2 expression in DA neurons is required for normal emotional reactivity as well as for psychostimulant-mediated behavioral activation.


Asunto(s)
Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Neuronas , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Anfetamina/farmacología , Animales , Conducta Animal/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Ácido Glutámico/metabolismo , Masculino , Mesencéfalo/citología , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Distribución Aleatoria , Asunción de Riesgos , Factores Sexuales , Proteína 2 de Transporte Vesicular de Glutamato/genética
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.
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
9.
Cancer Cell ; 6(4): 333-45, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15488757

RESUMEN

Cancer metastases are commonly found in the lymphatic system. Like tumor blood angiogenesis, stimulation of tumor lymphangiogenesis may require the interplay of several tumor-derived growth factors. Here we report that members of the PDGF family act as lymphangiogenic factors. In vitro, PDGF-BB stimulated MAP kinase activity and cell motility of isolated lymphatic endothelial cells. In vivo, PDGF-BB potently induced growth of lymphatic vessels. Expression of PDGF-BB in murine fibrosarcoma cells induced tumor lymphangiogenesis, leading to enhanced metastasis in lymph nodes. These data demonstrate that PDGF-BB is an important growth factor contributing to lymphatic metastasis. Thus, blockage of PDGF-induced lymphangiogenesis may provide a novel approach for prevention and treatment of lymphatic metastasis.


Asunto(s)
Linfangiogénesis/efectos de los fármacos , Metástasis Linfática , Neoplasias/patología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Becaplermina , División Celular/efectos de los fármacos , Línea Celular , Quimiotaxis/efectos de los fármacos , Femenino , Humanos , Sistema Linfático/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Neurobiol Dis ; 40(2): 460-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20656029

RESUMEN

Adenosine A(2A) receptor (A(2A)R) antagonists are being investigated as promising treatment strategy for Parkinson's disease (PD). To test whether A(2A)R antagonists are beneficial in early PD stages we used MitoPark mice, a genetic model with gradual degeneration of DA cells. Daily treatment of young MitoPark mice for eight weeks with the A(2A)R antagonist MSX-3 prevented the reduction of spontaneous locomotor activity observed in saline or L-DOPA treated animals. Chronic A(2A)R antagonist treatment neither induced desensitization of receptors nor accumulation of the drug in brain tissue. Despite beneficial effects on behavior, which are not improved upon addition of a low dose of L-DOPA, the characteristic decline of dopamine levels was not changed. Our results indicate that effective dosing with A(2A)R antagonists should be tested as monotherapy in early PD, and serves to remind us that positive behavioral effects of such treatment need not be reflected in rescue of striatal dopamine levels.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Dopamina/metabolismo , Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Xantinas/farmacología , Antagonistas del Receptor de Adenosina A2/efectos adversos , Animales , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , Levodopa/farmacología , Ratones , Ratones Endogámicos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Factores de Tiempo , Resultado del Tratamiento , Xantinas/efectos adversos
11.
Hum Mol Genet ; 17(10): 1418-26, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18245781

RESUMEN

Heteroplasmic mitochondrial DNA (mtDNA) mutations (mutations present only in a subset of cellular mtDNA copies) arise de novo during the normal ageing process or may be maternally inherited in pedigrees with mitochondrial disease syndromes. A pathogenic mtDNA mutation causes respiratory chain deficiency only if the fraction of mutated mtDNA exceeds a certain threshold level. These mutations often undergo apparently random mitotic segregation and the levels of normal and mutated mtDNA can vary considerably between cells of the same tissue. In human ageing, segregation of somatic mtDNA mutations leads to mosaic respiratory chain deficiency in a variety of tissues, such as brain, heart and skeletal muscle. A similar pattern of mutation segregation with mosaic respiratory chain deficiency is seen in patients with mitochondrial disease syndromes caused by inherited pathogenic mtDNA mutations. We have experimentally addressed the role of mosaic respiratory chain deficiency in ageing and mitochondrial disease by creating mouse chimeras with a mixture of normal and respiratory chain-deficient neurons in cerebral cortex. We report here that a low proportion (>20%) of respiratory chain-deficient neurons in the forebrain are sufficient to cause symptoms, whereas premature death of the animal occurs only if the proportion is high (>60-80%). The presence of neurons with normal respiratory chain function does not only prevent mortality but also delays the age at which onset of disease symptoms occur. Unexpectedly, respiratory chain-deficient neurons have adverse effect on normal adjacent neurons and induce trans-neuronal degeneration. In summary, our study defines the minimal threshold level of respiratory chain-deficient neurons needed to cause symptoms and also demonstrate that neurons with normal respiratory chain function ameliorate disease progression. Finally, we show that respiratory chain-deficient neurons induce death of normal neurons by a trans-neuronal degeneration mechanism. These findings provide novel insights into the pathogenesis of mosaic respiratory chain deficiency in ageing and mitochondrial disease.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Mitocondriales/metabolismo , Mosaicismo/embriología , Degeneración Nerviosa/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Quimera/genética , Quimera/metabolismo , Quimerismo , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Locomoción , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/mortalidad , Enfermedades Mitocondriales/patología , Actividad Motora , Degeneración Nerviosa/genética
12.
FASEB J ; 22(10): 3509-14, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18606870

RESUMEN

Alterations of brain and plasma alpha-synuclein levels and SNCA gene variability have been implicated in the pathogenesis of Parkinson's disease (PD). We therefore measured alpha-synuclein protein levels in postmortem PD and control cerebellum tissue using Western blot and investigated whether the levels correlated to SNCA genotype. We found markedly decreased alpha-synuclein levels in PD patients (n=16) compared to gender- and age-matched controls (n=14; P=0.004) normalized to alpha-tubulin. We also performed an association study of the noncoding polymorphisms rs2737029 (A/G) and rs356204 (A/G) (intron 4), and of rs356219 (T/C) (3'-region) of SNCA in a Swedish PD case-control material. Using a two-sided chi(2) test, we found significant association of rs2737029 (P=0.003; chi(2)=9.07) and rs356204 (P=0.048; chi(2)=3.91) with disease, strengthening the involvement of SNCA polymorphisms in sporadic PD. Stratification of the human postmortem brain material by genotype of the three investigated polymorphisms, did not indicate any influence of genotype on alpha-synuclein protein levels when comparing PD with controls. Taken together, our findings demonstrate that the investigated Parkinson patients have markedly reduced levels of alpha-synuclein in cerebellum, and that this reduction is general, rather then correlated to the investigated polymorphisms, although two of the polymorphisms also associated with disease in a Swedish material.


Asunto(s)
Cerebelo/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Anciano , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple , Suecia , alfa-Sinucleína/genética
13.
Neurosci Lett ; 452(1): 8-11, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19146923

RESUMEN

Mutations in DJ-1 lead to a monogenic form of early onset recessive parkinsonism. DJ-1 can respond to oxidative stress, which has been proposed to be involved in the pathogenesis of sporadic Parkinson disease (PD). We have recently reported that DJ-1 interacts with mRNA in an oxidation-dependent manner. Here, we confirm interaction of DJ-1 and RNA in human brain using immunoprecipitation followed by quantitative real time PCR. We confirmed previous reports that DJ-1 is more oxidized in cortex from cases of sporadic PD compared to controls. In the same samples, protein and RNA expression was measured for four DJ-1 target genes GPx4, MAPK8IP1, ND2 and ND5. While no alterations in mRNA expression were observed, an increase in protein expression was observed in PD cases for GPx4 and MAPK8IP1. In the same patients, we saw decreased mRNA and protein levels of two mitochondrial targets, ND2 and ND5. These results suggest that these proteins undergo regulation at the post-transcriptional level that may involve translational regulation by DJ-1.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Oncogénicas/genética , Enfermedad de Parkinson/genética , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Encéfalo/metabolismo , Electroforesis en Gel Bidimensional/métodos , Glutatión Peroxidasa/genética , Humanos , Inmunoprecipitación/métodos , NADH Deshidrogenasa/genética , Enfermedad de Parkinson/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proteína Desglicasa DJ-1 , Proteína Fosfatasa 2/genética , Proteínas Proto-Oncogénicas/genética , Selenoproteína W/genética , Proteína Letal Asociada a bcl/genética
14.
Mol Cell Neurosci ; 39(4): 586-91, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18790059

RESUMEN

LRRK2, alpha-synuclein, UCH-L1 and DJ-1 are implicated in the etiology of Parkinson's disease. We show for the first time that increase in striatal alpha-synuclein levels induce increased Lrrk2 mRNA levels while Dj-1 and Uch-L1 are unchanged. We also demonstrate that a mouse strain lacking the dopamine signaling molecule DARPP-32 has significantly reduced levels of both Lrrk2 and alpha-synuclein, while mice carrying a disabling mutation of the DARPP-32 phosphorylation site T34A or lack alpha-synuclein do not show any changes. To test if striatal dopamine depletion influences Lrrk2 or alpha-synuclein expression, we used the neurotoxin 6-hydroxydopamine in rats and MitoPark mice in which there is progressive degeneration of dopamine neurons. Because striatal Lrrk2 and alpha-synuclein levels were not changed by dopamine depletion, we conclude that Lrrk2 and alpha-synuclein mRNA levels are possibly co-regulated, but they are not influenced by striatal dopamine levels.


Asunto(s)
Cuerpo Estriado/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cuerpo Estriado/anatomía & histología , Dopamina/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Oxidopamina/metabolismo , Enfermedad de Parkinson/etiología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , alfa-Sinucleína/genética
15.
FEBS J ; 275(7): 1384-1391, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18279376

RESUMEN

Parkinson's disease (PD), a common progressive neurodegenerative disorder, is characterized by degeneration of dopamine neurons in the substantia nigra and neuronal proteinaceous aggregates called Lewy bodies (LBs). The etiology of PD is probably a combination of environmental and genetic factors. Recent progress in molecular genetics has identified several genes causing PD, including alpha-synuclein, leucine-rich repeat kinase 2 (LRRK2), Parkin, DJ-1 and PTEN-induced kinase 1 (PINK1), many of them coding for proteins found in LBs and/or implicated in mitochondrial function. However, the mechanism(s) leading to the development of the disease have not been identified, despite intensive research. Animal models help us to obtain insights into the mechanisms of several symptoms of PD, allowing us to investigate new therapeutic strategies and, in addition, provide an indispensable tool for basic research. As PD does not arise spontaneously in animals, characteristic and specific functional changes have to be induced by administration of toxins or by genetic manipulations. This review will focus on the comparison of three types of rodent animal models used to study different aspects of PD: (a) animal models using neurotoxins; (b) genetically modified mouse models reproducing findings from PD linkage studies or based on ablation of genes necessary for the development and survival of dopamine neurons; and (c) tissue-specific knockouts in mice targeting dopamine neurons. The advantages and disadvantages of these models are discussed.


Asunto(s)
Modelos Animales de Enfermedad , Neurotoxinas/toxicidad , Enfermedad de Parkinson/genética , Animales , Ratones , Ratones Noqueados , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Ratas , Técnicas Estereotáxicas
16.
Cell Tissue Res ; 334(2): 179-85, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18855017

RESUMEN

The membrane transporter multi-drug resistance 1 (MDR1, P-gp) regulates the bioavailability of endogenous and exogenous compounds and has been implicated in disorders such as Parkinson's disease, cancer, epilepsy, human immunodeficiency virus disease, and inflammatory bowel disease. To promote further understanding of the role of MDR1 in disease, we have characterized cellular MDR1 mRNA expression in post-mortem human and fresh-frozen Sprague-Dawley rat tissues by using radioactive oligonucleotide probe in situ hybridization. We report MDR1 mRNA in human and rat endothelial cells of small vessels in the brain and pia mater. Mdr1 mRNA is also expressed in the blood vessel walls of rat sensory dorsal root and sympathetic ganglia. In peripheral tissues, we have observed MDR1 mRNA in human and rat liver and renal tubules and in human adrenal cortex and the epithelial lining of rat intestine. In female and male reproductive tissues of rat, strong gene activity has been found in steroid-hormone-synthesizing cells. Quantification of MDR1 mRNA in human striatum has revealed reduced levels in Parkinson patients compared with control individuals. The high expression of MDR1 mRNA in blood vessels of the nervous system, in tissues involved in absorption and excretion, and in tissues forming barriers to the environment support the physiological role of MDR1 as a regulator of intracellular levels of endogenous and exogenous compounds.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Enfermedad de Parkinson/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Femenino , Expresión Génica , Humanos , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Distribución Tisular
17.
FEBS J ; 274(5): 1212-23, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17257171

RESUMEN

Parkinson's disease (PD) is a heterogeneous movement disorder characterized by progressive degeneration of dopamine neurons in substantia nigra. We have previously presented genetic evidence for the possible involvement of alcohol and aldehyde dehydrogenases (ADH; ALDH) by identifying genetic variants in ADH1C and ADH4 that associate with PD. The absence of the corresponding mRNA species in the brain led us to the hypothesis that one cause of PD could be defects in the defense systems against toxic aldehydes in the gastrointestinal tract. We investigated cellular expression of Adh1, Adh3, Adh4 and Aldh1 mRNA along the rodent GI tract. Using oligonucleotide in situ hybridization probes, we were able to resolve the specific distribution patterns of closely related members of the ADH family. In both mice and rats, Adh4 is transcribed in the epithelium of tongue, esophagus and stomach, whereas Adh1 was active from stomach to rectum in mice, and in duodenum, colon and rectum in rats. Adh1 and Adh4 mRNAs were present in the mouse gastric mucosa in nonoverlapping patterns, with Adh1 in the gastric glands and Adh4 in the gastric pits. Aldh1 was found in epithelial cells from tongue to jejunum in rats and from esophagus to colon in mice. Adh3 hybridization revealed low mRNA levels in all tissues investigated. The distribution and known physiological functions of the investigated ADHs and Aldh1 are compatible with a role in a defense system, protecting against alcohols, aldehydes and formaldehydes as well as being involved in retinoid metabolism.


Asunto(s)
Alcohol Deshidrogenasa/genética , Alcohol Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación Enzimológica de la Expresión Génica , Mucosa Intestinal/enzimología , Animales , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/enzimología , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Transcripción Genética
18.
Brain Res ; 1184: 10-6, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-17950257

RESUMEN

Mutations in PINK1 (PTEN-induced putative kinase 1) are causal for early onset recessive parkinsonism in humans, characterized by damage to the nigrostriatal system. In situ hybridization studies in rodent brains have suggested a predominantly neuronal expression of PINK1 mRNA but immunocytochemistry of human brain tissue has shown PINK1-like immunoreactivity in both neurons and glia. In this study, we assessed the comparative distribution of PINK1 mRNA in human, rat and mouse brain. We observe that in humans PINK1 message is expressed in neurons with very little to no signal in glia and confirms similar findings in rodent tissue. Highest levels of expression were observed in hippocampus, substantia nigra and cerebellar Purkinje cells. We also show that PINK1 mRNA expression is similar in nigral neurons from neurologically normal controls and sporadic Parkinson's disease cases.


Asunto(s)
Encéfalo/metabolismo , Expresión Génica/fisiología , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Animales , Encéfalo/patología , Humanos , Hibridación in Situ , Ratones , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Proteínas Quinasas/genética , Ratas
19.
Neurosci Lett ; 420(3): 257-62, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17537576

RESUMEN

Mitochondrial (mt) dysfunction has been implicated in Alzheimer's (AD) and Parkinson's disease (PD). Mitochondrial transcription factor A (TFAM) is needed for mtDNA maintenance, regulating mtDNA copy number and is absolutely required for transcriptional initiation at mtDNA promoters. Two genetic variants in TFAM have been reported to be associated with AD in a Caucasian case-control material collected from Germany, Switzerland and Italy. One of these variants was reported to show a tendency for association with AD in a pooled Scottish and Swedish case-control material and the other variant was reported to be associated with AD in a recent meta-analysis. We investigated these two genetic variants, rs1937 and rs2306604, in an AD and a PD case-control material, both from Sweden and found significant genotypic as well as allelic association to marker rs2306604 in the AD case-control material (P=0.05 and P=0.03, respectively), where the A-allele appears to increase risk for developing AD. No association was observed for marker rs1937. We did not find any association in the PD case-control material for either of the two markers. The distribution of the two-locus haplotype frequencies (based on rs1937 and rs2306604) did not differ significantly between affected individuals and controls in the two sample sets. However, the global P-value for haplotypic association testing indicated borderline association in the AD sample set. Our data suggests that the rs2306604 A-allele could be a moderate risk factor for AD, which is supported by the recent meta-analysis.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Unión al ADN/genética , Proteínas Mitocondriales/genética , Enfermedad de Parkinson/genética , Factores de Transcripción/genética , Anciano , Alelos , ADN/genética , Femenino , Frecuencia de los Genes , Variación Genética , Genotipo , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suecia
20.
Physiol Behav ; 92(1-2): 46-53, 2007 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-17599367

RESUMEN

DJ-1 (PARK7) has been implicated in early onset and familial cases of Parkinson's disease (PD). We therefore mapped cellular activity patterns of the DJ-1 gene in human and rodent brain tissue with radioactive in-situ hybridization. In all three mammals mRNA expression was restricted mainly to neurons in all regions analyzed. White matter, such as crus cerebri and capsula interna appeared negative, suggesting that glial cells express DJ-1 at levels below the detection limit of our method. We compared DJ-1 mRNA expression to the neuronal marker UCH-L1, which has also been implicated in PD, and found lower levels for DJ-1 but very similar patterns of expression. Measurement of the signal intensity revealed that human frontal cortex of control cases expressed DJ-1 mRNA more abundantly than other regions such as substantia nigra in the midbrain. Comparing DJ-1 expression in dopamine neurons on hemi-sections from controls and patients we could not detect any difference between 14 controls, 8 idiopathic Parkinson and 5 schizophrenia cases. Of note, DJ-1 is expressed in several other tissues such as the liver, gastrointestinal tract, adrenal and pituitary gland and during embryonic development, while UCH-L1 has a strictly neuronal expression also outside the CNS. We conclude that DJ-1 and UCH-L1, like other genes linked to PD, are not expressed specifically in DA neurons, but instead generally in neurons. The abundant expression of DJ-1 in certain peripheral tissues and of UCH-L1 in peripheral neurons may also be of relevance for the spectrum of symptoms in different forms of PD.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/metabolismo , Proteínas Oncogénicas/metabolismo , Enfermedad de Parkinson/metabolismo , Esquizofrenia/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/citología , Encéfalo/metabolismo , Estudios de Casos y Controles , Dopamina/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Oncogénicas/genética , Proteína Desglicasa DJ-1 , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Transducción de Señal/fisiología , Distribución Tisular , Ubiquitina Tiolesterasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA