Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Pharmacol Res ; 104: 197-205, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26731018

RESUMEN

HSP90 (Heat shock protein 90) is a molecular chaperone protein ubiquitously expressed throughout all tissues in the body. HSP90 has been proposed as a target to increase turnover of pathological proteins leading to neurodegeneration in Huntington's disease, Parkinson's disease and Alzheimer's disease. The mechanism of how HSP90 inhibition leads to clearance of misfolded proteins is not fully understood. It may involve direct effects of inhibiting ATPase function, indirect effects by inducing the heat-shock-response resulting in upregulation of other chaperone proteins like HSP70 or a combination of both. In the current work we established a methodology to investigate the relationship between HSP90 target occupancy and HSP70 induction in vivo. We also characterized the acute effect of two different HSP90 inhibitors in the rTg4510 transgenic mouse model of Alzheimer's disease which displays a tau-mediated synaptic dysfunction. We show that reversal of synaptic impairments in this model can be obtained with a compound which has a high HSP70 induction capacity. The current developed assay methodologies may thus be of significant use in the further elucidation of the mechanism involved in the in vivo effect of HSP90 inhibition in models of neurodegeneration. Further on, the ability of HSP90 inhibitors to normalize synaptic dysfunction in an in vivo disease model of Alzheimer's disease could have therapeutic relevance and further strengthens the usefulness of this animal model to establish pharmacodynamic effect of HSP90 inhibition.


Asunto(s)
Encéfalo/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Línea Celular , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Ratones Transgénicos , Proteínas tau/genética
2.
Pharmacol Res ; 64(3): 268-73, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21504793

RESUMEN

The stability and transcriptional activity of the hypoxia-inducible factors (HIFs) are regulated by oxygen-dependent hydroxylation that is catalyzed by three HIF prolyl 4-hydroxylases (HPHs). Use of HPH inhibition as a mean for HIF-upregulation has recently gained interest as a potential treatment paradigm against neurodegenerative diseases like ischemia and Parkinson's disease. In the present investigation we report the development of a new and robust assay to measure HPH activity. The assay is based on capture of hydroxylated peptide product by the von Hippel-Lindau protein which is directly measured in a scintillation proximity assay. In addition we describe the determination of HPH subtype potencies of HPH inhibitors which either directly or indirectly inhibit the HPH enzyme. The potencies of the HPH inhibitors displayed almost identical IC(50) values toward the HPH1 and HPH2 subtype while the potency against the HPH3 subtype was increased for several of the compounds. For the most potent compound, a hydroxyl thiazole derivative, the potency against HPH2 and HPH3 was 7nM and 0.49nM, respectively corresponding to a 14-fold difference. These results suggest that HPH subtype-selective compounds may be developed. In addition we determined the 2-oxoglutarate concentration in brain tissue and neuronal cell lines as 2-oxoglutarate is an important co-factor used by the HPH enzyme during the hydroxylation reaction. The high intracellular 2-oxoglutarate concentration provides an explanation for the diminished cellular HIF activating potency of a competitive HPH inhibitor compared to its orders of magnitude higher HPH inhibiting potency. The present reported data suggest that in the development of specific Hif prolyl hydroxylase inhibitors the high 2-oxoglutarate tissue level should be taken into account as this might affect the cellular potency. Thus to specifically inhibit the intracellular HPH enzymatic reaction a competitive inhibitor with a low Ki should be developed.


Asunto(s)
Dioxigenasas/metabolismo , Inhibidores Enzimáticos/farmacología , Ácidos Cetoglutáricos/metabolismo , Proteínas Nucleares/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Línea Celular , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/genética , Evaluación Preclínica de Medicamentos/métodos , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
J Neurochem ; 115(1): 209-19, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20649842

RESUMEN

Hypoxia-inducible factor (HIF) controls the expression of genes that adapts the cellular condition to accommodate oxidative stress. The potential beneficial effect of HIF up-regulation in ischemia has recently gained interest substantiated by the known HIF-regulation of erythropoietin and other hypoxia accommodating genes. So far the perspectives for HIF up-regulation has been focused on anemia and ischemia related diseases but little information is available about the relevance of HIF biology for neurodegenerative disease like Parkinson's disease. We therefore sought out to characterize the effect of HIF-up-regulation on survival and dopamine homeostasis in dopaminergic cells. We used a low molecular weight HIF prolyl hydroxylase (HPH) inhibitor and lentiviral based shRNA knockdown of HPH subtypes as molecular tools to increase HIF protein level and downstream HIF-regulated genes. We show that HIF induction results in protection against oxidative stress in cellular models based on PC12 cells and LUHMES cells. In addition, HPH inhibition elevates tyrosine hydroxylase expression and activity, which causes increased dopamine synthesis and release in both PC12 cells and a primary rat ventral mesencephalic cell culture. All together these findings suggest that prolyl hydroxylases may represent novel targets for therapeutic intervention in disorders characterized by dopamine homeostasis dysregulation like Parkinson's disease.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Dioxigenasas/antagonistas & inhibidores , Dopamina/metabolismo , Dopamina/fisiología , Neuronas/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , Animales , Western Blotting , Línea Celular , Dopamina/biosíntesis , Inhibidores Enzimáticos/farmacología , Genes Reporteros , Humanos , Luciferasas/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Neuronas/efectos de los fármacos , Células PC12 , Procolágeno-Prolina Dioxigenasa/metabolismo , Interferencia de ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirosina 3-Monooxigenasa/metabolismo
4.
Pharmaceutics ; 12(11)2020 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-33105895

RESUMEN

Drug distribution in the brain is generally associated with an affinity for fatty brain tissues and therefore known to be species- and concentration-independent. We report here the effect of target affinity on brain tissue binding for 10 small molecules designed to inhibit brain heat shock protein 90 (HSP90), a widespread protein whose expression is 1-2% of total cytosolic proteins in eucaryotes. Our results show that increasing the test item concentrations from 0.3 to 100 µM increased the unbound fraction 32-fold for the most potent molecules, with no change for the inactive one (1.1 fold change). Saturation of HSP90 led to normal concentration-independent brain tissue binding. In vivo pharmacokinetics performed in rats showed that the overall volume of distribution of compounds is correlated with their affinity for HSP90. The in vitro binding and in vivo pharmacokinetics (PK) performed in rats showed that small molecule HSP90 inhibitors followed the principle of target-mediated drug disposition. We demonstrate that assessing unbound fractions in brain homogenate was subject to HSP90 target interference; this may challenge the process of linking systemic-free drug concentrations to central nervous system unbound concentrations necessary to establish the proper pharmacokinetics/pharmacodynamics (PK/PD) relation needed for human dose prediction.

5.
J Neurosci Res ; 87(7): 1686-94, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19156859

RESUMEN

The transcription factor hypoxia-inducible factor (HIF) is essential for the activation of several genes that promote the survival of cells exposed to oxidative stress. Expression of tyrosine hydroxylase (TH), which is the rate-limiting enzyme in the dopamine (DA) synthesis, is one of the genes that are positively regulated by HIF. Accordingly, HIF induction results in elevated DA release in various cell lines in vitro. HIF prolyl hydroxylase (HPH) is critically involved in the negative regulation of HIF levels. We investigated the in vivo effects of the HPH inhibitor FG0041 on brain DA function in rats by microdialysis in freely moving rats, locomotor activity, and Western blot analysis. Administration of FG0041 (10 mg/kg i.p.), as an acute (single injection), or as subchronic (once daily for 6 days) treatment and cobalt chloride (CoCl2) (60 mg/kg s.c.) potentiated potassium (K+) induced increases in extracellular levels of DA levels in the rat striatum. The increase in extracellular DA of freely moving rats was sought in relationship to locomotor activity in rats. A significant increase in locomotor activity was observed in FG0041-treated rats compared with vehicle on a cocaine challenge. In support of these findings, protein levels of TH in the rat brain stem were increased after treatment with FG0041. These data indicate that FG0041 augments DA function in the rat brain. Inhibition of HPH enhances DA function by increasing DA release, which has implications for the use of HIF induction in the treatment of neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Inhibidores Enzimáticos/farmacología , Fenantrolinas/farmacología , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Cobalto/farmacología , Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Espacio Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Potasio/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Tirosina 3-Monooxigenasa/metabolismo
6.
Neurochem Int ; 112: 5-17, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29111377

RESUMEN

Deubiquitinating enzymes (DUBs) play important roles in a variety of cellular processes, including regulation of protein homeostasis. The DUB ataxin-3 is an enzyme implicated in protein quality control mechanisms. In the neurodegenerative disease spinocerebellar ataxia type 3 (SCA3), ataxin-3 contains an expanded polyglutamine (polyQ) stretch that leads to aggregation of the protein and neuronal dysfunction. Increasing the understanding of ataxin-3 protein interaction partners could help to elucidate disease mechanisms. Hence, we analyzed the repertoire of proteins interacting with normal and polyQ expanded ataxin-3 by mass spectrometry. This showed that both normal and polyQ expanded ataxin-3 interacted with components of the protein quality control system and mitochondria. Five proteins showed increased interaction with polyQ expanded ataxin-3 relative to normal and three of these were mitochondrial proteins. The analyses underline the role of ataxin-3 in ubiquitin biology and point towards a role in mitochondrial biology.


Asunto(s)
Ataxina-3/análisis , Ataxina-3/metabolismo , Mitocondrias/metabolismo , Péptidos/análisis , Péptidos/metabolismo , Mapas de Interacción de Proteínas/fisiología , Animales , Ataxina-3/genética , Células HEK293 , Humanos , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/metabolismo , Espectrometría de Masas/métodos , Ratones , Ratones Transgénicos , Mitocondrias/genética , Péptidos/genética
7.
Neurochem Int ; 105: 42-50, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28065793

RESUMEN

Ubiquitination and phosphorylation of proteins represent post translational modifications (PTMs) capable of regulating a variety of cellular processes. In the neurodegenerative disorder spinocerebellar ataxia type 3 (SCA3), the disease causing protein ataxin-3 carries an expanded polyglutamine (polyQ) stretch causing it to aggregate in nuclear inclusions. These inclusions are decorated with ubiquitin suggestive of a malfunction in the clearance of the mutant protein. Differences in ubiquitin chain topology between normal and polyQ expanded ataxin-3 could be involved in the differential clearance of the two proteins and play a role in SCA3 pathogenesis. Likewise, changes in phosphorylation patterns between the two variants could contribute to pathogenic processes involved in SCA3. We therefore determined the ubiquitination and phosphorylation patterns, together with the ubiquitin-linkage types associated with normal and polyQ expanded ataxin-3 by mass spectrometry (MS). This analysis revealed a similar ubiquitin linkage pattern on normal and expanded ataxin-3. However, the distribution of ubiquitinated lysine residues was altered in polyQ expanded ataxin-3, with increased ubiquitination at the new identified ubiquitination site lysine-8. MS analysis of phosphorylation also revealed novel phosphorylation sites in ataxin-3, and an increase in phosphorylation of expanded ataxin-3 at several positions. The study suggests that differences in clearance of normal and expanded ataxin-3 are not attributed to differences in ubiquitin-linkage pattern. However, the observed differences between the normal and polyQ expanded ataxin-3 with respect to the degree of ubiquitination and phosphorylation on specific sites may have an impact on ataxin-3 function and SCA3 pathogenesis.


Asunto(s)
Ataxina-3/genética , Ataxina-3/metabolismo , Péptidos/genética , Péptidos/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ubiquitinación/fisiología , Secuencia de Aminoácidos , Sitios de Unión/fisiología , Células HEK293 , Humanos , Fosforilación/fisiología
8.
Sci Rep ; 7(1): 10300, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860483

RESUMEN

Genetic variation in the leucine-rich repeat kinase 2 (LRRK2) gene is associated with risk of familial and sporadic Parkinson's disease (PD). To support clinical development of LRRK2 inhibitors as disease-modifying treatment in PD biomarkers for kinase activity, target engagement and kinase inhibition are prerequisite tools. In a combined proteomics and phosphoproteomics study on human peripheral mononuclear blood cells (PBMCs) treated with the LRRK2 inhibitor Lu AF58786 a number of putative biomarkers were identified. Among the phospho-site hits were known LRRK2 sites as well as two phospho-sites on human Rab10 and Rab12. LRRK2 dependent phosphorylation of human Rab10 and human Rab12 at positions Thr73 and Ser106, respectively, was confirmed in HEK293 and, more importantly, Rab10-pThr73 inhibition was validated in immune stimulated human PBMCs using two distinct LRRK2 inhibitors. In addition, in non-stimulated human PBMCs acute inhibition of LRRK2 with two distinct LRRK2 inhibitor compounds reduced Rab10-Thr73 phosphorylation in a concentration-dependent manner with apparent IC50's equivalent to IC50's on LRRK2-pSer935. The identification of Rab10 phosphorylated at Thr73 as a LRRK2 inhibition marker in human PBMCs strongly support inclusion of assays quantifying Rab10-pThr73 levels in upcoming clinical trials evaluating LRRK2 kinase inhibition as a disease-modifying treatment principle in PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Leucocitos Mononucleares/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Biología Computacional/métodos , Relación Dosis-Respuesta a Droga , Humanos , Inmunomodulación/efectos de los fármacos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Leucocitos Mononucleares/inmunología , Fosfoproteínas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteoma , Proteómica , Reproducibilidad de los Resultados
9.
J Med Chem ; 60(21): 8945-8962, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29023112

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2), such as G2019S, are associated with an increased risk of developing Parkinson's disease. Surrogates for the LRRK2 kinase domain based on checkpoint kinase 1 (CHK1) mutants were designed, expressed in insect cells infected with baculovirus, purified, and crystallized. X-ray structures of the surrogates complexed with known LRRK2 inhibitors rationalized compound potency and selectivity. The CHK1 10-point mutant was preferred, following assessment of surrogate binding affinity with LRRK2 inhibitors. Fragment hit-derived arylpyrrolo[2,3-b]pyridine LRRK2 inhibitors underwent structure-guided optimization using this crystallographic surrogate. LRRK2-pSer935 HEK293 IC50 data for 22 were consistent with binding to Ala2016 in LRRK2 (equivalent to Ala147 in CHK1 10-point mutant structure). Compound 22 was shown to be potent, moderately selective, orally available, and brain-penetrant in wild-type mice, and confirmation of target engagement was demonstrated, with LRRK2-pSer935 IC50 values for 22 in mouse brain and kidney being 1.3 and 5 nM, respectively.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Encéfalo/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cristalografía/métodos , Células HEK293 , Humanos , Riñón/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Mutación , Enfermedad de Parkinson/genética , Unión Proteica , Dominios Proteicos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética
10.
Neurotherapeutics ; 11(2): 433-49, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24477711

RESUMEN

Machado-Joseph disease (MJD) or spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disease currently with no treatment. We describe a novel mouse model of MJD which expresses mutant human ataxin-3 at near endogenous levels and manifests MJD-like motor symptoms that appear gradually and progress over time. CMVMJD135 mice show ataxin-3 intranuclear inclusions in the CNS and neurodegenerative changes in key disease regions, such as the pontine and dentate nuclei. Hsp90 inhibition has shown promising outcomes in some neurodegenerative diseases, but nothing is known about its effects in MJD. Chronic treatment of CMVMJD mice with Hsp90 inhibitor 17-DMAG resulted in a delay in the progression of their motor coordination deficits and, at 22 and 24 weeks of age, was able to rescue the uncoordination phenotype to wild-type levels; in parallel, a reduction in neuropathology was observed in treated animals. We observed limited induction of heat-shock proteins with treatment, but found evidence that 17-DMAG may be acting through autophagy, as LC3-II (both at mRNA and protein levels) and beclin-1 were induced in the brain of treated animals. This resulted in decreased levels of the mutant ataxin-3 and reduced intranuclear aggregation of this protein. Our data validate this novel mouse model as a relevant tool for the study of MJD pathogenesis and for pre-clinical studies, and show that Hsp90 inhibition is a promising therapeutic strategy for MJD.


Asunto(s)
Benzoquinonas/uso terapéutico , Lactamas Macrocíclicas/uso terapéutico , Enfermedad de Machado-Joseph/tratamiento farmacológico , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Equilibrio Postural/efectos de los fármacos , Proteínas Represoras/genética , Animales , Ataxina-3 , Autofagia/efectos de los fármacos , Benzoquinonas/administración & dosificación , Benzoquinonas/farmacología , Modelos Animales de Enfermedad , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Lactamas Macrocíclicas/administración & dosificación , Lactamas Macrocíclicas/farmacología , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo
11.
ISRN Neurosci ; 2013: 598587, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25006572

RESUMEN

The hypoxia inducible factor 1 (HIF-1) is a central transcription factor involved in the cellular and molecular adaptation to hypoxia and low glucose supply. The level of HIF-1 is to a large degree regulated by the HIF prolyl hydroxylase enzymes (HPHs) belonging to the Fe(II) and 2-oxoglutarate-dependent dioxygenase superfamily. In the present study, we compared competitive and noncompetitive HPH-inhibitor compounds in two different cell types (SH-SY5Y and PC12). Although the competitive HPH-inhibitor compounds were found to be pharmacologically more potent than the non-competitive compounds at inhibiting HPH2 and HPH1, this was not translated into the cellular effects of the compounds, where the non-competitive inhibitors were actually more potent than the competitive in stabilizing and translocatingHIF1 α to the nucleus (quantified with Cellomics ArrayScan technology). This could be explained by the high cellular concentrations of the cofactor 2-oxoglutarate (2-OG) as the competitive inhibitors act by binding to the 2-OG site of the HPH enzymes. Both competitive and non-competitive HPH inhibitors protected the cells against 6-OHDA induced oxidative stress. In addition, the protective effect of a specific HPH inhibitor was partially preserved when the cells were serum starved and exposed to 2-deoxyglucose, an inhibitor of glycolysis, indicating that other processes than restoring energy supply could be important for the HIF-mediated cytoprotection.

12.
ACS Med Chem Lett ; 2(4): 303-6, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24900311

RESUMEN

GPR139 is an orphan G-protein coupled receptor (GPCR) which is primarily expressed in the central nervous system (CNS). In order to explore the biological function of this receptor, selective tool compounds are required. A screening campaign identified compound 1a as a high potency GPR139 agonist with an EC50 = 39 nM in a calcium mobilization assay in CHO-K1 cells stably expressing the GPR139 receptor. In the absence of a known endogenous ligand, the maximum effect was set as 100% for 1a. Screening against 90 diverse targets revealed no cross-reactivity issues. Assessment of the pharmacokinetic properties showed limited utility as in vivo tool compound in rat with a poor whole brain exposure of 61 ng/g and a brain/plasma (b/p) ratio of 0.03. Attempts to identify a more suitable analogue identified the des-nitrogen analogue 1s with a reduced polar surface area of 76.7 Å(2) and an improved b/p ratio of 2.8. The whole brain exposure remained low at 95 ng/g due to a low plasma exposure.

13.
Behav Brain Res ; 208(2): 444-9, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20035793

RESUMEN

Systemic injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mice is one of the primary models used to evaluate neuroprotective and symptomatic treatment strategies for Parkinson's disease. Many behavioral methods for evaluation of MPTP toxicity have been described, but they often involve challenging scenarios that require handling and transfer of animals to novel environments and in some cases prior animal training. These factors can profoundly influence animal behavior and potentially influence experimental outcome. Presented here is a new nest building scoring paradigm based on the animals' normal home cage behavior that is a simple, non-invasive, and reproducible measure for estimating neurological dysfunction in MPTP intoxicated mice. Nest building behavior requires orofacial and forelimb movement and has been shown to be dopamine-dependent making it a possible method for assessing parkinsonian-like symptoms. Significant deficits in nest building scores after 2x20 and 2x25 mg/kg MPTP coincided with a 90% reduction in striatal dopamine. Nest building deficits could be detected for more than a week after intoxication. However, after 28 days the change in behavior was no longer detected, which may reflect the plasticity of the tyrosine hydroxylase positive neurons in the dorsolateral part of striatum.


Asunto(s)
Intoxicación por MPTP/fisiopatología , Comportamiento de Nidificación/fisiología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Conducta Animal , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neurotoxinas/toxicidad , Recuperación de la Función/fisiología , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
14.
Obesity (Silver Spring) ; 18(7): 1289-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19851307

RESUMEN

Using restriction fragment differential display (RFDD) technology, we have identified the imprinted gene neuronatin (Nnat) as a hypothalamic target under the influence of leptin. Nnat mRNA expression is decreased in several key appetite regulatory hypothalamic nuclei in rodents with impaired leptin signaling and during fasting conditions. Furthermore, peripheral administration of leptin to ob/ob mice normalizes hypothalamic Nnat expression. Comparative immunohistochemical analysis of human and rat hypothalami demonstrates that NNAT protein is present in anatomically equivalent nuclei, suggesting human physiological relevance of the gene product(s). A putative role of Nnat in human energy homeostasis is further emphasized by a consistent association between single nucleotide polymorphisms (SNPs) in the human Nnat gene and severe childhood and adult obesity.


Asunto(s)
Impresión Genómica/fisiología , Leptina/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Obesidad/genética , Obesidad/fisiopatología , Tejido Adiposo/fisiología , Animales , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Genotipo , Homeostasis/genética , Hipotálamo/fisiología , Leptina/genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Páncreas/fisiología , Hipófisis/fisiología , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología
15.
Microbiology (Reading) ; 146 ( Pt 6): 1381-1389, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10846216

RESUMEN

Pediocin PA-1, which is a bacteriocin produced by lactic acid bacteria, has potential as a biopreservative of food. However, such use may lead to the development of resistance in the target organism. Gene expression in two independent pediocin-resistant mutants of Listeria monocytogenes 412 was compared to the original isolate by restriction fragment differential display PCR (RFDD-PCR). This method amplifies cDNA restriction fragments under stringent PCR conditions, enabled by the use of specific primers complementary to ligated adaptor sequences. RFDD-PCR was very well suited for analysis of listerial gene expression, giving reproducible PCR product profiles. Three gene fragments having increased expression in both resistant mutants were identified. All three had homology to components of beta-glucoside-specific phosphoenolpyruvate-dependent phosphotransferase systems (PTS), one fragment having homology to enzyme II permeases, and the two others to phospho-beta-glucosidases. Overexpression of the putative PTS system was consistently observed in 10 additional pediocin-resistant mutants, isolated at different pH, salt content and temperature. The results suggest that RFDD-PCR is a strong approach for the analysis of prokaryotic gene expression and that the putative beta-glucoside-specific PTS system is involved in mediating pediocin resistance.


Asunto(s)
Bacteriocinas/farmacología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cartilla de ADN/genética , Farmacorresistencia Microbiana/genética , Conservación de Alimentos , Expresión Génica , Genes Bacterianos , Humanos , Listeria monocytogenes/patogenicidad , Datos de Secuencia Molecular , Mutación , Pediocinas , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Reacción en Cadena de la Polimerasa/métodos , Homología de Secuencia de Aminoácido
16.
Exp Cell Res ; 286(1): 102-14, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12729799

RESUMEN

Cardiac hypertrophy is induced by a number of stimuli and can lead to cardiomyopathy and heart failure. Cardiomyocyte hypertrophy is characterized by increased cell size and altered gene expression. By differential-display polymerase chain reaction and Western blotting we found that the transcriptional coactivator MBF1 was upregulated during hypertrophy in cardiomyocyte cultures. Furthermore, MBF1 protein level increased in two animal models of hypertrophy, angiotensin II treatment and aortic banding. MBF1 antisense oligodeoxynuclotides blocked phenylephrine-induced hypertrophy, suggesting MBF1 plays a key role in hypertrophic growth. In contrast, overexpression of MBF1 potentiated the hormone-induced response of the atrial natriuretic peptide promoter. MBF1 overexpressed by transient transfection cooperated with the transcription factor c-Jun in activation of transcription but not with GATA4. MBF1 and c-Jun induced the activity of a transiently transfected atrial natriuretic peptide promoter, whereas neither MBF1 nor c-Jun could induce the promoter alone. Moreover, MBF1 bound to c-Jun in vitro. These data suggest that MBF1 is a transcriptional coactivator of c-Jun regulating hypertrophic gene expression. Inhibitor studies suggested that MBF1 activates the atrial natriuretic peptide promoter independently of the calcineurin and CaMK signaling pathways. Our results indicate that MBF1 participates in hormone-induced cardiomyocyte hypertrophy and activates hypertrophic gene expression as a coactivator of c-Jun.


Asunto(s)
Proteínas de Unión a Calmodulina , Cardiomegalia/metabolismo , Miocitos Cardíacos/fisiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transactivadores/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Calcineurina/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Cardiotónicos/farmacología , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Ratones , Datos de Secuencia Molecular , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Oligonucleótidos Antisentido/metabolismo , Fenilefrina/farmacología , Proteínas Proto-Oncogénicas c-jun/genética , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA