Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Chem Phys ; 160(20)2024 May 28.
Article in English | MEDLINE | ID: mdl-38775240

ABSTRACT

The development and benchmarking of computational chemistry methods rely on comparison with benchmark data. More and larger benchmark datasets are becoming available, and working efficiently with them is a necessity. The Cuby framework provides rich functionality for working with datasets, comes with many ready-to-use predefined benchmark sets, and interfaces with a wide range of computational chemistry software packages. Here, we review the tools Cuby provides for working with datasets and provide examples of more advanced workflows, such as handling large numbers of computations on high performance computing resources and reusing previously computed data. Cuby has also been extended recently to include two important benchmark databases, NCIAtlas and GMTKN55.

2.
Phys Chem Chem Phys ; 25(17): 12479-12489, 2023 May 03.
Article in English | MEDLINE | ID: mdl-37097130

ABSTRACT

As the field of nanoelectronics based on biomolecules such as peptides and proteins rapidly grows, there is a need for robust computational methods able to reliably predict charge transfer properties at bio/metallic interfaces. Traditionally, hybrid quantum-mechanical/molecular-mechanical techniques are employed for systems where the electron hopping transfer mechanism is applicable to determine physical parameters controlling the thermodynamics and kinetics of charge transfer processes. However, these approaches are limited by a relatively high computational cost when extensive sampling of a configurational space is required, like in the case of soft biomatter. For these applications, semi-empirical approaches such as the perturbed matrix method (PMM) have been developed and successfully used to study charge-transfer processes in biomolecules. Here, we explore the performance of PMM on prototypical redox-active protein azurin in various environments, from solution to vacuum interfaces with gold surfaces and protein junction. We systematically benchmarked the robustness and convergence of the method with respect to the quantum-centre size, size of the Hamiltonian, number of samples, and level of theory. We show that PMM can adequately capture all the trends associated with the structural and electronic changes related to azurin oxidation at bio/metallic interfaces.


Subject(s)
Azurin , Azurin/chemistry , Electron Transport , Oxidation-Reduction , Proteins , Peptides/chemistry
3.
J Chem Phys ; 156(17): 175101, 2022 May 07.
Article in English | MEDLINE | ID: mdl-35525644

ABSTRACT

Metalloproteins, known to efficiently transfer electronic charge in biological systems, recently found their utilization in nanobiotechnological devices where the protein is placed into direct contact with metal surfaces. The feasibility of oxidation/reduction of the protein redox sites is affected by the reorganization free energies, one of the key parameters determining the transfer rates. While their values have been measured and computed for proteins in their native environments, i.e., in aqueous solution, the reorganization free energies of dry proteins or proteins adsorbed to metal surfaces remain unknown. Here, we investigate the redox properties of blue copper protein azurin, a prototypical redox-active metalloprotein previously probed by various experimental techniques both in solution and on metal/vacuum interfaces. We used a hybrid quantum mechanical/molecular mechanical computational technique based on density functional theory to explore protein dynamics, flexibility, and corresponding reorganization free energies in aqueous solution, vacuum, and on vacuum gold interfaces. Surprisingly, the reorganization free energy only slightly decreases when azurin is dried because the loss of the hydration shell leads to larger flexibility of the protein near its redox site. At the vacuum gold surfaces, the energetics of the structure relaxation depends on the adsorption geometry; however, significant reduction of the reorganization free energy was not observed. These findings have important consequences for the charge transport mechanism in vacuum devices, showing that the free energy barriers for protein oxidation remain significant even under ultra-high vacuum conditions.


Subject(s)
Azurin , Metalloproteins , Azurin/chemistry , Azurin/metabolism , Copper/chemistry , Copper/metabolism , Electron Transport , Gold , Metalloproteins/chemistry , Oxidation-Reduction , Vacuum , Water/chemistry
4.
PLoS One ; 15(12): e0243052, 2020.
Article in English | MEDLINE | ID: mdl-33370315

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by severe disruption of cognitive and motor functions, including changes in posture and gait. A number of HD mouse models have been engineered that display behavioral and neuropathological features of the disease, but gait alterations in these models are poorly characterized. Sensitive high-throughput tests of fine motor function and gait in mice might be informative in evaluating disease-modifying interventions. Here, we describe a hypothesis-free workflow that determines progressively changing locomotor patterns across 79 parameters in the R6/2 and Q175 mouse models of HD. R6/2 mice (120 CAG repeats) showed motor disturbances as early as at 4 weeks of age. Similar disturbances were observed in homozygous and heterozygous Q175 KI mice at 3 and 6 months of age, respectively. Interestingly, only the R6/2 mice developed forelimb ataxia. The principal components of the behavioral phenotypes produced two phenotypic scores of progressive postural instability based on kinematic parameters and trajectory waveform data, which were shared by both HD models. This approach adds to the available HD mouse model research toolbox and has a potential to facilitate the development of therapeutics for HD and other debilitating movement disorders with high unmet medical need.


Subject(s)
Gait Analysis/methods , Huntingtin Protein/genetics , Huntington Disease/physiopathology , Animals , Biomechanical Phenomena , Disease Models, Animal , Female , Huntington Disease/genetics , Male , Mice , Mice, Transgenic , Motor Activity , Mutation , Posture
5.
EJNMMI Radiopharm Chem ; 4(1): 20, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31659519

ABSTRACT

PURPOSE: Dopamine receptors are involved in pathophysiology of neuropsychiatric diseases, including Huntington's disease (HD). PET imaging of dopamine D2 receptors (D2R) in HD patients has demonstrated 40% decrease in D2R binding in striatum, and D2R could be a reliable quantitative target to monitor disease progression. A D2/3R antagonist, [18F] fallypride, is a high-affinity radioligand that has been clinically used to study receptor density and occupancy in neuropsychiatric disorders. Here we report an improved synthesis method for [18F]fallypride. In addition, high molar activity of the ligand has allowed us to apply PET imaging to characterize D2/D3 receptor density in striatum of the recently developed zQ175DN knock-in (KI) mouse model of HD. METHODS: We longitudinally characterized in vivo [18F] fallypride -PET imaging of D2/D3 receptor densities in striatum of 9 and 12 month old wild type (WT) and heterozygous (HET) zQ175DN KI mouse. Furthermore, we verified the D2/D3 receptor density in striatum with [3H] fallypride autoradiography at 12 months of age. RESULTS: We implemented an improved synthesis method for [18F] fallypride to yield high molar activity (MA, 298-360 GBq/µmol) and good reproducibility. In the HET zQ175DN KI mice, we observed a significant longitudinal decrease in binding potential (BPND) (30.2%, p < 0.001, 9 months of age and 51.6%, p < 0.001, 12 months of age) compared to WT littermates. No mass effect was observed when the MA of [18F] fallypride was > 100 GBq/µmol at the time of injection. Furthermore, the decrease of D2/D3 receptor density in striatum in HET zQ175DN KI was consistent using [3H] fallypride autoradiography. CONCLUSIONS: We observed a significant decrease in D2/D3R receptor densities in the striatum of HET zQ175DN KI mice compared to WT mice at 9 and 12 months of age. These results are in line with clinical findings in HD patients, suggesting [18F] fallypride PET imaging has potential as a quantitative translational approach to monitor disease progression in preclinical studies.

6.
Beilstein J Org Chem ; 15: 1758-1768, 2019.
Article in English | MEDLINE | ID: mdl-31435447

ABSTRACT

We report two novel functional dyes based on a boron-dipyrromethene (BODIPY) core displaying a panchromatic absorption with an extension to the near-infrared (NIR) range. An innovative synthetic approach for preparing the 2,3,5,6-tetramethyl-BODIPY unit is disclosed, and a versatile way to further functionalize this unit has been developed. The optoelectronic properties of the two dyes were computed by density functional theory modelling (DFT) and characterized through UV-vis spectroscopy and cyclic voltammetry (CV) measurements. Finally, we report preliminary results obtained using these functional dyes as photosensitizers in dye-sensitized solar cells (DSSCs).

7.
Eur J Pharmacol ; 803: 11-23, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28336400

ABSTRACT

Histaminergic H3 inverse agonists, by stimulating central histamine release, represent attractive drug candidates to treat cognitive disorders. The present studies aimed to describe the mechanistic profile of S 38093 a novel H3 receptors inverse agonist. S 38093 displays a moderate affinity for rat, mouse and human H3 receptors (Ki=8.8, 1.44 and 1.2µM, respectively) with no affinity for other histaminergic receptors. In cellular models, the compound was able to antagonize mice H3 receptors (KB=0.65µM) and to suppress cAMP decrease induced by an H3 agonist via human H3 receptors (KB=0.11µM). The antagonism properties of the compound were confirmed by electrophysiological studies on rat hippocampal slices (from 0.1µM). In cells expressing a high H3 density, S 38093 behaved as a moderate inverse agonist at rat and human H3 receptors (EC50=9 and 1.7µM, respectively). S 38093 was rapidly absorbed in mouse and rat (Tmax=0.25-0.5h), slowly in monkey (2h), with a bioavailability ranging from 20% to 60% and t1/2 ranging from 1.5 to 7.4h. The compound was widely distributed with a moderate volume of distribution and low protein binding. The brain distribution of S 38093 was rapid and high. In mice, S 38093 significantly increased ex vivo N-tele-Methylhistamine cerebral levels from 3mg/kg p.o. and antagonized R-α-Methylhistamine-induced dipsogenia from 10mg/kg i.p. Taken together, these data suggest that S 38093, a novel H3 inverse agonist, is a good candidate for further in vivo evaluations, in particular in animal models of cognition.


Subject(s)
Azabicyclo Compounds/pharmacology , Benzamides/pharmacology , Drug Inverse Agonism , Histamine Agonists/pharmacokinetics , Histamine H3 Antagonists/pharmacokinetics , Receptors, Histamine H3/metabolism , Animals , Arachidonic Acid/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Histamine/metabolism , Histamine Agonists/metabolism , Histamine Agonists/pharmacology , Histamine H3 Antagonists/metabolism , Histamine H3 Antagonists/pharmacology , Humans , Male , Mice , Rats
8.
PLoS One ; 12(2): e0171127, 2017.
Article in English | MEDLINE | ID: mdl-28182673

ABSTRACT

The aim of these studies was to demonstrate the therapeutic capacity of an antisense oligonucleotide with the sequence (CUG)7 targeting the expanded CAG repeat in huntingtin (HTT) mRNA in vivo in the R6/2 N-terminal fragment and Q175 knock-in Huntington's disease (HD) mouse models. In a first study, R6/2 mice received six weekly intracerebroventricular infusions with a low and high dose of (CUG)7 and were sacrificed 2 weeks later. A 15-60% reduction of both soluble and aggregated mutant HTT protein was observed in striatum, hippocampus and cortex of (CUG)7-treated mice. This correction at the molecular level resulted in an improvement of performance in multiple motor tasks, increased whole brain and cortical volume, reduced levels of the gliosis marker myo-inositol, increased levels of the neuronal integrity marker N-aceyl aspartate and increased mRNA levels of the striatal marker Darpp-32. These neuroanatomical and neurochemical changes, together with the improved motor performance, suggest that treatment with (CUG)7 ameliorates basal ganglia dysfunction. The HTT-lowering was confirmed by an independent study in Q175 mice using a similar (CUG)7 AON dosing regimen, further demonstrating a lasting reduction of mutant HTT protein in striatum, hippocampus and cortex for up to 18 weeks post last infusion along with an increase in motor activity. Based on these encouraging results, (CUG)7 may thus offer an interesting alternative HTT-lowering strategy for HD.


Subject(s)
Genetic Therapy , Huntingtin Protein/genetics , Huntington Disease/therapy , RNA, Antisense/genetics , Trinucleotide Repeat Expansion , Animals , Brain/metabolism , Brain/pathology , Female , Gliosis , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Motor Activity
9.
Exp Neurol ; 282: 99-118, 2016 08.
Article in English | MEDLINE | ID: mdl-27163548

ABSTRACT

Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntington's disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.


Subject(s)
Electrophysiological Phenomena/drug effects , Enzyme Inhibitors/therapeutic use , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Pyrimidines/therapeutic use , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiological Phenomena/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hippocampus/drug effects , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , In Vitro Techniques , Kynurenic Acid/metabolism , Kynurenine 3-Monooxygenase/metabolism , Male , Mice , Mice, Transgenic , Microdialysis , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Quinolinic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Trinucleotide Repeats/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
10.
PLoS One ; 9(12): e113317, 2014.
Article in English | MEDLINE | ID: mdl-25469887

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder that primarily affects the medium-size GABAergic neurons of striatum. The R6/2 mouse line is one of the most widely used animal models of HD. Previously the hallmarks of HD-related pathology have been detected in photoreceptors and interneurons of R6/2 mouse retina. Here we aimed to explore the survival of retinal ganglion cells (RGCs) and functional integrity of distinct retinal cell populations in R6/2 mice. The pattern electroretinography (PERG) signal was lost at the age of 8 weeks in R6/2 mice in contrast to the situation in wild-type (WT) littermates. This defect may be attributable to a major reduction in photopic ERG responses in R6/2 mice which was more evident in b- than a-wave amplitudes. At the age of 4 weeks R6/2 mice had predominantly the soluble form of mutant huntingtin protein (mHtt) in the RGC layer cells, whereas the aggregated form of mHtt was found in the majority of those cells from the 12-week-old R6/2 mice and onwards. Retinal astrocytes did not contain mHtt deposits. The total numbers of RGC layer cells, retinal astrocytes as well as optic nerve axons did not differ between 18-week-old R6/2 mice and their WT controls. Our data indicate that mHtt deposition does not cause RGC degeneration or retinal astrocyte loss in R6/2 mice even at a late stage of HD-related pathology. However, due to functional deficits in the rod- and cone-pathways, the R6/2 mice suffer progressive deficits in visual capabilities starting as early as 4 weeks; at 8 weeks there is severe impairment. This should be taken into account in any behavioral testing conducted in R6/2 mice.


Subject(s)
Huntington Disease/physiopathology , Retina/physiopathology , Retinal Ganglion Cells/metabolism , Animals , Astrocytes/metabolism , Disease Models, Animal , Electroretinography , Female , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retina/cytology , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Ganglion Cells/pathology
11.
PLoS Curr ; 62014 Feb 13.
Article in English | MEDLINE | ID: mdl-24558637

ABSTRACT

Huntington's disease is a neurodegenerative disorder caused by mutations in the CAG tract of huntingtin. Several studies in HD cellular and rodent systems have identified disturbances in cyclic nucleotide signaling, which might be relevant to pathogenesis and therapeutic intervention. To investigate whether selective phosphodiesterase (PDE) inhibitors can improve some aspects of disease pathogenesis in HD models, we have systematically evaluated the effects of a variety of cAMP and cGMP selective PDE inhibitors in various HD models. Here we present the lack of effect in a variety of endpoints of the PDE subtype selective inhibitor SCH-51866, a PDE1/5 inhibitor, in the R6/2 mouse model of HD, after chronic oral dosing.

12.
PLoS One ; 7(12): e50717, 2012.
Article in English | MEDLINE | ID: mdl-23284644

ABSTRACT

Huntington's disease (HD) is an autosomal neurodegenerative disorder, characterized by severe behavioral, cognitive, and motor deficits. Since the discovery of the huntingtin gene (HTT) mutation that causes the disease, several mouse lines have been developed using different gene constructs of Htt. Recently, a new model, the zQ175 knock-in (KI) mouse, was developed (see description by Menalled et al, [1]) in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. Here we confirm the behavioral phenotypes reported by Menalled et al [1], and extend the characterization to include brain volumetry, striatal metabolite concentration, and early neurophysiological changes. The overall reproducibility of the behavioral phenotype across the two independent laboratories demonstrates the utility of this new model. Further, important features reminiscent of human HD pathology are observed in zQ175 mice: compared to wild-type neurons, electrophysiological recordings from acute brain slices reveal that medium spiny neurons from zQ175 mice display a progressive hyperexcitability; glutamatergic transmission in the striatum is severely attenuated; decreased striatal and cortical volumes from 3 and 4 months of age in homo- and heterozygous mice, respectively, with whole brain volumes only decreased in homozygotes. MR spectroscopy reveals decreased concentrations of N-acetylaspartate and increased concentrations of glutamine, taurine and creatine + phosphocreatine in the striatum of 12-month old homozygotes, the latter also measured in 12-month-old heterozygotes. Motor, behavioral, and cognitive deficits in homozygotes occur concurrently with the structural and metabolic changes observed. In sum, the zQ175 KI model has robust behavioral, electrophysiological, and histopathological features that may be valuable in both furthering our understanding of HD-like pathophyisology and the evaluation of potential therapeutic strategies to slow the progression of disease.


Subject(s)
Behavior, Animal , Brain/pathology , Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease/pathology , Huntington Disease/physiopathology , Neurophysiology , Animals , Body Weight , Brain/metabolism , Brain/physiopathology , Cell Count , Disease Progression , Endpoint Determination , Female , Glutamic Acid/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Neostriatum/pathology , Nerve Tissue Proteins/genetics , Neurons/pathology , Organ Size , Repetitive Sequences, Nucleic Acid , Swimming , Synaptic Transmission
13.
Twin Res Hum Genet ; 8(3): 185-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15989746

ABSTRACT

Information about linkage disequilibrium (LD) is important in understanding the genome structure and has its applications in association studies. Here we present the first genome-wide LD study based on a founder population (East Finland). The LD data consist of 118 unrelated individuals and around 480,000 SNP pairs genotyped with the Affymetrix 100K genotyping assay. Using the minor allele frequency (MAF) limit of .05, the squared correlation coefficient between two loci (r(2)) was .48, .37, .28, and .20 for distances of 5, 10, 20, and 40 kb respectively. MAF had a significant effect on the mean r(2) so that the extent of useful LD (r(2) > .3) varied from 17 kb to 80 kb depending on the limit set for the MAF. For D' the effect of MAF was smaller but reflected the possible age of the mutation: SNPs with high MAF had lower D' than those with low MAF. The X chromosome showed higher D' values than autosomes and the extent of useful LD (r(2) > .3) was twice as long on the X chromosome than on the autosomes. Based on the results, LD varies across the genome and is correlated to local recombination rate between and within chromosomes. However, the recombination rate does not explain all the variation found in LD. We also report a number of long chromosomal regions where exceptionally high or low LD were detected.


Subject(s)
Genetics, Population , Genome, Human , Linkage Disequilibrium/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Case-Control Studies , Chi-Square Distribution , Chromosomes, Human, X , Female , Finland , Founder Effect , Gene Frequency , Genotype , Humans , Male
14.
J Neurochem ; 83(5): 1043-53, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12437575

ABSTRACT

Antipsychotic drug treatment is known to modulate gene expression in experimental animals. In this study, candidate target genes for antipsychotic drug action were searched using microarrays after acute clozapine treatment (1, 6 and 24 h) in the rat prefrontal cortex. Microarray data clustering with a self-organizing map algorithm revealed differential expression of genes involved in presynaptic function following acute clozapine treatment. The differential expression of 35 genes most profoundly regulated in expression arrays was further examined using in situ hybridization following acute clozapine, and chronic clozapine and haloperidol treatments. Acute administration of clozapine regulated the expression of chromogranin A, synaptotagmin V and calcineurin A mRNAs in the cortex. Chronic clozapine treatment induced differential cortical expression of chromogranin A, son of sevenless (SoS) and Sec-1. Chronic treatment with haloperidol regulated the mRNA expression of inhibitor of DNA-binding 2 (ID-2) and Rab-12. Furthermore, the expression of visinin-like proteins-1, -2 and -3 was regulated by chronic drug treatments in various brain regions. Our data suggest that acute and chronic treatments with haloperidol and clozapine modulate the expression of genes involved in synaptic function and in regulation of intracellular Ca2+ in cortex.


Subject(s)
Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Gene Expression Regulation/drug effects , Repressor Proteins , Animals , Calcineurin/genetics , Calcineurin/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebral Cortex/chemistry , Chromogranin A , Chromogranins/genetics , Chromogranins/metabolism , Clozapine/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation/physiology , Haloperidol/pharmacology , In Situ Hybridization , Inhibitor of Differentiation Protein 2 , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Munc18 Proteins , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , Synaptotagmins , Transcription Factors/genetics , Transcription Factors/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
15.
Expert Opin Ther Targets ; 6(3): 363-74, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12223073

ABSTRACT

The rapidly accumulating amount of information concerning gene and protein expression patterns produced by functional genomics, proteomics and bioinformatics is presently providing new targets for drug development. Furthermore, the analysis of gene expression in cells and tissues affected by a disease may reveal the underlying metabolic pathways and cellular processes affected. Finally, changes in gene expression may be used in either diagnostics or the monitoring of drug responses. This review focuses on advances in the use of functional genomics in neurological and neuropsychiatric diseases and neuropsychopharmacology. Although the number of published studies in this field is still limited, it already appears that this strategy may become a fruitful means in the analysis of the aetiology of neuropsychiatric disorders and the search for novel neuropharmacological drugs.


Subject(s)
Central Nervous System Agents/pharmacology , Drug Design , Gene Expression Profiling/methods , Genomics , Mental Disorders/drug therapy , Nervous System Diseases/drug therapy , Neuropharmacology/methods , Central Nervous System Agents/therapeutic use , Epilepsy/drug therapy , Epilepsy/genetics , Gene Expression Regulation/drug effects , Humans , Illicit Drugs/adverse effects , Illicit Drugs/pharmacology , Mental Disorders/genetics , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nervous System Diseases/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Schizophrenia/drug therapy , Schizophrenia/genetics , Substance-Related Disorders/genetics
16.
Neuropsychopharmacology ; 27(2): 152-62, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12093589

ABSTRACT

We have characterized the effects of chronic clozapine and haloperidol treatments on the expression of fos (c-fos, fosB, fra-2) and jun (c-jun, junB, junD) family genes in the rat forebrain. The effects of chronic (17d) clozapine and haloperidol on mRNA expression were determined two hours, 24 hours, and six days after the last drug injection, and the DNA-binding activity of the activator protein-1 (AP-1) complex was studied after washout periods of 24 hours and six days. Chronic clozapine treatment with a 6 d washout period induced the expression of several fos and jun family genes in cortical regions, including the prefrontal cortex (PFC), and in the caudate putamen and nucleus accumbens. Moreover, the DNA-binding activity of the AP-1 complex was greatly increased in the anterior cingulate cortex-PFC in mobility shift assays already after 24 h, and remained increased after a 6d washout period. Chronic administration of haloperidol upregulated fos and jun family mRNA expression that was detectable 24 h and 6 d after cessation of the treatment mainly in the cortex. However, the DNA-binding activity of the AP-1 complex was not altered in the anterior cingulate cortex-PFC by chronic haloperidol administration at any of the time points studied. Thus, chronic treatments with clozapine and haloperidol induce a long-lasting enhancement of fos and jun family transcription factors that continues for several days after the cessation of the treatments in the cortex. These lasting effects might represent events that are potentially involved in the mechanisms of antipsychotic drug action.


Subject(s)
Antipsychotic Agents/pharmacology , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/drug effects , Schizophrenia/drug therapy , Transcription Factor AP-1/genetics , Animals , Clozapine/pharmacology , DNA-Binding Proteins/genetics , Drug Administration Schedule , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Haloperidol/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Schizophrenia/pathology , Schizophrenia/physiopathology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...