Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Alzheimers Dement ; 19(8): 3379-3388, 2023 08.
Article in English | MEDLINE | ID: mdl-36795603

ABSTRACT

INTRODUCTION: We assessed the use of cerebrospinal fluid (CSF) biomarkers as an alternative to positron emission tomography (PET) for brain amyloid beta (Aß) pathology confirmation in the EMERGE and ENGAGE clinical trials. METHODS: EMERGE and ENGAGE were randomized, placebo-controlled, Phase 3 trials of aducanumab in participants with early Alzheimer's disease. Concordance between CSF biomarkers (Aß42, Aß40, phosphorylated tau 181, and total tau) and amyloid PET status (visual read) at screening was examined. RESULTS: Robust concordance between CSF biomarkers and amyloid PET visual status was observed (for Aß42/Aß40, AUC: 0.90; 95% CI: 0.83-0.97; p < 0.0001), confirming CSF biomarkers as a reliable alternative to amyloid PET in these studies. Compared with single CSF biomarkers, CSF biomarker ratios showed better agreement with amyloid PET visual reads, demonstrating high diagnostic accuracy. DISCUSSION: These analyses add to the growing body of evidence supporting CSF biomarkers as reliable alternatives to amyloid PET imaging for brain Aß pathology confirmation. HIGHLIGHTS: CSF biomarkers and amyloid PET concordance were assessed in Ph3 aducanumab trials. Robust concordance between CSF biomarkers and amyloid PET was observed. CSF biomarker ratios increased diagnostic accuracy over single CSF biomarkers. CSF Aß42/Aß40 demonstrated high concordance with amyloid PET. Results support CSF biomarker testing as a reliable alternative to amyloid PET.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Alzheimer Disease/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Positron-Emission Tomography , Amyloid , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid
2.
Alzheimers Dement ; 17(9): 1575-1582, 2021 09.
Article in English | MEDLINE | ID: mdl-33788410

ABSTRACT

The core cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers amyloid beta (Aß42 and Aß40), total tau, and phosphorylated tau, have been extensively clinically validated, with very high diagnostic performance for AD, including the early phases of the disease. However, between-center differences in pre-analytical procedures may contribute to variability in measurements across laboratories. To resolve this issue, a workgroup was led by the Alzheimer's Association with experts from both academia and industry. The aim of the group was to develop a simplified and standardized pre-analytical protocol for CSF collection and handling before analysis for routine clinical use, and ultimately to ensure high diagnostic performance and minimize patient misclassification rates. Widespread application of the protocol would help minimize variability in measurements, which would facilitate the implementation of unified cut-off levels across laboratories, and foster the use of CSF biomarkers in AD diagnostics for the benefit of the patients.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Clinical Laboratory Techniques , Guidelines as Topic/standards , Internationality , Specimen Handling , tau Proteins/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Clinical Laboratory Techniques/instrumentation , Clinical Laboratory Techniques/standards , Humans , Phosphorylation , Specimen Handling/instrumentation , Specimen Handling/standards
3.
Arthritis Rheumatol ; 69(3): 643-654, 2017 03.
Article in English | MEDLINE | ID: mdl-27723281

ABSTRACT

OBJECTIVE: To characterize baseline gene expression and pharmacodynamically induced changes in whole blood gene expression in 1,760 systemic lupus erythematosus (SLE) patients from 2 phase III, 52-week, randomized, placebo-controlled, double-blind studies in which patients were treated with the BAFF-blocking IgG4 monoclonal antibody tabalumab. METHODS: Patient samples were obtained from SLE patients from the ILLUMINATE-1 and ILLUMINATE-2 studies, and control samples were obtained from healthy donors. Blood was collected in Tempus tubes at baseline, week 16, and week 52. RNA was analyzed using Affymetrix Human Transcriptome Array 2.0 and NanoString. RESULTS: At baseline, expression of the interferon (IFN) response gene was elevated in patients compared with controls, with 75% of patients being positive for this IFN response gene signature. There was, however, substantial heterogeneity of IFN response gene expression and complex relationships among gene networks. The IFN response gene signature was a predictor of time to disease flare, independent of anti-double-stranded DNA (anti-dsDNA) antibody and C3 and C4 levels, and overall disease activity. Pharmacodynamically induced changes in gene expression following tabalumab treatment were extensive, occurring predominantly in B cell-related and immunoglobulin genes, and were consistent with other pharmacodynamic changes including anti-dsDNA antibody, C3, and immunoglobulin levels. CONCLUSION: SLE patients demonstrated increased expression of an IFN response gene signature (75% of patients had an elevated IFN response gene signature) at baseline in ILLUMINATE-1 and ILLUMINATE-2. Substantial heterogeneity of gene expression was detected among individual patients and in gene networks. The IFN response gene signature was an independent risk factor for future disease flares. Pharmacodynamic changes in gene expression were consistent with the mechanism of BAFF blockade by tabalumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B-Cell Activating Factor/antagonists & inhibitors , Gene Expression/genetics , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Gene Expression/drug effects , Humans , Male , Middle Aged , Young Adult
4.
J Pers Med ; 6(1)2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26861400

ABSTRACT

This study aims to confirm the initial pharmacogenetic finding observed within the clinical proof-of-concept trial of an enhanced response to treatment with pomaglumetad methionil (LY2140023 monohydrate) in Caucasian schizophrenia patients homozygous for T/T at single nucleotide polymorphism rs7330461 in the serotonin (5-hydroxytryptamine) 2A receptor gene compared to A/A homozygous patients. The effect of the rs7330461 genotype on the response to pomaglumetad methionil treatment was assessed in three additional clinical trials and in an integrated analysis. Overall, this study includes data from 1115 Caucasian patients for whom genotyping information for rs7330461 was available, consisting of 513 A/A homozygous, 466 A/T heterozygous and 136 T/T homozygous patients. Caucasian T/T homozygous patients showed significantly (p ≤ 0.05) greater improvement in Positive and Negative Syndrome Scale (PANSS) total scores during treatment with pomaglumetad methionil 40 mg twice daily compared to A/A homozygous patients. Additionally, T/T homozygous patients receiving pomaglumetad methionil had significantly (p ≤ 0.05) greater improvements in PANSS total scores compared to placebo and similar improvements as T/T homozygous patients receiving standard-of-care (SOC) treatment. The findings reported here in conjunction with prior reports show that in Caucasian patients with schizophrenia, the T/T genotype at rs7330461 is consistently associated with an increased treatment response to pomaglumetad methionil compared to the A/A genotype.

5.
BMC Genet ; 17: 16, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26738766

ABSTRACT

BACKGROUND: The serotonin 2A receptor is widely implicated in genetic association studies and remains an important drug target for psychiatric, neurological, and cardiovascular conditions. RNA sequencing redefined the architecture of the serotonin 2A receptor gene (HTR2A), revealing novel mRNA transcript isoforms utilizing unannotated untranslated regions of the gene. Expression of these untranslated regions is modulated by common single nucleotide polymorphisms (SNPs), namely rs6311. Previous studies did not fully capture the complexity of the sense- and antisense-encoded transcripts with respect to novel exons in the HTR2A gene locus. Here, we comprehensively catalogued exons and RNA isoforms for both HTR2A and HTR2A-AS1 using RNA-Seq from human prefrontal cortex and multiple mouse tissues. We subsequently tested associations between expression of newfound gene features and common SNPs in humans. RESULTS: We find that the human HTR2A gene spans ~66 kilobases and consists of 7, rather than 4 exons. Furthermore, the revised human HTR2A-AS1 gene spans ~474 kilobases and consists of 18, rather than 3 exons. Three HTR2A exons directly overlap with HTR2A-AS1 exons, suggesting potential for complementary nucleotide interactions. The repertoire of possible mouse Htr2a splice isoforms is remarkably similar to humans and we also find evidence for overlapping sense-antisense transcripts in the same relative positions as the human transcripts. rs6311 and SNPs in high linkage disequilibrium are associated with HTR2A-AS1 expression, in addition to previously described associations with expression of the extended 5' untranslated region of HTR2A. CONCLUSIONS: Our proposed HTR2A and HTR2A-AS1 gene structures dramatically differ from current annotations, now including overlapping exons on the sense and anti-sense strands. We also find orthologous transcript isoforms expressed in mice, providing opportunities to elucidate the biological roles of the human isoforms using a model system. Associations between rs6311 and expression of HTR2A and HTR2A-AS1 suggest this polymorphism is capable of modulating the expression of the sense or antisense transcripts. Still unclear is whether these SNPs act directly on the expression of the sense or antisense transcripts and whether overlapping exons are capable of interacting through complimentary base-pairing. Additional studies are necessary to determine the extent and nature of interactions between the SNPs and the transcripts prior to interpreting these findings in the context of phenotypes associated with HTR2A.


Subject(s)
DNA, Antisense , Exons , Receptor, Serotonin, 5-HT2A/genetics , Alternative Splicing , Animals , Humans , Mice , Polymorphism, Single Nucleotide , Prefrontal Cortex/metabolism , RNA Splice Sites , Schizophrenia/genetics , Sequence Alignment , Transcription, Genetic
6.
Mol Neuropsychiatry ; 1(2): 82-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26405684

ABSTRACT

The NMDA receptor antagonist phencyclidine (PCP) creates schizophrenia-like symptoms in normal controls. The effect of PCP on non-human primate brain gene expression was examined and compared to changes induced by olanzapine treatment. Experimental studies of PCP and antipsychotic drugs have direct relevance to understanding the patho-physiology and treatment of schizophrenia. Genome-wide changes in prefrontal cortex gene expression revealed alterations of 146 transcripts in the PCP treatment group compared to vehicle controls. Dysregulated genes were enriched in identified classes implicated in neurological and genetic disorders, including schizophrenia genes from the Psychiatric Genomics Consortium 108 loci as well as cell death in PCP-treated primates. Canonical pathway analysis revealed a significant overrepresentation of several groups including synaptic long-term potentiation and calcium signaling. Olanzapine coadministered with PCP normalized 34% of the 146 PCP-induced probe set expression changes, and a network of 17 olanzapine-normalized genes was identified enriched in schizophrenia candidate genes containing RGS4, SYN1 and AKT as nodes. The results of this study support the use of PCP administration in non-human primates as a glutamatergic model of schizophrenia and suggest that a large number of PCP-induced expression differences can be reversed by olanzapine. The results of this study may be informative for identification of potential candidates for pharmacogenetics and biomarker research related to the treatment of schizophrenia.

7.
J Clin Pharmacol ; 55(10): 1167-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25919121

ABSTRACT

Atomoxetine, which is indicated for treatment of attention-deficit hyperactivity disorder (ADHD), is predominantly metabolized by genetically polymorphic cytochrome P450 2D6 (CYP2D6). Based on identified CYP2D6 genotypes, individuals can be categorized into 4 phenotypic metabolizer groups as ultrarapid, extensive, intermediate, and poor. Previous studies have focused on observed differences between poor and extensive metabolizers, but it is not well understood whether the safety profile of intermediate metabolizers differs from that of ultrarapid and extensive metabolizers. This study compared safety and tolerability among the different CYP2D6 metabolizer groups in the 12-week open-label phase of an atomoxetine study in adult patients with ADHD. Genotyping identified 1039 patients as extensive/ultrarapid metabolizers, 780 patients as intermediate metabolizers, and 117 patients as poor metabolizers. Common (≥5% frequency) treatment-emergent adverse events did not significantly differ between extensive/ultrarapid and intermediate metabolizers (odds ratios were <2.0 or >0.5). Poor metabolizers had higher frequencies of dry mouth, erectile dysfunction, hyperhidrosis, insomnia, and urinary retention compared with the other metabolizer groups. There were no significant differences between extensive/ultrarapid and intermediate metabolizers in changes from baseline in vital signs. These results suggest that data from CYP2D6 intermediate and extensive/ultrarapid metabolizers can be combined when considering safety analyses related to atomoxetine.


Subject(s)
Adrenergic Uptake Inhibitors/adverse effects , Atomoxetine Hydrochloride/adverse effects , Attention Deficit Disorder with Hyperactivity/drug therapy , Cytochrome P-450 CYP2D6/genetics , Adrenergic Uptake Inhibitors/pharmacokinetics , Adrenergic Uptake Inhibitors/therapeutic use , Adult , Atomoxetine Hydrochloride/pharmacokinetics , Atomoxetine Hydrochloride/therapeutic use , Attention Deficit Disorder with Hyperactivity/genetics , Cytochrome P-450 CYP2D6/metabolism , Female , Genotype , Humans , Male , Phenotype , Young Adult
8.
BMC Psychiatry ; 14: 351, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25539791

ABSTRACT

BACKGROUND: Pomaglumetad methionil (LY2140023 monohydrate) is a potent and highly selective agonist for the metabotropic glutamate mGluR2 and mGluR3 receptors. We present results of a pivotal clinical study H8Y-MC-HBBM assessing the efficacy of LY2140023 in improving symptoms as a monotherapy in patients with an acute exacerbation of schizophrenia. METHODS: Enrolled adult patients (ages 18-65) with schizophrenia who had experienced an exacerbation of symptoms within 2 weeks prior to study entry. Patients (N = 1013) were randomized 2:2:2:1 to treatment with placebo, LY40 mg twice daily (BID), LY80 mg BID, or risperidone (RIS) 2 mg BID for 6 weeks after a one-week blinded placebo lead-in. The primary outcome assessed change from baseline in the Positive and Negative Syndrome Scale (PANSS) total score in an overall schizophrenia population and a predefined subpopulation which excluded non-Hispanic white patients with the A/A genotype at the HTR2A SNP rs7330461. RESULTS: Neither LY2140023 dose showed significant improvement compared to placebo on PANSS total in either population (1-sided p-value [significance level], overall: LY40, p = .154 [0.01]; LY80, p = .698 [0.01], subpopulation: LY40, p = .033 [0.0025]; LY80, p = .659 [0.0025], MMRM analysis). RIS statistically separated from placebo in both populations (p < .001 [0.05]). There were no statistically significant differences in the incidence of serious adverse events, and no seizures on LY2140023. CONCLUSION: LY2140023 treatment did not demonstrate efficacy in populations studied. Overall, LY2140023 treatment was generally well tolerated with no new adverse safety findings compared to previous trials. Further understanding of the role of glutamate as a therapeutic target in schizophrenia is needed. CLINICAL TRIALS REGISTRATION: A Phase 2, Multicenter, Double-Blind, Placebo-Controlled Comparator Study of 2 Doses of LY2140023 Versus Placebo in Patients With DSM-IV-TR SchizophreniaClinicalTrials.gov identifier: NCT01086748.


Subject(s)
Amino Acids/administration & dosage , Antipsychotic Agents/administration & dosage , Schizophrenia/drug therapy , Adult , Aged , Dopamine Antagonists/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Psychiatric Status Rating Scales , Receptors, Metabotropic Glutamate/agonists , Risperidone/administration & dosage , Treatment Outcome , Young Adult
9.
Biol Psychiatry ; 73(6): 546-54, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23158458

ABSTRACT

BACKGROUND: The 5-hydroxytryptamine 2A receptor, encoded by HTR2A, is a major postsynaptic target for serotonin in the human brain and a therapeutic drug target. Despite hundreds of genetic associations investigating HTR2A polymorphisms in neuropsychiatric disorders and therapies, the role of genetic HTR2A variability in health and disease remains uncertain. METHODS: To discover and characterize regulatory HTR2A variants, we sequenced whole transcriptomes from 10 human brain regions with massively parallel RNA sequencing and measured allelic expression of multiple HTR2A messenger (m)RNA transcript variants. Following discovery of functional variants, we further characterized their impact on genetic expression in vitro. RESULTS: Three polymorphisms modulate the use of novel alternative exons and untranslated regions (UTRs), changing expression of RNA and protein. The frequent promoter variant rs6311, widely implicated in human neuropsychiatric disorders, decreases usage of an upstream transcription start site encoding a longer 5'UTR with greater translation efficiency. rs76665058, located in an extended 3'UTR and unique to individuals of African descent, modulates allelic HTR2A mRNA expression. The third single nucleotide polymorphism, unannotated and present in only a single subject, directs alternative splicing of exon 2. Targeted analysis of HTR2A in the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study reveals associations between functional variants and depression severity or citalopram response. CONCLUSIONS: Regulatory polymorphisms modulate HTR2A mRNA expression in an isoform-specific manner, directing the usage of novel untranslated regions and alternative exons. These results provide a foundation for delineating the role of HTR2A and serotonin signaling in central nervous system disorders.


Subject(s)
Cerebral Cortex/metabolism , Gene Expression Regulation/genetics , Genetic Variation/genetics , Promoter Regions, Genetic/genetics , Receptor, Serotonin, 5-HT2A/biosynthesis , Alleles , Clinical Trials as Topic , Depression/drug therapy , Depression/genetics , Exons/genetics , Humans , Methylation , Polymorphism, Single Nucleotide/genetics , Protein Isoforms/biosynthesis , Selective Serotonin Reuptake Inhibitors/therapeutic use , Transcriptome/genetics , Untranslated Regions/genetics
10.
J Neurosci Res ; 90(3): 588-96, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22038504

ABSTRACT

Orexinergic neurons are discretely localized within the lateral hypothalamus and have widespread projections to the whole brain. Here, the role of orexin/hypocretin-2 receptors (OX2) in modulating extracellular concentrations of neurotransmitters was evaluated in the hypothalamus and the prefrontal cortex (PFC) of OX2 knockout (KO) mice by using a microdialysis technique. In the hypothalamus, basal concentrations of norephinephrine (NE), acetylcholine (ACh), and histamine (Hist) were significantly higher in KO mice, whereas KCl perfusion (147 mM) resulted in significantly lesser increases in NE, ACh, and Hist release in KO compared with wild-type (WT) mice. No differences in basal concentrations or evoked release of serotonin (5-HT) or dopamine (DA) were found in the hypothalamus between genotypes. In the PFC, no differences in the basal concentrations of the studied neurotransmitters were found between genotypes. After KCl perfusion, significantly higher increases in NE, 5-HT, and DA release were found in KO compared with WT mice. No differences in the evoked release of ACh and Hist in the PFC were found between genotypes. The present results demonstrate that genetic deletion of OX2 receptors differentially modulates extracellular concentrations of distinct neurotransmitters in the somatodendritic region vs. a nerve terminal region of the orexinergic neurons. In the hypothalamus, an inhibitory role of the OX2 receptors in modulating basal concentrations of NE, ACh, and Hist was revealed, which probably accounts for the reduced responsiveness to KCl as well. In the PFC, the evoked release of the monoamines NE, 5-HT, and DA seems to be controlled negatively by OX2 receptors.


Subject(s)
Hypothalamus/physiology , Prefrontal Cortex/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Synaptic Transmission/physiology , Acetylcholine/metabolism , Animals , Dopamine/metabolism , Histamine/metabolism , Mice , Mice, Knockout , Microdialysis , Norepinephrine/metabolism , Orexin Receptors , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Serotonin/metabolism , Synaptic Transmission/genetics
11.
J Neurochem ; 113(1): 175-87, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20070867

ABSTRACT

The molecular mechanism of action of antipsychotic drugs is not well understood. Their complex receptor affinity profiles indicate that their action could extend beyond dopamine receptor blockade. Single gene expression studies and high-throughput gene profiling have shown the induction of genes from several molecular classes and functional categories. Using a focused microarray approach, we investigated gene regulation in rat striatum, frontal cortex, and hippocampus after chronic administration of haloperidol or olanzapine. Regulated genes were validated by in situ hybridization, real-time PCR, and immunohistochemistry. Only limited overlap was observed in genes regulated by haloperidol and olanzapine. Both drugs elicited maximal gene regulation in the striatum and least in the hippocampus. Striatal gene induction by haloperidol was predominantly in neurotransmitter signaling, G-protein coupled receptors, and transcription factors. Olanzapine prominently induced retinoic acid and trophic factor signaling genes in the frontal cortex. The data also revealed the induction of several genes that could be targeted in future drug development efforts. The study uncovered the induction of several novel genes, including somatostatin receptors and metabotropic glutamate receptors. The results demonstrating the regulation of multiple receptors and transcription factors suggests that both typical and atypical antipsychotics could possess a complex molecular mechanism of action.


Subject(s)
Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Brain/drug effects , Gene Expression Regulation/drug effects , Haloperidol/analogs & derivatives , Animals , Brain/metabolism , Gene Expression Profiling/methods , Haloperidol/pharmacology , Male , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Olanzapine , Oligonucleotide Array Sequence Analysis/methods , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
J Pharmacol Exp Ther ; 315(3): 1265-77, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16141369

ABSTRACT

FMPD [6-fluoro-10-[3-(2-methoxyethyl)-4-methyl-piperazin-1-yl]-2-methyl-4H-3-thia-4,9-diaza-benzo[f]azulene] is a potential novel antipsychotic with high affinity for dopamine D2 (Ki= 6.3 nM), 5-HT(2A) (Ki= 7.3 nM), and 5-HT6 (Ki= 8.0 nM) human recombinant receptors and lower affinity for histamine H1 (Ki= 30 nM) and 5-HT2C (Ki= 102 nM) human recombinant receptors than olanzapine. Oral administration of FMPD increased rat nucleus accumbens 3,4-dihyroxyphenylacetic acid concentrations (ED200 = 6 mg/kg), blocked 5-HT2A agonist-induced increases in rat serum corticosterone levels (ED50= 1.8 mg/kg), and inhibited the ex vivo binding of [125I]SB-258585 [4-iodo-N-[4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyl]-benzenesulfonamide] to striatal 5-HT6 receptors (ED50= 10 mg/kg) but failed to inhibit ex vivo binding of [3H]pyrilamine to hypothalamic histamine H1 receptors at doses of up to 30 mg/kg. In electrophysiology studies, acute administration of FMPD selectively elevated the number of spontaneously active A10 (versus A9) dopamine neurons and chronic administration selectively decreased the number of spontaneously active A10 (versus A9) dopamine neurons. FMPD did not produce catalepsy at doses lower than 25 mg/kg p.o. In Fos-induction studies, FMPD had an atypical antipsychotic profile in the striatum and nucleus accumbens and increased Fos expression in orexin-containing neurons of the hypothalamus. FMPD produced only a transient elevation of prolactin levels. These data indicate that FMPD is an orally available potent antagonist of dopamine D2, 5-HT2A, and 5-HT6 receptors and a weak antagonist of H1 and 5-HT2C receptors. FMPD has the potential to have efficacy in treating schizophrenia and bipolar mania with a low risk of treatment-emergent extrapyramidal symptoms, prolactin elevation, and weight gain. Clinical trials are needed to test these hypotheses.


Subject(s)
Antipsychotic Agents/pharmacology , Piperazines/pharmacology , Receptors, Histamine H1/metabolism , 3,4-Dihydroxyphenylacetic Acid/analysis , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Antipsychotic Agents/metabolism , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Body Weight/drug effects , Catalepsy/chemically induced , Cocaine/pharmacology , Corticosterone/blood , Drug Evaluation, Preclinical , Electrochemistry , Electrophysiology , Fasting , Female , Immunohistochemistry , Male , Molecular Structure , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Olanzapine , Piperazines/chemistry , Piperazines/metabolism , Prolactin/blood , Quipazine/pharmacology , Radioligand Assay , Rats , Rats, Inbred F344 , Rats, Inbred Strains , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology , Thiophenes/therapeutic use , Time Factors
13.
J Neurosci Methods ; 141(1): 9-20, 2005 Jan 30.
Article in English | MEDLINE | ID: mdl-15585284

ABSTRACT

Immunohistochemical localization of c-Fos immunoreactivity has been used successfully for over a decade to visualize patterns of neuronal activity in the brain and spinal cord. These experiments are extremely useful in identifying physiological or pharmacological activation of specific populations of neurons. Unfortunately, conventional c-Fos immunohistochemical protocols are very time and resource intensive. We have adapted and optimized established c-Fos immunohistochemistry (IHC) methodologies for use with fresh frozen brain tissue mounted directly onto slides. The resultant rapid protocols, which we refer to as "Fast Fos", include applications for single- and double-labeling, utilizing either enzyme-substrate or fluorescent detection systems. These protocols provide increased assay throughput and reproducibility, which can be further enhanced by use of an automated slide stainer. Taken as a whole, the c-Fos IHC protocols described in this report provide a flexible system for the identification of neuronal activation that substantially reduces time and resource expenditure while increasing quality and reproducibility of data.


Subject(s)
Brain/metabolism , Immunohistochemistry/methods , Proto-Oncogene Proteins c-fos/analysis , Staining and Labeling/methods , Animals , Brain/cytology , Fluorescent Antibody Technique/methods , Frozen Sections/methods , Intracellular Signaling Peptides and Proteins/analysis , Male , Neuropeptides/analysis , Orexins , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Software Design , Time Factors , Tissue Fixation/methods
14.
Neurobiol Dis ; 16(1): 220-35, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15207279

ABSTRACT

Acute phencyclidine induces schizophrenia-like symptoms in healthy humans and psychotic episodes in schizophrenics. Although phencyclidine is known as a N-methyl d-aspartate receptor antagonist (NMDA-R), the molecular events underlying the behavioral symptoms remain largely unknown. Statistical analysis of oligonucleotide microarray data was used to identify phencyclidine-induced alterations in rat cortical gene expression. Acute phencyclidine produced a statistically significant change in 477 genes in rat prefrontal cortex (PFC), a brain area associated with cognitive dysfunction in schizophrenics. Real-time quantitative PCR (RTQ-PCR) confirmed a subset of these changes ranging from -59% to 255% (smallest confirmation: -19%). Subsequent time-course and dose-response studies using RTQ-PCR confirmed and extended the original microarray results. At the molecular level, genes altered by phencyclidine are related to diverse biological processes including stress, inflammatory response, growth and development, neural plasticity and signal transduction. Further analysis, aimed at assessing the relevance of our results to schizophrenia, revealed dysregulation of genes related to: (i) thalamocortical projections, (ii) neurotransmission and neuromodulation, (iii) thyroid hormone activity, (iv) oligodendrocyte linage, (v) brain lipid metabolism, (vi) sleep architecture and (viii) the velocardiofacial syndrome.


Subject(s)
Cerebral Cortex/drug effects , Gene Expression Regulation/drug effects , Oligonucleotide Array Sequence Analysis , Phencyclidine/administration & dosage , Schizophrenia/genetics , Animals , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Male , Oligonucleotide Array Sequence Analysis/methods , Phencyclidine/pharmacology , Rats , Rats, Sprague-Dawley , Schizophrenia/metabolism
15.
Am J Pharmacogenomics ; 4(2): 129-39, 2004.
Article in English | MEDLINE | ID: mdl-15059035

ABSTRACT

INTRODUCTION: The hybridization intensities derived from microarray experiments, for example Affymetrix's MAS5 signals, are very often transformed in one way or another before statistical models are fitted. The motivation for performing transformation is usually to satisfy the model assumptions such as normality and homogeneity in variance. Generally speaking, two types of strategies are often applied to microarray data depending on the analysis need: correlation analysis where all the gene intensities on the array are considered simultaneously, and gene-by-gene ANOVA where each gene is analyzed individually. AIM: We investigate the distributional properties of the Affymetrix GeneChip signal data under the two scenarios, focusing on the impact of analyzing the data at an inappropriate scale. METHODS: The Box-Cox type of transformation is first investigated for the strategy of pooling genes. The commonly used log-transformation is particularly applied for comparison purposes. For the scenario where analysis is on a gene-by-gene basis, the model assumptions such as normality are explored. The impact of using a wrong scale is illustrated by log-transformation and quartic-root transformation. RESULTS: When all the genes on the array are considered together, the dependent relationship between the expression and its variation level can be satisfactorily removed by Box-Cox transformation. When genes are analyzed individually, the distributional properties of the intensities are shown to be gene dependent. Derivation and simulation show that some loss of power is incurred when a wrong scale is used, but due to the robustness of the t-test, the loss is acceptable when the fold-change is not very large.


Subject(s)
Data Interpretation, Statistical , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Algorithms , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Databases, Genetic , Fluorescent Dyes , Gene Expression , Protein Folding , RNA/biosynthesis , RNA/genetics , Rats , Reference Values
16.
J Pharmacol Exp Ther ; 309(2): 825-32, 2004 May.
Article in English | MEDLINE | ID: mdl-14724223

ABSTRACT

This study characterizes the sumatriptan-sensitive [5-hydroxytryptamine (5-HT)(1B/1D)] receptor in rabbit saphenous vein and basilar artery. (S)-(-)-1-[2-[4-(4-Methoxy-phenyl)-piperazin-1-yl]-ethyl]isochroman-6-carboxylic acid methylamide (PNU-109291), a 5-HT(1D) subtype-selective agonist (human K(i) = 2.5 +/- 0.07 nM), did not contract either tissue, whereas o-methoxyphenylpiperazide derivative 4F (MPPA-4F), a 5-HT(1B) subtype-selective antagonist (human K(i) = 4.6 +/- 0.6 nM) potently inhibited sumatriptan-induced contraction in the saphenous vein and basilar artery. These results suggested that sumatriptan-induced contraction was mediated via the 5-HT(1B) receptor in these blood vessels. 5-HT(1B) receptor-mediated contraction was then compared in endothelium-intact and denuded vessels to evaluate the role of the endothelium in regulating sumatriptan-induced contractility in these tissues. The presence of an intact endothelium inhibited 5-HT(1B)-induced contraction in both tissues. Endothelial denudation or nitric-oxide synthase inhibition with N(omega) nitro-L-arginine methyl ester (L-NAME) (100 microM) increased the efficacy and potency of sumatriptan in the saphenous vein and basilar artery. Surprisingly, in endothelial-denuded vascular tissues, L-NAME (100 microM) also significantly increased the maximal 5-HT(1B) receptor-induced contraction in both tissues, with no effect on potency of sumatriptan. The effect of L-NAME after endothelial denudation may reflect the presence of a low density of residual endothelial cells as estimated by CD31 antibody staining combined with the modulating effect of nitric oxide released from nonendothelial cells in vascular tissue. Endothelial modulation was specific to 5-HT(1B) receptors because removal of the endothelium did not significantly alter contraction to norepinephrine, histamine, prostaglandin, or potassium chloride in the saphenous vein or basilar artery.


Subject(s)
Basilar Artery/physiology , Endothelium, Vascular/physiology , Receptor, Serotonin, 5-HT1B/physiology , Saphenous Vein/physiology , Vasoconstriction/physiology , Animals , Antigens, CD34/immunology , Basilar Artery/drug effects , Benzopyrans/pharmacology , Binding Sites , Endothelium, Vascular/drug effects , NG-Nitroarginine Methyl Ester , Piperazines/pharmacology , Rabbits , Saphenous Vein/drug effects , Serotonin 5-HT1 Receptor Agonists , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
17.
Physiol Genomics ; 16(1): 1-7, 2003 Dec 16.
Article in English | MEDLINE | ID: mdl-14679298

ABSTRACT

In the postgenomic era, integrating data obtained from array technologies (e.g., oligonucleotide microarrays) with published information on eukaryotic genomes is beginning to yield biomarkers and therapeutic targets that are key for the diagnosis and treatment of disease. Nevertheless, identifying and validating these drug targets has not been a trivial task. Although a plethora of bioinformatics tools and databases are available, major bottlenecks for this approach reside in the interpretation of vast amounts of data, its integration into biologically representative models, and ultimately the identification of pathophysiologically and therapeutically useful information. In the field of neuroscience, accomplishing these goals has been particularly challenging because of the complex nature of nerve tissue, the relatively small adaptive nature of induced-gene expression changes, as well as the polygenic etiology of most neuropsychiatric diseases. This report combines published data sets from multiple transcript profiling studies that used GeneChip microarrays to illustrate a postanalysis approach for the interpretation of data from neuroscience microarray studies. By defining common gene expression patterns triggered by diverse events (administration of psychoactive drugs and trauma) in different nerve tissues (telencephalic brain areas and spinal cord), we broaden the conclusions derived from each of the original studies. In addition, the evaluation of the identified overlapping gene lists provides a foundation for generating hypotheses relating alterations in specific sets of genes to common physiological processes. Our approach demonstrates the significance of interpreting transcript profiling data within the context of common pathways and mechanisms rather than specific to a given tissue or stimulus. We also highlight the use of gene expression patterns in predictive biology (e.g., in toxicogenomics) as well as the utility of combining data derived from multiple microarray studies that examine diverse biological events for a broader interpretation of data from a particular microarray study.


Subject(s)
Gene Expression Profiling , Nervous System/metabolism , Animals , Gene Expression Regulation/drug effects , Hallucinogens/pharmacology , Hippocampus/injuries , Hippocampus/metabolism , Lysergic Acid Diethylamide/pharmacology , Nervous System/drug effects , Oligonucleotide Array Sequence Analysis , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spinal Cord Injuries/genetics , Telencephalon/drug effects , Telencephalon/metabolism
18.
Neuropsychopharmacology ; 28(1): 45-52, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12496939

ABSTRACT

Recent studies have demonstrated that the hallucinogen 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) enhances glutamatergic transmission in the prefrontal cortex. This increase can be suppressed by metabotropic glutamate2/3 (mGlu2/3) receptor activation. In addition to enhancing glutamatergic transmission, DOI increases cortical c-fos expression. We tested if a reduction in glutamate release produced by mGlu2/3 receptor activation attenuates DOI-induced c-fos expression in the cortex. Similar to previous studies, DOI produced a robust increase in c-fos mRNA throughout the cortex, including the prefrontal, frontoparietal, and somatosensory regions. Pretreatment with the mGlu2/3 agonist LY379268 attenuated the DOI-induced increase in the prefrontal cortex. This suppression was blocked by the mGlu2/3 antagonist LY341495. In contrast, the DOI-induced increase in c-fos mRNA in the frontoparietal and somatosensory cortex was unaffected by the mGlu2/3 agents. These findings suggest that Group II metabotropic glutamate receptor agonists are capable of modulating postsynaptic function preferentially in the limbic cortex under conditions of enhanced glutamate release.


Subject(s)
Amphetamines/pharmacology , Cerebral Cortex/metabolism , Excitatory Postsynaptic Potentials/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Metabotropic Glutamate/metabolism , Serotonin Receptor Agonists/pharmacology , Amino Acids/pharmacology , Animals , Autoradiography , Bridged Bicyclo Compounds/pharmacology , Cerebral Cortex/drug effects , DNA/biosynthesis , DNA/genetics , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , GABA Agonists/pharmacology , Image Interpretation, Computer-Assisted , In Situ Hybridization , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/drug effects , Xanthenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...