Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nature ; 629(8013): 945-950, 2024 May.
Article in English | MEDLINE | ID: mdl-38720069

ABSTRACT

Lipoprotein(a) (Lp(a)), an independent, causal cardiovascular risk factor, is a lipoprotein particle that is formed by the interaction of a low-density lipoprotein (LDL) particle and apolipoprotein(a) (apo(a))1,2. Apo(a) first binds to lysine residues of apolipoprotein B-100 (apoB-100) on LDL through the Kringle IV (KIV) 7 and 8 domains, before a disulfide bond forms between apo(a) and apoB-100 to create Lp(a) (refs. 3-7). Here we show that the first step of Lp(a) formation can be inhibited through small-molecule interactions with apo(a) KIV7-8. We identify compounds that bind to apo(a) KIV7-8, and, through chemical optimization and further application of multivalency, we create compounds with subnanomolar potency that inhibit the formation of Lp(a). Oral doses of prototype compounds and a potent, multivalent disruptor, LY3473329 (muvalaplin), reduced the levels of Lp(a) in transgenic mice and in cynomolgus monkeys. Although multivalent molecules bind to the Kringle domains of rat plasminogen and reduce plasmin activity, species-selective differences in plasminogen sequences suggest that inhibitor molecules will reduce the levels of Lp(a), but not those of plasminogen, in humans. These data support the clinical development of LY3473329-which is already in phase 2 studies-as a potent and specific orally administered agent for reducing the levels of Lp(a).


Subject(s)
Drug Discovery , Lipoprotein(a) , Macaca fascicularis , Animals , Female , Humans , Male , Mice , Administration, Oral , Kringles , Lipoprotein(a)/antagonists & inhibitors , Lipoprotein(a)/blood , Lipoprotein(a)/chemistry , Lipoprotein(a)/metabolism , Mice, Transgenic , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Plasminogen/chemistry , Plasminogen/metabolism , Species Specificity , Clinical Trials, Phase II as Topic , Apolipoproteins A/chemistry , Apolipoproteins A/metabolism
2.
Physiol Genomics ; 51(10): 506-515, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31530226

ABSTRACT

The genomic regulatory networks underlying the pathogenesis of non-ST-segment elevation acute coronary syndrome (NSTE-ACS) are incompletely understood. As intermediate traits, protein biomarkers report on underlying disease severity and prognosis in NSTE-ACS. We hypothesized that integration of dense microRNA (miRNA) profiling with biomarker measurements would highlight potential regulatory pathways that underlie the relationships between prognostic biomarkers, miRNAs, and cardiovascular phenotypes. We performed miRNA sequencing using whole blood from 186 patients from the TRILOGY-ACS trial. Seven circulating prognostic biomarkers were measured: NH2-terminal pro-B-type natriuretic peptide (NT-proBNP), high-sensitivity C-reactive protein, osteopontin (OPN), myeloperoxidase, growth differentiation factor 15, monocyte chemoattractant protein, and neopterin. We tested miRNAs for association with each biomarker with generalized linear models and controlled the false discovery rate at 0.05. Ten miRNAs, including known cardiac-related miRNAs 25-3p and 423-3p, were associated with NT-proBNP levels (min. P = 7.5 × 10-4) and 48 miRNAs, including cardiac-related miRNAs 378a-3p, 20b-5p and 320a, -b, and -d, were associated with OPN levels (min. P = 1.6 × 10-6). NT-proBNP and OPN were also associated with time to cardiovascular death, myocardial infarction (MI), or stroke in the sample. By integrating large-scale miRNA profiling with circulating biomarkers as intermediate traits, we identified associations of known cardiac-related and novel miRNAs with two prognostic biomarkers and identified potential genomic networks regulating these biomarkers. These results, highlighting plausible biological pathways connecting miRNAs with biomarkers and outcomes, may inform future studies seeking to delineate genomic pathways underlying NSTE-ACS outcomes.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/genetics , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Natriuretic Peptide, Brain/blood , Osteopontin/blood , Peptide Fragments/blood , Aged , Biomarkers/blood , Cohort Studies , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Phenotype , Prognosis , Risk Factors , Sequence Analysis, RNA
3.
Nephrol Dial Transplant ; 33(11): 1950-1959, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29481660

ABSTRACT

Background: Inflammation signaled by Janus kinases (JAKs) promotes progression of diabetic kidney disease (DKD). Baricitinib is an oral, reversible, selective inhibitor of JAK1 and JAK2. This study tested the efficacy of baricitinib versus placebo on albuminuria in adults with Type 2 diabetes at high risk for progressive DKD. Methods: In this Phase 2, double-blind, dose-ranging study, participants were randomized 1:1:1:1:1 to receive placebo or baricitinib (0.75 mg daily; 0.75 mg twice daily; 1.5 mg daily; or 4 mg daily), for 24 weeks followed by 4-8 weeks of washout. Results: Participants (N = 129) were 63±9.1 (mean±standard deviation) years of age, 27.1% (35/129) women and 11.6% (15/129) African-American race. Baseline hemoglobin A1c (HbA1c) was 7.3±1% and estimated glomerular filtration rate was 45.0±12.1 mL/min/1.73 m2 with first morning urine albumin-creatinine ratio (UACR) of 820 (407-1632) (median; interquartile range) mg/g. Baricitinib, 4 mg daily, decreased morning UACR by 41% at Week 24 compared with placebo (ratio to baseline 0.59, 95% confidence interval 0.38-0.93, P = 0.022). UACR was decreased at Weeks 12 and 24 and after 4-8 weeks of washout. Baricitinib 4 mg decreased inflammatory biomarkers over 24 weeks (urine C-X-C motif chemokine 10 and urine C-C motif ligand 2, plasma soluble tumor necrosis factor receptors 1 and 2, intercellular adhesion molecule 1 and serum amyloid A). The only adverse event rate that differed between groups was anemia at 32.0% (8/25) for baricitinib 4 mg daily versus 3.7% (1/27) for placebo. Conclusions: Baricitinib decreased albuminuria in participants with Type 2 diabetes and DKD. Further research is required to determine if baricitinib reduces DKD progression.


Subject(s)
Albuminuria/drug therapy , Azetidines/therapeutic use , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/complications , Gene Expression Regulation, Enzymologic/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Sulfonamides/therapeutic use , Albuminuria/etiology , Albuminuria/pathology , Double-Blind Method , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/therapeutic use , Purines , Pyrazoles
4.
J Biol Chem ; 288(9): 6386-96, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23322769

ABSTRACT

Control of plasma cholesterol levels is a major therapeutic strategy for management of coronary artery disease (CAD). Although reducing LDL cholesterol (LDL-c) levels decreases morbidity and mortality, this therapeutic intervention only translates into a 25-40% reduction in cardiovascular events. Epidemiological studies have shown that a high LDL-c level is not the only risk factor for CAD; low HDL cholesterol (HDL-c) is an independent risk factor for CAD. Apolipoprotein A-I (ApoA-I) is the major protein component of HDL-c that mediates reverse cholesterol transport from tissues to the liver for excretion. Therefore, increasing ApoA-I levels is an attractive strategy for HDL-c elevation. Using genome-wide siRNA screening, targets that regulate hepatocyte ApoA-I secretion were identified through transfection of 21,789 siRNAs into hepatocytes whereby cell supernatants were assayed for ApoA-I. Approximately 800 genes were identified and triaged using a convergence of information, including genetic associations with HDL-c levels, tissue-specific gene expression, druggability assessments, and pathway analysis. Fifty-nine genes were selected for reconfirmation; 40 genes were confirmed. Here we describe the siRNA screening strategy, assay implementation and validation, data triaging, and example genes of interest. The genes of interest include known and novel genes encoding secreted enzymes, proteases, G-protein-coupled receptors, metabolic enzymes, ion transporters, and proteins of unknown function. Repression of farnesyltransferase (FNTA) by siRNA and the enzyme inhibitor manumycin A caused elevation of ApoA-I secretion from hepatocytes and from transgenic mice expressing hApoA-I and cholesterol ester transfer protein transgenes. In total, this work underscores the power of functional genetic assessment to identify new therapeutic targets.


Subject(s)
Apolipoprotein A-I/metabolism , Hepatocytes/metabolism , Liver/metabolism , Animals , Apolipoprotein A-I/genetics , Cholesterol, HDL/genetics , Cholesterol, HDL/metabolism , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Genome-Wide Association Study , Hep G2 Cells , Humans , Liver/cytology , Mice , Mice, Transgenic , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , RNA, Small Interfering/genetics
5.
PLoS One ; 19(1): e0292091, 2024.
Article in English | MEDLINE | ID: mdl-38277356

ABSTRACT

Many of the pathological consequences of chronic kidney disease can be attributed to an elevation in serum phosphate levels. Current therapies focused on decreasing intestinal phosphate absorption to treat hyperphosphatemia are inadequate. The most effective therapeutic strategy may be to target multiple absorptive pathways. In this study, the ability of a novel inhibitor of the intestinal sodium hydrogen exchanger 3 (NHE3), LY3304000, which inhibits paracellular, diffusional uptake of phosphate, to work in combination with an inhibitor of the active transporter, sodium dependent phosphate cotransporter 2b (NPT2b), LY3358966, was explored. LY3304000 modestly inhibited the acute uptake of phosphate into plasma of rats, while surprisingly, it doubled the rate of phosphate uptake in mice, an animal model dominated by NPT2b mediated acute phosphate uptake. In rats, LY3004000 and LY3358966 work in concert to inhibit acute phosphate uptake. On top of LY3358966, LY3304000 further decreased the acute uptake of phosphate into plasma. Studies measuring the recovery of radiolabeled phosphate in the intestine demonstrated LY3304000 and LY3358966 synergistically inhibited the absorption of phosphate in rats. We hypothesize the synergism is because the NHE3 inhibitor, LY3304000, has two opposing effects on intestinal phosphate absorption in rats, first it decreases diffusion mediated paracellular phosphate absorption, while second, it simultaneously increases phosphate absorption through the NPT2b pathway. NHE3 inhibition decreases proton export from enterocytes and raises the cell surface pH. In vitro, NPT2b mediated phosphate transport is increased at higher pHs. The increased NPT2b mediated transport induced by NHE3 inhibition is masked in rats which have relatively low levels of NPT2b mediated phosphate transport, by the more robust inhibition of diffusion mediated phosphate absorption. Thus, the inhibition of NPT2b mediated phosphate transport in rats in the presence of NHE3 inhibition has an effect that exceeds its effect in the absence of NHE3 inhibition, leading to the observed synergism on phosphate absorption between NPT2b and NHE3 inhibition.


Subject(s)
Phosphates , Renal Insufficiency, Chronic , Rats , Mice , Animals , Phosphates/metabolism , Sodium-Hydrogen Exchanger 3 , Rodentia , Intestinal Absorption , Renal Insufficiency, Chronic/metabolism , Sodium-Hydrogen Exchangers/metabolism
6.
Pharmacol Res Perspect ; 10(2): e00938, 2022 04.
Article in English | MEDLINE | ID: mdl-35194979

ABSTRACT

An excess phosphate burden in renal disease has pathological consequences for bone, kidney, and heart. Therapies to decrease intestinal phosphate absorption have been used to address the problem, but with limited success. Here, we describe the in vivo effects of a novel potent inhibitor of the intestinal sodium-dependent phosphate cotransporter NPT2b, LY3358966. Following treatment with LY3358966, phosphate uptake into plasma 15 min following an oral dose of radiolabeled phosphate was decreased 74% and 22% in mice and rats, respectively, indicating NPT2b plays a much more dominant role in mice than rats. Following the treatment with LY3358966 and radiolabeled phosphate, mouse feces were collected for 48 h to determine the ability of LY3358966 to inhibit phosphate absorption. Compared to vehicle-treated animals, there was a significant increase in radiolabeled phosphate recovered in feces (8.6% of the dose, p < .0001). Similar studies performed in rats also increased phosphate recovered in feces (5.3% of the dose, p < .05). When used in combination with the phosphate binder sevelamer in rats, there was a further small, but not significant, increase in fecal phosphate. In conclusion, LY3358966 revealed a more prominent role for NPT2b on acute intestinal phosphate uptake into plasma in mice than rats. However, the modest effects on total intestinal phosphate absorption observed in mice and rats with LY3359866 when used alone or in combination with sevelamer highlights the challenge to identify new more effective therapeutic targets and/or drug combinations to treat the phosphate burden in patients with renal disease.


Subject(s)
Intestinal Absorption , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/antagonists & inhibitors , Animals , CHO Cells , Chelating Agents/administration & dosage , Chelating Agents/pharmacology , Cricetulus , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Sevelamer/administration & dosage , Sevelamer/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism , Species Specificity
7.
Clin Transl Sci ; 11(5): 461-470, 2018 09.
Article in English | MEDLINE | ID: mdl-29877628

ABSTRACT

The Assay Guidance Manual (AGM) is an eBook of best practices for the design, development, and implementation of robust assays for early drug discovery. Initiated by pharmaceutical company scientists, the manual provides guidance for designing a "testing funnel" of assays to identify genuine hits using high-throughput screening (HTS) and advancing them through preclinical development. Combined with a workshop/tutorial component, the overall goal of the AGM is to provide a valuable resource for training translational scientists.


Subject(s)
Biological Assay/methods , Drug Discovery , Geography , High-Throughput Screening Assays , Humans , Translational Research, Biomedical
8.
J Med Chem ; 59(24): 10891-10916, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27749056

ABSTRACT

The G protein-coupled receptor 40 (GPR40) also known as free fatty acid receptor 1 (FFAR1) is highly expressed in pancreatic, islet ß-cells and responds to endogenous fatty acids, resulting in amplification of insulin secretion only in the presence of elevated glucose levels. Hypothesis driven structural modifications to endogenous FFAs, focused on breaking planarity and reducing lipophilicity, led to the identification of spiropiperidine and tetrahydroquinoline acid derivatives as GPR40 agonists with unique pharmacology, selectivity, and pharmacokinetic properties. Compounds 1 (LY2881835), 2 (LY2922083), and 3 (LY2922470) demonstrated potent, efficacious, and durable dose-dependent reductions in glucose levels along with significant increases in insulin and GLP-1 secretion during preclinical testing. A clinical study with 3 administered to subjects with T2DM provided proof of concept of 3 as a potential glucose-lowering therapy. This manuscript summarizes the scientific rationale, medicinal chemistry, preclinical, and early development data of this new class of GPR40 agonists.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Drug Discovery , Hypoglycemic Agents/pharmacology , Piperidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Spiro Compounds/pharmacology , Animals , Dose-Response Relationship, Drug , Glucose Tolerance Test , HEK293 Cells , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Rats , Rats, Zucker , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Structure-Activity Relationship
9.
Eur J Pharmacol ; 482(1-3): 31-7, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14660002

ABSTRACT

Four dopamine receptor agonists used for the treatment of Parkinson's disease (apomorphine, pergolide, ropinirole and sumanirole) were evaluated for the ability to block human ether-a-go-go related gene (hERG) K(+) channels and to modify the duration of canine Purkinje fiber action potentials. Apomorphine, pergolide and ropinirole blocked the hERG-mediated currents with IC(50) values of 2.4, 0.12 and 1.2 microM, respectively. When evaluated in an action potential duration assay, pergolide significantly shortened action potential duration at 90% repolarization (APD(90)) whereas apomorphine and ropinirole significantly prolonged repolarization. Sumanirole only partially blocked hERG K(+) channels at the highest tested concentration (10 microM) and did not modify action potential duration over the tested concentration range (0.65-65 microM). Taken together, these data provide evidence that dopamine receptor agonists developed for the treatment of Parkinson's disease differentially influence hERG K(+) channel function and cardiac action potential duration.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Dopamine Agonists/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Purkinje Fibers/drug effects , Purkinje Fibers/metabolism , Receptors, Dopamine/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , CHO Cells , Cricetinae , Dogs , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels , In Vitro Techniques , Male , Potassium Channel Blockers/pharmacology
10.
Int J Antimicrob Agents ; 23(3): 226-34, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15164962

ABSTRACT

Staphylococcus epidermidis has emerged as a major nosocomial pathogen that is often associated with infections of indwelling medical devices. Microbial adhesion to implanted foreign materials is a prerequisite for establishing infection. We studied the time-dependent anti-adhesion effects of linezolid and vancomycin on three S. epidermidis clinical isolates. Minimum inhibitory concentration (MIC) values were identical for both agents for all three isolates (2 mg/l). Bacterial suspensions were added to polystyrene wells and treated with 0.5-4 times the MIC of linezolid or vancomycin at 0, 2, 4 or 6 h post-inoculation. Supra-inhibitory (2 and 4 x MIC) and inhibitory (MIC) concentrations of linezolid demonstrated potent anti-adhesion activity following 2 and 4 h deferred treatments. Even at sub-inhibitory concentrations (0.5 x MIC), suppression of staphylococcal adherence to polystyrene was still evident in most cultures. Linezolid at two and four times the MIC also exerted significant inhibitory effects in cultures that had been treated with a 6-h delay. Supra-inhibitory and inhibitory concentrations of vancomycin administered 2 h post-infection appeared equally effective as linezolid. However, sub-inhibitory concentrations of vancomycin showed minimal or no activity against bacterial adhesion. When vancomycin treatments were delayed by 4 h, only concentrations above the MIC prevented adherence. Linezolid has promising in vitro anti-adhesion activity that merits further studies to determine its role in the management of foreign-body infections.


Subject(s)
Acetamides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Oxazolidinones/pharmacology , Staphylococcus epidermidis/drug effects , Acetamides/administration & dosage , Anti-Bacterial Agents/administration & dosage , Cross Infection/drug therapy , Humans , In Vitro Techniques , Linezolid , Microscopy, Electron, Scanning , Oxazolidinones/administration & dosage , Staphylococcal Infections/drug therapy , Staphylococcus epidermidis/pathogenicity , Time Factors , Vancomycin/administration & dosage , Vancomycin/pharmacology
11.
PLoS One ; 9(4): e93297, 2014.
Article in English | MEDLINE | ID: mdl-24695114

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is a syndrome characterized by the rapid loss of the kidney excretory function and is strongly associated with increased early and long-term patient morbidity and mortality. Early diagnosis of AKI is challenging; therefore we profiled plasma microRNA in an effort to identify potential diagnostic circulating markers of renal failure. The goal of the present study was to investigate the dynamic relationship of circulating and renal microRNA profiles within the first 24 hours after bilateral ischemia-reperfusion kidney injury in mice. METHODOLOGY/PRINCIPAL FINDINGS: Bilateral renal ischemia was induced in C57Bl/6 mice (n = 10 per group) by clamping the renal pedicle for 27 min. Ischemia-reperfusion caused highly reproducible, progressive, concordant elevation of miR-714, miR-1188, miR-1897-3p, miR-877*, and miR-1224 in plasma and kidneys at 3, 6 and 24 hours after acute kidney injury compared to the sham-operated mice (n = 5). These dynamics correlated with histologic findings of kidney injury and with a conventional plasma marker of renal dysfunction (creatinine). Pathway analysis revealed close association between miR-1897-3p and Nucks1 gene expression, which putative downstream targets include genes linked to renal injury, inflammation and apoptosis. CONCLUSIONS/SIGNIFICANCE: Systematic profiling of renal and plasma microRNAs in the early stages of experimental AKI provides the first step in advancing circulating microRNAs to the level of promising novel biomarkers.


Subject(s)
Acute Kidney Injury/metabolism , Ischemia/metabolism , Kidney/metabolism , MicroRNAs/metabolism , Plasma/metabolism , Reperfusion Injury/metabolism , Animals , Biomarkers/metabolism , Creatinine/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Reperfusion/methods
12.
Atherosclerosis ; 213(2): 632-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21040917

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that regulates cholesterol metabolism through low-density lipoprotein receptor (LDLR) degradation. Gain-of-function and loss-of-function mutations within PCSK9 gene lead to hypercholesterolemia or hypocholesterolemia respectively. Studies in the U.S. and Canada reported a correlation between multiple metabolic factors and circulating PCSK9 concentrations. However, there is no data available on circulating PCSK9 levels in Chinese. A sandwich ELISA assay was applied to measure serum PCSK9 levels in a Chinese population of 2719 adults from Nanjing district, China, which represents a large and uniform ethnic population of Han Chinese. Serum PCSK9 levels ranged from 12.85 to 222.50 ng/ml with a mean concentration of 69.35 ng/ml in this population. Serum PCSK9 levels were slightly higher in women than in men. Compared to premenopausal women, postmenopausal women had significantly higher PCSK9 levels. Serum PCSK9 levels were correlated with multiple metabolic variables including age, BMI, total cholesterol, LDL cholesterol, triglycerides, fasting blood glucose, systolic blood pressure (SP) and diastolic blood pressure (DP) in this population. After stepwise regression analysis, there was a significant positive association between serum PCSK9 levels and total cholesterol, triglycerides and SP in men. In women, there was a positive correlation between PCSK9 levels and total cholesterol, age and DP. Our study indicates that the serum PCSK9 level may be a biomarker of metabolic status and cardiovascular disease.


Subject(s)
Biomarkers/blood , Cardiovascular Diseases/blood , Serine Endopeptidases/blood , Adult , Aged , Aged, 80 and over , Asian People/genetics , Blood Glucose/metabolism , Blood Pressure , China , Cholesterol/blood , Cholesterol, LDL/blood , Female , Humans , Male , Middle Aged , Postmenopause , Proprotein Convertase 9 , Proprotein Convertases , Serine Endopeptidases/genetics , Triglycerides/blood
13.
PLoS One ; 2(12): e1360, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18159243

ABSTRACT

Schizophrenia is often associated with emotional blunting--the diminished ability to respond to emotionally salient stimuli--particularly those stimuli representative of negative emotional states, such as fear. This disturbance may stem from dysfunction of the amygdala, a brain region involved in fear processing. The present article describes a novel animal model of emotional blunting in schizophrenia. This model involves interfering with normal fear processing (classical conditioning) in rats by means of acute ketamine administration. We confirm, in a series of experiments comprised of cFos staining, behavioral analysis and neurochemical determinations, that ketamine interferes with the behavioral expression of fear and with normal fear processing in the amygdala and related brain regions. We further show that the atypical antipsychotic drug clozapine, but not the typical antipsychotic haloperidol nor an experimental glutamate receptor 2/3 agonist, inhibits ketamine's effects and retains normal fear processing in the amygdala at a neurochemical level, despite the observation that fear-related behavior is still inhibited due to ketamine administration. Our results suggest that the relative resistance of emotional blunting to drug treatment may be partially due to an inability of conventional therapies to target the multiple anatomical and functional brain systems involved in emotional processing. A conceptual model reconciling our findings in terms of neurochemistry and behavior is postulated and discussed.


Subject(s)
Emotions , Models, Animal , Schizophrenic Psychology , Animals , Behavior, Animal , Conditioning, Classical , Dopamine/metabolism , Fear , Glutamic Acid/metabolism , Male , Rats , Rats, Sprague-Dawley
14.
Antimicrob Agents Chemother ; 47(8): 2655-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12878535

ABSTRACT

The efficacy of linezolid, alone or in combination with rifampin, against methicillin-susceptible Staphylococcus aureus in rabbits with experimental endocarditis was investigated. Linezolid (50 or 75 mg/kg of body weight), rifampin, and linezolid (25, 50, or 75 mg/kg) plus rifampin produced statistically significant reductions in bacterial counts compared with those in untreated controls. Plasma or valvular vegetation levels of linezolid in the groups treated with the linezolid-rifampin combination were similar to those in the respective linezolid-only treatment groups. At therapeutic levels of linezolid, rifampin resistance was not observed. The results from this experimental model of endocarditis suggest that while rifampin did not provide synergy to the linezolid dosing, it did not antagonize the efficacy of linezolid.


Subject(s)
Acetamides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Antibiotics, Antitubercular/therapeutic use , Endocarditis, Bacterial/drug therapy , Methicillin/pharmacology , Oxazolidinones/therapeutic use , Penicillins/pharmacology , Rifampin/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Animals , Colony Count, Microbial , Drug Synergism , Drug Therapy, Combination/therapeutic use , Endocarditis, Bacterial/microbiology , Heart Valves/microbiology , Kidney/microbiology , Linezolid , Male , Microbial Sensitivity Tests , Rabbits , Staphylococcal Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL