Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Jt Comm J Qual Patient Saf ; 50(7): 492-499, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705745

ABSTRACT

BACKGROUND: Safety event reporting and review is well established within US hospitals, but systems to ensure implementation of changes to improve patient safety are less developed. METHODS: Contributing factors and corrective actions for events brought to a tertiary care academic medical center's multidisciplinary hospital-level safety event review meeting were prospectively collected from 2020 to 2021. Corrective actions were tracked to completion through 2023. The authors retrospectively coded corrective actions by category and strength using the US Department of Veterans Affairs/Institute for Healthcare Improvement Action Hierarchy Tool. RESULTS: In the analysis of 67 events, 15 contributing factor themes were identified and resulted in 148 corrective actions. Of these events, 85.1% (57/67) had more than one corrective action. Of the 148 corrective actions, 84 (56.8%) were rated as weak, 36 (24.3%) as intermediate, 15 (10.1%) strong, and 13 (8.8%) needed more information. The completion rate was 97.6% (for weak corrective actions), 80.6% (intermediate), and 73.3% (strong) (p < 0.0001). CONCLUSION: Safety events were often addressed with multiple corrective actions. There was an inverse relationship between intervention strength and completion, the strongest interventions with the lowest rate of completion. By integrating action strength and completion status into corrective action follow-up, health care organizations may more effectively identify and address those barriers to completing the strongest interventions that ultimately achieve high reliability.


Subject(s)
Medical Errors , Patient Safety , Humans , Patient Safety/standards , Medical Errors/prevention & control , Medical Errors/statistics & numerical data , Quality Improvement/organization & administration , United States , Safety Management/organization & administration , Safety Management/standards , Academic Medical Centers/organization & administration , Retrospective Studies
2.
J Neurophysiol ; 116(6): 2663-2675, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27655963

ABSTRACT

Agonists of the α7-nicotinic acetylcholine receptor (α7-nAChR) have entered clinical trials as procognitive agents for treating schizophrenia and Alzheimer's disease. The most advanced compounds are orthosteric agonists, which occupy the ligand binding site. At the molecular level, agonist activation of α7-nAChR is reasonably well understood. However, the consequences of activating α7-nAChRs on neural circuits underlying cognition remain elusive. Here we report that an α7-nAChR agonist (FRM-17848) enhances long-term potentiation (LTP) in rat septo-hippocampal slices far below the cellular EC50 but at a concentration that coincides with multiple functional outcome measures as we reported in Stoiljkovic M, Leventhal L, Chen A, Chen T, Driscoll R, Flood D, Hodgdon H, Hurst R, Nagy D, Piser T, Tang C, Townsend M, Tu Z, Bertrand D, Koenig G, Hajós M. Biochem Pharmacol 97: 576-589, 2015. In this same concentration range, we observed a significant increase in spontaneous γ-aminobutyric acid (GABA) inhibitory postsynaptic currents and a moderate suppression of excitability in whole cell recordings from rat CA1 pyramidal neurons. This modulation of GABAergic activity is necessary for the LTP-enhancing effects of FRM-17848, since inhibiting GABAA α5-subunit-containing receptors fully reversed the effects of the α7-nAChR agonist. These data suggest that α7-nAChR agonists may increase synaptic plasticity in hippocampal slices, at least in part, through a circuit-level enhancement of a specific subtype of GABAergic receptor.


Subject(s)
GABAergic Neurons/drug effects , Hippocampus/cytology , Inhibitory Postsynaptic Potentials/drug effects , Nerve Net/physiology , Nicotinic Agonists/pharmacology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Cholinesterase Inhibitors/pharmacology , Donepezil , Evoked Potentials/drug effects , Evoked Potentials/genetics , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Agents/pharmacology , GABAergic Neurons/physiology , Humans , Indans/pharmacology , Male , Nerve Net/drug effects , Oocytes , Piperidines/pharmacology , Quinuclidines/pharmacology , Rats , Rats, Sprague-Dawley , Thiophenes/pharmacology , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
3.
Pharmacol Rev ; 67(4): 1025-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26419447

ABSTRACT

Progress in the fields of neuroscience and molecular biology has identified the forebrain cholinergic system as being important in many higher order brain functions. Further analysis of the genes encoding the nicotinic acetylcholine receptors (nAChRs) has highlighted, in particular, the role of α7 nAChRs in these higher order brain functions as evidenced by their peculiar physiologic and pharmacological properties. As this receptor has gained the attention of scientists from academia and industry, our knowledge of its roles in various brain and bodily functions has increased immensely. We have also seen the development of small molecules that have further refined our understanding of the roles of α7 nAChRs, and these molecules have begun to be tested in clinical trials for several indications. Although a large body of data has confirmed a role of α7 nAChRs in cognition, the translation of small molecules affecting α7 nAChRs into therapeutics has to date only progressed to the stage of testing in clinical trials. Notably, however, most recent human genetic and biochemical studies are further underscoring the crucial role of α7 nAChRs and associated genes in multiple organ systems and disease states. The aim of this review is to discuss our current knowledge of α7 nAChRs and their relevance as a target in specific functional systems and disease states.


Subject(s)
Brain/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Humans , Immune System Diseases/genetics , Immune System Diseases/physiopathology , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Polymorphism, Single Nucleotide , Signal Transduction , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor/genetics
4.
Biochem Pharmacol ; 97(4): 576-589, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26206187

ABSTRACT

Pharmacological activation of α7 nicotinic acetylcholine receptors (α7 nAChRs) may improve cognition in schizophrenia and Alzheimer's disease. The present studies describe an integrated pharmacological analysis of the effects of FRM-17874, an analogue of encenicline, on α7 nAChRs in vitro and in behavioral and neurophysiological assays relevant to cognitive function. FRM-17874 demonstrated high affinity binding to human α7 nAChRs, displacing [(3)H]-methyllacaconitine (Ki=4.3nM). In Xenopus laevis oocytes expressing human α7 nAChRs, FRM-17874 acted as an agonist, evoking inward currents with an EC50 of 0.42µM. Lower concentrations of FRM-17874 (0.01-3nM) elicited no detectable current, but primed receptors to respond to sub-maximal concentrations of acetylcholine. FRM-17874 improved novel object recognition in rats, and enhanced memory acquisition and reversal learning in the mouse water T-maze. Neurophysiological correlates of cognitive effects of drug treatment, such as synaptic transmission, long-term potentiation, and hippocampal theta oscillation were also evaluated. Modulation of synaptic transmission and plasticity was observed in rat hippocampal slices at concentrations of 3.2 and 5nM. FRM-17874 showed a dose-dependent facilitation of stimulation-induced hippocampal theta oscillation in mice and rats. The FRM-17874 unbound brain concentration-response relationship for increased theta oscillation power was similar in both species, exhibited a biphasic pattern peaking around 3nM, and overlapped with active doses and exposures observed in cognition assays. In summary, behavioral and neurophysiological assays indicate a bell-shaped effective concentration range and this report represents the first attempt to explain the concentration-response function of α7 nAChR-mediated pro-cognitive effects in terms of receptor pharmacology.


Subject(s)
Quinuclidines/pharmacology , Thiophenes/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Behavior, Animal/drug effects , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Gene Expression Regulation , Hippocampus/metabolism , Humans , Learning/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Oocytes/drug effects , Oocytes/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor/genetics
5.
Behav Pharmacol ; 26(4): 403-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25794332

ABSTRACT

We investigated whether the effects of acutely administered EVP-6124, an α7 nicotinic acetylcholine receptor (α7 nAChR) agonist, on cognition were maintained after 6-day continuous minipump administration. Performance in a delay-dependent forgetting test was measured in the object recognition task after single-oral doses of 0.3 or 1 mg/kg, or at plasma steady-state concentrations (Css) of 0.6 or 2 ng/ml, which were similar to the efficacious plasma concentrations after single-oral dosing. The 0.3 mg/kg acute dose enhanced memory at a total plasma concentration of ∼0.3 ng/ml at 1-4 h after dosing. Continuous treatment produced total plasma Css values of 0.48 and 1.93 ng/ml on day 6 and enhanced memory. At EVP-6124 plasma concentrations that optimally enhance memory in the object recognition task, tolerance did not develop after 6 days of continuous treatment.


Subject(s)
Nootropic Agents/administration & dosage , Nootropic Agents/pharmacokinetics , Quinuclidines/administration & dosage , Quinuclidines/pharmacokinetics , Recognition, Psychology/drug effects , Thiophenes/administration & dosage , Thiophenes/pharmacokinetics , alpha7 Nicotinic Acetylcholine Receptor/agonists , Administration, Oral , Animals , Blood Chemical Analysis , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Tolerance , Male , Models, Animal , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacokinetics , Pilot Projects , Rats, Wistar , Recognition, Psychology/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
6.
Biochem Pharmacol ; 91(4): 543-51, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25091561

ABSTRACT

Two investigational compounds (FRM-1, (R)-7-fluoro-N-(quinuclidin-3-yl)benzo[b]thiophene-2-carboxamide and FRM-2, (R)-7-cyano-N-(quinuclidin-3-yl)benzo[b]thiophene-2-carboxamide) resided in rat brain longer than in systemic circulation. In Caco-2 directional transport studies, they both showed good intrinsic passive permeability but differed significantly in efflux susceptibility (efflux ratio of <2 and ∼7, respectively), largely attributed to P-glycoprotein (P-gp). Capitalizing on these interesting properties, we investigated how cerebrospinal fluid (CSF) concentration (CCSF) would be shaped by unbound plasma concentration (Cu,p) and unbound brain concentration (Cu,b) in disequilibrium conditions and at steady state. Following subcutaneous administration, FRM-1CCSF largely followed Cu,p initially and leveled between Cu,p and Cu,b. However, it gradually approached Cu,b and became lower than, but parallel to Cu,b at the terminal phase. In contrast, FRM-2CCSF temporal profile mostly paralleled the Cu,p but was at a much lower level. Upon intravenous infusion to steady state, FRM-1CCSF and Cu,b were similar, accounting for 61% and 69% of the Cu,p, indicating a case of largely passive diffusion-governed brain penetration where CCSF served as a good surrogate for Cu,b. On the contrary, FRM-2CCSF and Cu,b were remarkably lower than Cu,p (17% and 8% of Cu,p, respectively), suggesting that FRM-2 brain penetration was severely impaired by P-gp-mediated efflux and CCSF underestimated this impact. A semi-physiologically based pharmacokinetic (PBPK) model was constructed that adequately described the temporal profiles of the compounds in the plasma, brain and CSF. Our work provided some insight into the relative importance of blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) in modulating CCSF.


Subject(s)
Brain/metabolism , Thiophenes/pharmacokinetics , Animals , Blood-Brain Barrier , Caco-2 Cells , Humans , Male , Models, Theoretical , Rats , Rats, Wistar , Thiophenes/cerebrospinal fluid
7.
Psychopharmacology (Berl) ; 231(23): 4541-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24810107

ABSTRACT

BACKGROUND: Alpha7 and α4ß2 nicotinic acetylcholine receptor (nAChR) agonists have been shown to improve cognition in various animal models of cognitive impairment and are of interest as treatments for schizophrenia, Alzheimer's disease, and other cognitive disorders. Increased release of dopamine (DA), acetylcholine (ACh), glutamate (Glu), and γ-aminobutyric acid (GABA) in cerebral cortex, hippocampus, and nucleus accumbens (NAC) has been suggested to contribute to their beneficial effects on cognition. RESULTS: Using in vivo microdialysis, we found that EVP-6124 [(R)-7-chloro-N-quinuclidin-3-yl)benzo[b]thiophene-2-carboxamide], a high-affinity α7 nAChR partial agonist, at 0.1 mg/kg, s.c., increased DA efflux in the medial prefrontal cortex (mPFC) and NAC. EVP-6124, 0.1 and 0.3 mg/kg, also increased efflux of ACh in the mPFC but not in the NAC. Similarly, EVP-6124, 0.1 mg/kg, but not 0.03 and 0.3 mg/kg, significantly increased mPFC Glu efflux. Thus, EVP-6124 produced an inverted U-shaped curve for DA and Glu release, as previously reported for other α7 nAChR agonists. The three doses of EVP-6124 did not produce a significant effect on GABA efflux in either region. Pretreatment with the selective α7 nAChR antagonist, methyllycaconitine (MLA, 1.0 mg/kg), significantly blocked cortical DA and Glu efflux induced by EVP-6124 (0.1 mg/kg), suggesting that the effects of EVP-6124 on these neurotransmitters were due to α7 nAChR agonism. MLA only partially blocked the effects of EVP-6124 on ACh efflux in the mPFC. CONCLUSION: These results suggest increased cortical DA, ACh, and Glu release, which may contribute to the ability of the α7 nAChR agonist, EVP-6124, to treat cognitive impairment and possibly other dimensions of psychopathology.


Subject(s)
Acetylcholine/metabolism , Dopamine/metabolism , Glutamic Acid/metabolism , Nicotinic Agonists/pharmacology , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Quinuclidines/pharmacology , Thiophenes/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Male , Microdialysis , Nicotinic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor/agonists , gamma-Aminobutyric Acid/metabolism
8.
Mol Neurodegener ; 7: 61, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23249765

ABSTRACT

BACKGROUND: A hallmark of Alzheimer's disease is the presence of senile plaques in human brain primarily containing the amyloid peptides Aß42 and Aß40. Many drug discovery efforts have focused on decreasing the production of Aß42 through γ-secretase inhibition. However, identification of γ-secretase inhibitors has also uncovered mechanism-based side effects. One approach to circumvent these side effects has been modulation of γ-secretase to shift Aß production to favor shorter, less amyloidogenic peptides than Aß42, without affecting the overall cleavage efficiency of the enzyme. This approach, frequently called γ-secretase modulation, appears more promising and has lead to the development of new therapeutic candidates for disease modification in Alzheimer's disease. RESULTS: Here we describe EVP-0015962, a novel small molecule γ-secretase modulator. EVP-0015962 decreased Aß42 in H4 cells (IC50 = 67 nM) and increased the shorter Aß38 by 1.7 fold at the IC50 for lowering of Aß42. AßTotal, as well as other carboxyl-terminal fragments of amyloid precursor protein, were not changed. EVP-0015962 did not cause the accumulation of other γ-secretase substrates, such as the Notch and ephrin A4 receptors, whereas a γ-secretase inhibitor reduced processing of both. A single oral dose of EVP-0015962 (30 mg/kg) decreased Aß42 and did not alter AßTotal peptide levels in a dose-dependent manner in Tg2576 mouse brain at an age when overt Aß deposition was not present. In Tg2576 mice, chronic treatment with EVP-0015962 (20 or 60 mg/kg/day in a food formulation) reduced Aß aggregates, amyloid plaques, inflammatory markers, and cognitive deficits. CONCLUSIONS: EVP-0015962 is orally bioavailable, detected in brain, and a potent, selective γ-secretase modulator in vitro and in vivo. Chronic treatment with EVP-0015962 was well tolerated in mice and lowered the production of Aß42, attenuated memory deficits, and reduced Aß plaque formation and inflammation in Tg2576 transgenic animals. In summary, these data suggest that γ-secretase modulation with EVP-0015962 represents a viable therapeutic alternative for disease modification in Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/drug effects , Amyloid beta-Peptides/drug effects , Behavior, Animal/drug effects , Biphenyl Compounds/pharmacology , Phenylpropionates/pharmacology , Propionates/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Transfection
9.
Neuropharmacology ; 62(2): 1099-110, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22085888

ABSTRACT

EVP-6124, (R)-7-chloro-N-quinuclidin-3-yl)benzo[b]thiophene-2-carboxamide, is a novel partial agonist of α7 neuronal nicotinic acetylcholine receptors (nAChRs) that was evaluated here in vitro and in vivo. In binding and functional experiments, EVP-6124 showed selectivity for α7 nAChRs and did not activate or inhibit heteromeric α4ß2 nAChRs. EVP-6124 had good brain penetration and an adequate exposure time. EVP-6124 (0.3 mg/kg, p.o.) significantly restored memory function in scopolamine-treated rats (0.1 mg/kg, i.p.) in an object recognition task (ORT). Although donepezil at 0.1 mg/kg, p.o. or EVP-6124 at 0.03 mg/kg, p.o. did not improve memory in this task, co-administration of these sub-efficacious doses fully restored memory. In a natural forgetting test, an ORT with a 24 h retention time, EVP-6124 improved memory at 0.3 mg/kg, p.o. This improvement was blocked by the selective α7 nAChR antagonist methyllycaconitine (0.3 mg/kg, i.p. or 10 µg, i.c.v.). In co-application experiments of EVP-6124 with acetylcholine, sustained exposure to EVP-6124 in functional investigations in oocytes caused desensitization at concentrations greater than 3 nM, while lower concentrations (0.3-1 nM) caused an increase in the acetylcholine-evoked response. These actions were interpreted as representing a co-agonist activity of EVP-6124 with acetylcholine on α7 nAChRs. The concentrations of EVP-6124 that resulted in physiological potentiation were consistent with the free drug concentrations in brain that improved memory performance in the ORT. These data suggest that the selective partial agonist EVP-6124 improves memory performance by potentiating the acetylcholine response of α7 nAChRs and support new therapeutic strategies for the treatment of cognitive impairment. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Subject(s)
Brain/drug effects , Memory/drug effects , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Receptors, Nicotinic/metabolism , Thiophenes/pharmacology , Animals , Brain/metabolism , Cholinesterase Inhibitors/pharmacology , Donepezil , Dose-Response Relationship, Drug , Drug Partial Agonism , Indans/pharmacology , Male , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , alpha7 Nicotinic Acetylcholine Receptor
10.
J Pharmacol Exp Ther ; 340(1): 124-33, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22001260

ABSTRACT

CEP-26401 [irdabisant; 6-{4-[3-((R)-2-methyl-pyrrolidin-1-yl)-propoxy]-phenyl}-2H-pyridazin-3-one HCl] is a novel, potent histamine H3 receptor (H3R) antagonist/inverse agonist with drug-like properties. High affinity of CEP-26401 for H3R was demonstrated in radioligand binding displacement assays in rat brain membranes (K(i) = 2.7 ± 0.3 nM) and recombinant rat and human H3R-expressing systems (K(i) = 7.2 ± 0.4 and 2.0 ± 1.0 nM, respectively). CEP-26401 displayed potent antagonist and inverse agonist activities in [³5S]guanosine 5'-O-(γ-thio)triphosphate binding assays. After oral dosing of CEP-26401, occupancy of H3R was estimated by the inhibition of ex vivo binding in rat cortical slices (OCC50 = 0.1 ± 0.003 mg/kg), and antagonism of the H3R agonist R-α-methylhistamine- induced drinking response in the rat dipsogenia model was demonstrated in a similar dose range (ED50 = 0.06 mg/kg). CEP-26401 improved performance in the rat social recognition model of short-term memory at doses of 0.01 to 0.1 mg/kg p.o. and was wake-promoting at 3 to 30 mg/kg p.o. In DBA/2NCrl mice, CEP-26401 at 10 and 30 mg/kg i.p. increased prepulse inhibition (PPI), whereas the antipsychotic risperidone was effective at 0.3 and 1 mg/kg i.p. Coadministration of CEP-26401 and risperidone at subefficacious doses (3 and 0.1 mg/kg i.p., respectively) increased PPI. These results demonstrate potent behavioral effects of CEP-26401 in rodent models and suggest that this novel H3R antagonist may have therapeutic utility in the treatment of cognitive and attentional disorders. CEP-26401 may also have therapeutic utility in treating schizophrenia or as adjunctive therapy to approved antipsychotics.


Subject(s)
Cognition/drug effects , Histamine H3 Antagonists/pharmacology , Nootropic Agents , Pyridazines/pharmacology , Pyrrolidines/pharmacology , Wakefulness/drug effects , Animals , Autoradiography , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Drinking/drug effects , Electroencephalography/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Memory, Short-Term/drug effects , Radioligand Assay , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Reflex, Startle/drug effects , Sleep/drug effects , Social Behavior
11.
Neurobiol Dis ; 43(3): 565-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21605674

ABSTRACT

In vivo detection of Alzheimer's disease (AD) neuropathology in living patients using positron emission tomography (PET) in conjunction with high affinity molecular imaging probes for ß-amyloid (Aß) and tau has the potential to assist with early diagnosis, evaluation of disease progression, and assessment of therapeutic interventions. Animal models of AD are valuable for exploring the in vivo binding of these probes, particularly their selectivity for specific neuropathologies, but prior PET experiments in transgenic mice have yielded conflicting results. In this work, we utilized microPET imaging in a transgenic rat model of brain Aß deposition to assess [F-18]FDDNP binding profiles in relation to age-associated accumulation of neuropathology. Cross-sectional and longitudinal imaging demonstrated that [F-18]FDDNP binding in the hippocampus and frontal cortex progressively increases from 9 to 18months of age and parallels age-associated Aß accumulation. Specificity of in vivo [F-18]FDDNP binding was assessed by naproxen pretreatment, which reversibly blocked [F-18]FDDNP binding to Aß aggregrates. Both [F-18]FDDNP microPET imaging and neuropathological analyses revealed decreased Aß burden after intracranial anti-Aß antibody administration. The combination of this non-invasive imaging method and robust animal model of brain Aß accumulation allows for future longitudinal in vivo assessments of potential therapeutics for AD that target Aß production, aggregation, and/or clearance. These results corroborate previous analyses of [F-18]FDDNP PET imaging in clinical populations.


Subject(s)
Aging/pathology , Alzheimer Disease/diagnostic imaging , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/immunology , Antibodies, Blocking/pharmacology , Nitriles , Positron-Emission Tomography/methods , Aging/immunology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/diagnostic imaging , Amyloidosis/genetics , Amyloidosis/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Binding, Competitive/immunology , Disease Models, Animal , Fluorine Radioisotopes , Humans , Naproxen/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Transgenic
12.
Psychopharmacology (Berl) ; 215(3): 441-54, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21301810

ABSTRACT

RATIONALE: Naturally low prepulse inhibition (PPI) in DBA/2 mice is increased by marketed antipsychotics and compounds acting at novel targets relevant to schizophrenia. Whether other mouse strains with naturally low PPI respond similarly and could be translational models of schizophrenia is unknown. OBJECTIVE: Baseline levels of PPI were determined in outbred CF-1 and Black Swiss mice. CF-1 and Black Swiss mice were then compared to DBA/2 mice for their responses to typical (haloperidol) and atypical (clozapine) antipsychotics and to compounds with potential antipsychotic activity, a histamine H(3) receptor antagonist (thioperamide) and a glycine transporter-1 inhibitor (SSR504734). RESULTS: CF-1 and Black Swiss mice had naturally low PPI, similar to the level in C57BL/6 mice, but higher than that in DBA/2 mice. Haloperidol (0.3-1 mg/kg) increased PPI in DBA/2, CF-1, and Black Swiss mice. Clozapine (3 mg/kg) increased PPI in DBA/2 and CF-1 mice, but not in Black Swiss mice. Thioperamide (10-30 mg/kg) and SSR504734 (30 mg/kg) increased PPI only in DBA/2 mice. Strain differences in PPI responsiveness were not due to differences in brain concentrations of the tested compounds. CONCLUSIONS: CF-1 mice with naturally low PPI may be useful for testing typical and atypical antipsychotics while Black Swiss mice only responded to a typical antipsychotic. DBA/2 mice remain the only strain with naturally low PPI that responds to marketed antipsychotics, as well as to compounds with novel mechanisms of action. Thus, DBA/2 mice may be the strain of choice for screening novel chemical entities for their ability to increase PPI.


Subject(s)
Antipsychotic Agents/pharmacology , Reflex, Startle/drug effects , Schizophrenia/drug therapy , Animals , Animals, Outbred Strains , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Schizophrenia/physiopathology , Species Specificity
13.
Biochem Pharmacol ; 81(12): 1422-34, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21295552

ABSTRACT

The need to develop effective treatments for Alzheimer's disease has been confounded by repeated clinical failures where promising new chemical entities that have been extensively characterized in preclinical models of Alzheimer's disease have failed to show efficacy in the human disease state. This has been attributed to: the selection of drug targets that have yet to be shown as causal to the disease as distinct from being the result of the disease process, a lack of congruence in the animal models of Alzheimer's disease, wild-type and transgenic, to the human disease, and the enrollment of patients in proof of concept clinical trials who are at too advanced a stage of the disease to respond to any therapeutic. The development of validated biomarkers that can be used for disease diagnosis and progression is anticipated to improve patient enrollment in clinical trials, to develop new animal models and to identify new disease targets for drug discovery. The present review assesses the status of current efforts in developing CSF biomarkers for Alzheimer's disease and briefly discusses the status of CSF biomarker efforts in schizophrenia, depression, Parkinson's disease and multiple sclerosis.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/drug therapy , Central Nervous System Agents/therapeutic use , Translational Research, Biomedical/methods , Animals , Biomarkers/cerebrospinal fluid , Disease Models, Animal , Humans , Neuropsychiatry
14.
J Pharmacol Exp Ther ; 337(2): 380-90, 2011 May.
Article in English | MEDLINE | ID: mdl-21300706

ABSTRACT

Sydnocarb is a psychomotor stimulant structurally similar to d-amphetamine (D-AMPH) and is used in Russia for the treatment of a variety of neuropsychiatric comorbidities. The nature of sydnocarb-induced facilitation of dopamine (DA) neurotransmission [DA release versus DA transporter (DAT) inhibition] is not clear. The present study characterized the pharmacological actions and behavioral effects of intraperitoneal sydnocarb in male Sprague-Dawley rats. Where relevant, comparisons were made with intraperitoneal D-AMPH. Unlike D-AMPH, which causes release of DA from rat synaptosomes (EC(50) = 0.10 µM; 95% confidence limits, 0.06-0.18), sydnocarb (up to 100 µM) did not. Sydnocarb potently (K(i) = 8.3 ± 0.7 nM) blocked recombinant human DAT expressed in Chinese hamster ovary-K1 cells and less potently blocked the norepinephrine transporter (K(i) = 10.1 ± 1.5 µM). Sydnocarb at 10 µM did not bind to 64 other targets. In rats, 10 and 30 mg/kg sydnocarb showed a 2-fold longer half-life in plasma and brain and a 5-fold lower brain-to-plasma ratio compared with 0.3 and 1 mg/kg D-AMPH. In the Irwin assay, sydnocarb was well tolerated up to 30 mg/kg; D-AMPH-like stereotypic behaviors were evident at 100 mg/kg. Behavioral effects of 30 mg/kg sydnocarb and 0.3 mg/kg D-AMPH were comparable. In a sleep/wake assay, 10 mg/kg sydnocarb and 1 mg/kg D-AMPH increased wakefulness comparably; however, sydnocarb (up to 30 mg/kg) did not induce D-AMPH-like rebound hypersomnolence (RHS). Like D-AMPH, sydnocarb enhanced theta power, an electrophysiological measure of cognitive function. In conclusion, sydnocarb is a selective and potent DAT inhibitor that produces robust increases in the wake state without RHS, and with potential cognitive-enhancing properties.


Subject(s)
Central Nervous System Stimulants/pharmacology , Dopamine Agents/pharmacology , Sydnones/pharmacology , Animals , Behavior, Animal/drug effects , Body Temperature/drug effects , Central Nervous System Stimulants/metabolism , Dextroamphetamine/pharmacology , Dopamine/metabolism , Dopamine Agents/metabolism , Electroencephalography , Electromyography , Ion Channels/metabolism , Male , Motor Activity/drug effects , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/psychology , Sydnones/metabolism , Sydnones/pharmacokinetics , Theta Rhythm/drug effects , Wakefulness/drug effects
15.
Biochem Pharmacol ; 80(9): 1407-17, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20637735

ABSTRACT

Inhibition of the glycine transporter type 1 (GlyT1) leading to potentiation of the glycine site (GlyB) on the N-methyl-d-aspartate (NMDA) receptor has been proposed as a novel therapeutic approach for schizophrenia. However, sarcosine-based GlyT1 inhibitors produce undesirable side effects including compulsive walking and respiratory distress. The influence of specific biochemical properties of GlyT1 inhibitors, such as mode of inhibition and residence time, on adverse effects is unknown. Two GlyT1 inhibitors that contain a sarcosine moiety, sarcosine and ALX-5407, and two compounds that do not contain a sarcosine moiety, Roche-7 and Merck (S)-13h, were evaluated for their potency, mode of inhibition, and target residence times in vitro, and modulation of prepulse inhibition (PPI) and locomotor activity in vivo. (S)-13h and sarcosine were competitive inhibitors while ALX-5407 and Roche-7 demonstrated mixed noncompetitive inhibition. Potency of GlyT1 inhibition (ALX-5407>(S)-13h>Roche-7≫sarcosine) did not correlate with residence time on GlyT1 (sarcosine=Roche-7≪(S)-13h

Subject(s)
Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Motor Activity/drug effects , Neural Inhibition/drug effects , Animals , Glycine/metabolism , Glycine Plasma Membrane Transport Proteins/physiology , Male , Mice , Mice, Inbred DBA , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Serine/pharmacology , Strychnine/pharmacology , Time Factors
16.
Psychopharmacology (Berl) ; 211(3): 325-36, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20549488

ABSTRACT

RATIONALE: Dopamine (DA) agonists decrease prepulse inhibition (PPI) and are widely used in translational models for the sensorimotor gating deficits in schizophrenia. Reductions in PPI induced by DA agonists are routinely reversed by antipsychotics in these translational models. Nevertheless, under conditions of low-baseline PPI, DA agonists may increase PPI in humans and experimental animals. DBA/2 mice have naturally low-baseline PPI, which as in the drug-induced translational models, is increased by antipsychotics. OBJECTIVE: Determine whether DBA/2 mice respond like other models of low-baseline PPI by evaluating the effect of psychostimulants (caffeine, 30-100 mg/kg IP) and the indirect DA agonists d-amphetamine (0.3-10 mg/kg IP), methylphenidate (10-100 mg/kg IP), and sydnocarb (10-30 mg/kg IP), a selective DA transporter inhibitor on PPI. Furthermore, baseline PPI in DBA/2 mice was increased by noise exposure and the effect of d-amphetamine was assessed. RESULTS: PPI was increased at one dose for each of the psychostimulants when baseline PPI was low in naïve DBA/2 mice. Effective doses were 3 mg/kg of d-amphetamine, 30 mg/kg of methylphenidate, 30 mg/kg of sydnocarb, and 100 mg/kg of caffeine. Higher doses of d-amphetamine (10 mg/kg) and methylphenidate (100 mg/kg IP) decreased PPI. When the baseline PPI was increased by noise exposure, 10 mg/kg of d-amphetamine only reduced PPI. CONCLUSION: Lower doses of psychostimulants increased PPI in naïve DBA/2 mice in a manner consistent with their naturally low-baseline PPI, and higher doses decreased PPI, consistent with effects observed in most mouse strains.


Subject(s)
Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Dextroamphetamine/pharmacology , Methylphenidate/pharmacology , Mice, Inbred DBA/physiology , Sensory Gating/drug effects , Sydnones/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Mice , Noise
17.
Psychopharmacology (Berl) ; 205(3): 369-77, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19404613

ABSTRACT

RATIONALE: Lithium and several antiepileptic drugs have mood-stabilizing effects in bipolar disorder and schizophrenia. Both disorders are characterized by deficits in prepulse inhibition (PPI) of the acoustic startle response. OBJECTIVES: Using the DBA/2 model of naturally low PPI, which is reliably increased by antipsychotics, five mood stabilizers in clinical use were tested to determine whether they would also increase PPI in this model. All drugs were administered intraperitoneally (i.p.) 30 min before testing. RESULTS: Lithium chloride (30 mg/kg), topiramate (100 and 300 mg/kg), carbamazepine (30, 60, and 100 mg/kg), valproic acid (178 and 316 mg/kg), and lamotrigine (3, 10, and 30 mg/kg) increased percent PPI. The antiepileptic drugs carbamazepine, valproic acid, and lamotrigine at high doses also decreased no-stimulus amplitudes and increased startle amplitudes. At high doses of carbamazepine, valproic acid, and lamotrigine, increases in percent PPI were independent of the increases in startle amplitude. CONCLUSIONS: The demonstrated efficacy of five mood stabilizers in the DBA/2 model of naturally low PPI points to the translational value of this model in predicting therapeutic activity in schizophrenia and bipolar disorder of compounds with diverse mechanisms of action.


Subject(s)
Anticonvulsants/pharmacology , Antimanic Agents/pharmacology , Central Nervous System Stimulants/pharmacology , Reflex, Startle/drug effects , Acoustic Stimulation , Animals , Carbamazepine/pharmacology , Dose-Response Relationship, Drug , Fructose/analogs & derivatives , Fructose/pharmacology , Injections, Intraperitoneal , Lamotrigine , Lithium Chloride/pharmacology , Male , Mice , Mice, Inbred DBA , Topiramate , Triazines/pharmacology , Valproic Acid/pharmacology
18.
Pharmacol Biochem Behav ; 92(3): 549-57, 2009 May.
Article in English | MEDLINE | ID: mdl-19249327

ABSTRACT

Modafinil increases waking and labeling of Fos, a marker of neuronal activation. In the present study, armodafinil, the R-enantiomer of racemic modafinil, was administered to rats at 30 or 100 mg/kg i.p. about 5 h after lights on (circadian time 5 and near the midpoint of the sleep phase of the sleep:wake cycle) to assess its effects on sleep/wake activity and Fos activation. Armodafinil at 100 mg/kg increased wakefulness for 2 h, while 30 mg/kg armodafinil only briefly increased wakefulness. Armodafinil (30 and 100 mg/kg) also increased latencies to the onset of sleep and motor activity. Armodafinil had differential effects in increasing neuronal Fos immunolabeling 2 h after administration. Armodafinil at 100 mg/kg increased numbers of Fos-labeled neurons in striatum and anterior cingulate cortex, without affecting nucleus accumbens. Armodafinil at 30 mg/kg only increased numbers of light Fos-labeled neurons in the anterior cingulate cortex. In brainstem arousal centers, 100 mg/kg armodafinil increased numbers of Fos-labeled neurons in the tuberomammillary nucleus, pedunculopontine tegmentum, laterodorsal tegmentum, locus coeruleus, and dorsal raphe nucleus. Fos activation of these brainstem arousal centers, as well as of the cortex and striatum, is consistent with the observed arousal effects of armodafinil.


Subject(s)
Benzhydryl Compounds/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Wakefulness/drug effects , Animals , Benzhydryl Compounds/pharmacokinetics , Body Temperature/drug effects , Brain/metabolism , Central Nervous System Stimulants/pharmacokinetics , Dose-Response Relationship, Drug , Male , Modafinil , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
19.
Neurobiol Aging ; 30(7): 1078-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18053619

ABSTRACT

Many transgenic mouse models of Alzheimer's disease (AD) that deposit amyloid (Abeta) have been produced, but development of an Abeta-depositing rat model has not been successful. Here, we describe a rat model with extracellular fibrillar Abeta deposition. Two lines of Sprague Dawley rats with transgenes expressing human amyloid precursor protein (APP) with the familial AD (FAD) mutations K670N/M671L and K670N/M671L/V717I were crossed. Abeta production in the double homozygous rats was sufficient for deposition by 17-18 months of age. The age of onset of Abeta deposition was reduced by crossing in a third rat line carrying a human presenilin-1 (PS-1) transgene with the FAD M146V mutation. The triple homozygous line had an onset of Abeta deposition by 7 months of age. Deposits appeared similar to those observed in the mouse models and displayed surrounding glial and phosphorylated tau reactivity. Abeta levels measured by ELISA were comparable to those reported in mouse models, suggesting that substantially greater amounts of soluble Abeta are not required in the rat to generate Abeta deposition.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Extracellular Fluid/metabolism , Plaque, Amyloid/metabolism , Age of Onset , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Biomarkers , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gliosis/metabolism , Gliosis/pathology , Gliosis/physiopathology , Humans , Mice , Mice, Transgenic , Mutation/genetics , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Transgenes/genetics , tau Proteins/metabolism
20.
Psychopharmacology (Berl) ; 195(2): 203-11, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17668187

ABSTRACT

RATIONALE: DBA/2 mice demonstrate poor prepulse inhibition (PPI) as is also observed in schizophrenic patients, and their PPI is improved by antipsychotics. Thus, the DBA/2 mouse is increasingly used for testing of novel antipsychotics in PPI; however, the strain has not been fully characterized for relevant variables affecting compound testing. OBJECTIVES: The objectives of this study were to compare four DBA/2 substrains, evaluate light- and dark-phase testing on startle, PPI, and drug-induced improvement in PPI in DBA/2NCrl mice, test chamber lighting on startle and PPI in DBA/2NCrl mice and to evaluate vehicles on baseline PPI in DBA/2NCrl mice. RESULTS: DBA/2NCrl and DBA/2J mice were acceptable for PPI testing, while DBA/2NHsd mice had diminished startle reflexes. Startle responses to the prepulses alone were observed in 46% of the DBA/2NTac mice. PPI and startle did not show diurnal variations or variations due to chamber lighting. Olanzapine and aripiprazole showed better drug-induced improvements in PPI during the light phase. The vehicle 25% (2-hydroxypropyl)-beta-cyclodextrin variably improved PPI, an effect not observed with other vehicles. CONCLUSIONS: DBA/2NHsd and DBA/2NTac mice were unacceptable for PPI experiments. The finding of responses to the prepulses alone by DBA/2NTac mice further indicates the advisability of routinely monitoring responses to prepulses alone. Unlike rats, DBA/2NCrl mice did not have greater startle amplitudes during the dark phase. Compound efficacy was better during the light phase because of poorer PPI in the vehicle group. Some vehicles may have unacceptable effects on PPI in DBA/2NCrl mice and may not be appropriate for studies evaluating novel compounds.


Subject(s)
Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Lighting , Piperazines/pharmacology , Quinolones/pharmacology , Reflex, Startle/drug effects , 2-Hydroxypropyl-beta-cyclodextrin , Acoustic Stimulation , Animals , Aripiprazole , Circadian Rhythm , Excipients/pharmacology , Male , Mice , Mice, Inbred DBA , Olanzapine , Species Specificity , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...