Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Behav Pharmacol ; 28(2 and 3-Spec Issue): 199-206, 2017 04.
Article in English | MEDLINE | ID: mdl-28234659

ABSTRACT

Agitation associated with dementia is frequently reported clinically but has received little attention in preclinical models of dementia. The current study used a 7PA2 CM intracerebroventricular injection model of Alzheimer's disease (AD) to assess acute memory impairment, and a bilateral intrahippocampal (IH) injection model of AD (aggregated Aß1-42 injections) and a bilateral IH injection model of dementia with Lewy bodies (aggregated NAC61-95 injections) to assess chronic memory impairment in the rat. An alternating-lever cyclic-ratio schedule of operant responding was used for data collection, where incorrect lever perseverations measured executive function (memory) and running response rates (RRR) measured behavioral output (agitation). The results indicate that bilateral IH injections of Aß1-42 and bilateral IH injections of NAC61-95 decreased memory function and increased RRRs, whereas intracerebroventricular injections of 7PA2 CM decreased memory function but did not increase RRRs. These findings show that using the aggregated peptide IH injection models of dementia to induce chronic neurotoxicity, memory decline was accompanied by elevated behavioral output. This demonstrates that IH peptide injection models of dementia provide a preclinical screen for pharmacological interventions used in the treatment of increased behavioral output (agitation), which also establish detrimental side effects on memory.


Subject(s)
Alzheimer Disease/physiopathology , Lewy Body Disease/physiopathology , Memory Disorders/physiopathology , Psychomotor Agitation/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Behavior, Animal/physiology , Conditioning, Operant/physiology , Disease Models, Animal , Executive Function/physiology , Hippocampus , Injections, Intraventricular , Male , Peptide Fragments/toxicity , Rats , Rats, Sprague-Dawley , alpha-Synuclein/toxicity
2.
Neuropharmacology ; 100: 124-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26025659

ABSTRACT

Bexarotene has been reported to reduce brain amyloid-ß (Aß) levels and to improve cognitive function in transgenic mouse models of Alzheimer's disease (AD). Four groups failed to fully replicate the primary results but the original authors claimed overall support for the general conclusions. Because of its potential clinical importance, the current work studied the effects of bexarotene using two animal species and highly relevant paradigms. Rats were tested for the ability of bexarotene to prevent changes induced by an Aß challenge in the form intracerebroventricular (i.c.v) administration of 7PA2 conditioned medium (7PA2 CM) which contains high levels of Aß species. Bexarotene had no effect on the long-term potentiation of evoked extracellular field excitatory postsynaptic potentials induced by i.c.v. 7PA2 CM. It also had no effect following subcutaneous administration of 2, 5, 10 and 15 mg/kg on behavioral/cognitive impairment using an alternating-lever cyclic-ratio schedule of operant responding in the rat. The effects of bexarotene were further tested using the APPSwFILon, PSEN1*M146L*L286V transgenic mouse model of AD, starting at the time Aß deposits first begin to develop. Mice were sacrificed after 48 days of exposure to 100 mg bexarotene per day. No significant difference between test and control mice was found using a water-maze test, and no significant difference in the number of Aß deposits in cerebral cortex, using two different antibodies, was apparent. These results question the potential efficacy of bexarotene for AD treatment, even if instigated in the preclinical period prior to the onset of cognitive deficits reported for human AD. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , CA1 Region, Hippocampal/drug effects , Tetrahydronaphthalenes/administration & dosage , Alzheimer Disease/chemically induced , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/toxicity , Animals , Bexarotene , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , CHO Cells , Conditioning, Operant/drug effects , Cricetulus , Culture Media, Conditioned , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley
3.
Neuropharmacology ; 92: 170-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25637092

ABSTRACT

ß-amyloid1-42 (Aß1-42) is a major endogenous pathogen underlying the aetiology of Alzheimer's disease (AD). Recent evidence indicates that soluble Aß oligomers, rather than plaques, are the major cause of synaptic dysfunction and neurodegeneration. Small molecules that suppress Aß aggregation, reduce oligomer stability or promote off-pathway non-toxic oligomerization represent a promising alternative strategy for neuroprotection in AD. MRZ-99030 was recently identified as a dipeptide that modulates Aß1-42 aggregation by triggering a non-amyloidogenic aggregation pathway, thereby reducing the amount of intermediate toxic soluble oligomeric Aß species. The present study evaluated the relevance of these promising results with MRZ-99030 under pathophysiological conditions i.e. against the synaptotoxic effects of Aß oligomers on hippocampal long term potentiation (LTP) and two different memory tasks. Aß1-42 interferes with the glutamatergic system and with neuronal Ca(2+) signalling and abolishes the induction of LTP. Here we demonstrate that MRZ-99030 (100-500 nM) at a 10:1 stoichiometric excess to Aß clearly reversed the synaptotoxic effects of Aß1-42 oligomers on CA1-LTP in murine hippocampal slices. Co-application of MRZ-99030 also prevented the two-fold increase in resting Ca(2+) levels in pyramidal neuron dendrites and spines triggered by Aß1-42 oligomers. In anaesthetized rats, pre-administration of MRZ-99030 (50 mg/kg s.c.) protected against deficits in hippocampal LTP following i.c.v. injection of oligomeric Aß1-42. Furthermore, similar treatment significantly ameliorated cognitive deficits in an object recognition task and under an alternating lever cyclic ratio schedule after the i.c.v. application of Aß1-42 and 7PA2 conditioned medium, respectively. Altogether, these results demonstrate the potential therapeutic benefit of MRZ-99030 in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Cognition Disorders , Dipeptides/pharmacology , Dipeptides/therapeutic use , Long-Term Potentiation/drug effects , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Animals , Calcium/metabolism , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Conditioning, Operant/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , In Vitro Techniques , Injections, Intraventricular , Inositol/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Putamen/drug effects , Putamen/metabolism , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects
4.
Int J Neuropsychopharmacol ; 17(1): 117-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24103729

ABSTRACT

Prefibrillar assembly of amyloid-ß (Aß) is a major event underlying the development of neuropathology and dementia in Alzheimer's disease (AD). This study determined the neuroprotective properties of an orally bioavailable Aß synaptotoxicity inhibitor, SEN1576. Binding of SEN1576 to monomeric Aß 1-42 was measured using surface plasmon resonance. Thioflavin-T and MTT assays determined the ability of SEN1576 to block Aß 1-42-induced aggregation and reduction in cell viability, respectively. In vivo long-term potentiation (LTP) determined effects on synaptic toxicity induced by intracerebroventricular (i.c.v.) injection of cell-derived Aß oligomers. An operant behavioural schedule measured effects of oral administration following i.c.v. injection of Aß oligomers in normal rats. SEN1576 bound to monomeric Aß 1-42, protected neuronal cells exposed to Aß 1-42, reduced deficits in in vivo LTP and behaviour. SEN1576 exhibits the necessary features of a drug candidate for further development as a disease modifying treatment for the early stages of AD-like dementia.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Pyrimidines/pharmacology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Infusions, Intraventricular , Long-Term Potentiation/drug effects , Male , Neurons/drug effects , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Rats
5.
J Alzheimers Dis ; 37(1): 77-88, 2013.
Article in English | MEDLINE | ID: mdl-23780661

ABSTRACT

The current study examined behavioral and histological effects of amyloid-ß (Aß) protein precursor (AßPP) overexpression in transgenic (Tg) rats created using the same gene, mutation, and promoter as the Tg2576 mouse model of Alzheimer's disease (AD). Male Tg+ rats were bred with female wild-type rats to generate litters of hemizygous Tg+ and Tg- offspring. Tg+ rats and Tg- littermates were tested for memory deficits at 4, 8, and 12 months old using a water-maze procedure. There were no significant behavioral differences between Tg+ rats and Tg- littermates at 4 months old but there were significant differences at 8 and 12 months old, and in probe trials at 8 and 12 months old, the Tg+ rats spent significantly less time and covered less distance in the platform zone. Under acquisition of a fixed-consecutive number schedule at 3 months old, Tg- littermates demonstrated a longer latency to learning the response rule than Tg+ rats; while this might seem paradoxical, it is consistent with the role of overexpression of AßPP in learning. Histological analyses revealed activated astrocytes in brains of Tg+ rats but not Tg- littermates at 6 months old, and thioflavin-S positive staining in the hippocampus and cortex of 17-month old Tg+ rats but not Tg- littermates. Quantification of Aß load in the brain at 22 months indicated high levels of Aß38, Aß40, and Aß42 in the Tg+ rats. These data suggest this model might provide a valuable resource for AD research.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Cricetinae , Female , Humans , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Rats , Rats, Transgenic
6.
Neurobiol Aging ; 34(4): 1116-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23164690

ABSTRACT

Oligomers of beta-amyloid (Aß) are implicated in the early memory impairment seen in Alzheimer's disease before to the onset of discernable neurodegeneration. Here, the capacity of a novel orally bioavailable, central nervous system-penetrating small molecule 5-aryloxypyrimidine, SEN1500, to prevent cell-derived (7PA2 [conditioned medium] CM) Aß-induced deficits in synaptic plasticity and learned behavior was assessed. Biochemically, SEN1500 bound to Aß monomer and oligomers, produced a reduction in thioflavin-T fluorescence, and protected a neuronal cell line and primary cortical neurons exposed to synthetic soluble oligomeric Aß(1-42). Electrophysiologically, SEN1500 alleviated the in vitro depression of long-term potentiation induced by both synthetic Aß(1-42) and 7PA2 CM, and alleviated the in vivo depression of long-term potentiation induced by 7PA2 CM, after systemic administration. Behaviorally, oral administration of SEN1500 significantly reduced memory-related deficits in operant responding induced after intracerebroventricular injection of 7PA2 CM. SEN1500 reduced cytotoxicity, acute synaptotoxicity, and behavioral deterioration after in vitro and in vivo exposure to synthetic Aß and 7PA2 CM, and shows promise for development as a clinically viable disease-modifying Alzheimer's disease treatment.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Memory/drug effects , Pyrimidines/administration & dosage , Synaptic Transmission/drug effects , Administration, Oral , Alzheimer Disease/complications , Animals , Male , Memory Disorders/complications , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley
7.
J Alzheimers Dis ; 25(2): 219-29, 2011.
Article in English | MEDLINE | ID: mdl-21654060

ABSTRACT

Behavioral effects of a novel anti-inflammatory SEN1176 were investigated. This pyrrolo[3,2-e][1,2,4]triazolo[1,5-a]pyrimidine suppresses amyloid-ß (Aß)1-42-induced macrophage production of nitric oxide, TNF-α, IL-1ß, and IL-6 in a dose-dependent fashion, an activity profile consistent with SEN1176 being a neuroinflammation inhibitor. Using male Sprague-Dawley rats, SEN1176 was examined relative to detrimental behavioral effects induced following bilateral intrahippocampal (IH) injections of aggregated Aß1-42. The rats were trained to respond under an alternating-lever cyclic-ratio (ALCR) schedule of food reinforcement, enabling measurement of parameters of operant performance that reflect aspects of learning and memory. Under the ALCR schedule, orally administered SEN1176 at 5, 20, or 30 mg/kg was effective in reducing the behavioral deficit caused by bilateral IH aggregated Aß1-42 injections in a dose-related manner over a 90-day treatment period. SEN1176 at 20 and 30 mg/kg significantly reduced lever switching errors and, at doses of 5, 10, and 30 mg/kg, significantly reduced incorrect lever perseverations, indicating a reduction of the behavioral deficit induced as a result of inflammation following IH Aß1-42 injections. When treatment with SEN1176 was instigated 30 days after IH Aß1-42 injections, it resulted in progressive protection, and withdrawal of SEN1176 treatment 60 days after IH Aß1-42 injections revealed partial retention of the protective effect. SEN1176 also significantly reduced numbers of activated astrocytes adjacent to the aggregated Aß1-42 injection sites. These results indicate the potential of SEN1176 for alleviating chronic neuroinflammatory processes related to brain Aß deposition that affect learning and memory in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Anti-Inflammatory Agents/therapeutic use , Behavioral Symptoms/chemically induced , Behavioral Symptoms/drug therapy , Hippocampus/drug effects , Peptide Fragments/toxicity , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Behavior, Animal/drug effects , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Macrophages/chemistry , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Triazoles/pharmacokinetics , Triazoles/therapeutic use
8.
Nanomedicine (Lond) ; 5(9): 1447-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21128725

ABSTRACT

Quantification of nanoparticles in biological systems (i.e., cells, tissues and organs) is becoming a vital part of nanotoxicological and nanomedical fields. Dose is a key parameter when assessing behavior and any potential risk of nanomaterials. Various techniques for nanoparticle quantification in cells and tissues already exist but will need further development in order to make measurements reliable, reproducible and intercomparable between different techniques. Microscopy allows detection and location of nanoparticles in cells and has been used extensively in recent years to characterize nanoparticles and their pathways in living systems. Besides microscopical techniques (light microscopy and electron microscopy mainly), analytical techniques such as mass spectrometry, an established technique in trace element analysis, have been used in nanoparticle research. Other techniques require 'labeled' particles, fluorescently, radioactively or magnetically. However, these techniques lack spatial resolution and subcellular localization is not possible. To date, only electron microscopy offers the resolving power to determine accumulation of nanoparticles in cells due to its ability to image particles individually. So-called super-resolution light microscopy techniques are emerging to provide sufficient resolution on the light microscopy level to image or 'see' particles as individual particles. Nevertheless, all microscopy techniques require statistically sound sampling strategies in order to provide quantitative results. Stereology is a well-known sampling technique in various areas and, in combination with electron microscopy, proves highly successful with regard to quantification of nanoparticle uptake by cells.


Subject(s)
Microscopy/methods , Nanoparticles , Animals , Fractionation, Field Flow , Humans , Mass Spectrometry , Microscopy, Electron
9.
Behav Brain Res ; 210(1): 32-7, 2010 Jun 26.
Article in English | MEDLINE | ID: mdl-20138088

ABSTRACT

Clinically accessible compounds that arrest or reverse the effects of amyloid-beta (Abeta) on progressively developing behavioural symptomatology and neuropathology in Alzheimer's disease (AD) have yet to become available. However, a viable strategy may be to target and neutralise soluble Abeta oligomers, which have been shown to mediate synaptic dysfunction and to produce cognitive deficits in the intact organism. Inhibiting the aggregation of Abeta is therapeutically attractive, as Abeta aggregation is a pathological event and pharmacological interventions targeting this are likely to have a non-toxic profile. A behavioural assay, the alternating-lever cyclic-ratio schedule, was used to assess the effect of Abeta oligomers and the non-peptide small molecule RS-0406 in male Sprague-Dawley rats. RS-0406 has been shown to inhibit Abeta(1-42) fibrillogenesis and protect against Abeta(1-42)-induced cytotoxicity in primary hippocampal neurons. In the current study, RS-0406 ameliorated the adverse effects of secreted oligomers of human Abeta on behaviour and dose dependently reduced the behavioural effects of Abeta oligomers, with the highest dose, 10microM, maintaining behaviour approximately at control levels. This effect appeared to be central; peripheral confounds having been extensively investigated. This is the first published report on the effects of RS-0406 in vivo and indicates that RS-0406 has potential as a pharmacotherapeutic intervention for behavioural deficits seen in the early stages of AD, and possibly as an intervention in the development of AD neuropathology. Indeed, an analogue of RS-0406 that could be administered peripherally might be a realistic candidate for the clinical treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Diamines/pharmacology , Neuroprotective Agents/pharmacology , Pyridazines/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cell Line , Cognition Disorders/chemically induced , Diamines/administration & dosage , Diamines/chemistry , Dose-Response Relationship, Drug , Humans , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuropsychological Tests , Pyridazines/administration & dosage , Pyridazines/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
10.
Neurosci Res ; 66(2): 213-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19913060

ABSTRACT

Residues 61-95 of the non-amyloid component (NAC(61-95)) domain of alpha-synuclein are responsible for the aggregation and neurotoxicity of this protein. This study evaluated the effect of N((omega))-nitro-l-arginine methyl ester (l-NAME), a nitric oxide synthase inhibitor, on the behavior of rats bilaterally injected into the CA3 area of the dorsal hippocampus with aggregated NAC(61-95). Twenty-four male Sprague-Dawley rats were trained to respond under an alternating-lever cyclic-ratio (ALCR) operant schedule. When responding was stable, 12 rats were injected bilaterally into the CA3 area of the hippocampus with aggregated NAC(61-95) (5muicro per side; concentration 10(-4)M), the remaining 12 rats were similarly injected with sterile water (SW), six of the NAC(61-95) injected rats and six of the SW injected rats were treated for 90 days post-injections with 0.05mg/ml l-NAME in drinking water available ad libitum. Treatment with l-NAME alleviated NAC(61-95)-induced behavioral deficits, indicated through the measurement of lever switching errors and incorrect lever perseverations under the ALCR schedule, and reduced the number of activated astrocytes proximal to the injection sites.


Subject(s)
CA3 Region, Hippocampal/drug effects , Conditioning, Operant/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Peptide Fragments/pharmacology , alpha-Synuclein/pharmacology , Analysis of Variance , Animals , Astrocytes/drug effects , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
11.
Behav Brain Res ; 208(1): 274-7, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-19931572

ABSTRACT

Pre-aggregated non-amyloid component of alpha-synuclein, NAC(61-95), was injected bilaterally into the CA3 area of rat hippocampus and behaviour was assessed using an alternating-lever cyclic-ratio (ALCR) schedule of operant responding. Four groups of rats were used (n=6 per group), subgroups were treated orally with either ibuprofen (40mg/kg) or vehicle (10% sucrose) twice daily. Intrahippocampal injection of NAC(61-95) increased lever switching errors and numbers of activated astrocytes, and ibuprofen treatment alleviated these effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CA3 Region, Hippocampal/drug effects , Ibuprofen/therapeutic use , Mental Disorders/chemically induced , Mental Disorders/drug therapy , Peptide Fragments , alpha-Synuclein , Animals , Behavior, Animal/drug effects , CA3 Region, Hippocampal/pathology , Conditioning, Operant/drug effects , Ibuprofen/pharmacology , Male , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
12.
Appetite ; 53(3): 457-60, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19748538

ABSTRACT

Male Sprague-Dawley rats were fitted with two cannulae in the VTA and one cannula in the NTS for co-administration of the micro-opioid receptor agonist DAMGO in one site and the opioid antagonist naltrexone in the other. Injection of DAMGO into the VTA or the NTS stimulated feeding. The increase in food intake after DAMGO injection into the VTA was decreased following injection of naltrexone into the NTS. Furthermore, the increase in food intake after DAMGO injection into the NTS was decreased following injection of naltrexone into the VTA. These results suggest an opioid-mediated feeding association between the VTA and NTS.


Subject(s)
Eating/physiology , Solitary Nucleus/physiology , Ventral Tegmental Area/physiology , Animals , Eating/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Male , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Solitary Nucleus/drug effects , Ventral Tegmental Area/drug effects
13.
Prog Neuropsychopharmacol Biol Psychiatry ; 33(5): 782-90, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19393282

ABSTRACT

The subiculum is in a pivotal position governing the output of the hippocampal formation. Despite this, it is a rather under-explored and sometimes ignored structure. Here, we discuss recent data indicating that the subiculum participates in a wide range of neurocognitive functions and processes. Some of the functions of subiculum are relatively well-known-these include providing a relatively coarse representation of space and participating in, and supporting certain aspects of, memory (particularly in the dynamic bridging of temporal intervals). The subiculum also participates in a wide variety of other neurocognitive functions too, however. Much less well-known are roles for the subiculum, and particularly the ventral subiculum, in the response to fear, stress and anxiety, and in the generation of motivated behaviour (particularly the behaviour that underlies drug addiction and the response to reward). There is an emerging suggestion that the subiculum participates in the temporal control of behaviour. It is notable that these latter findings have emerged from a consideration of instrumental behaviour using operant techniques; it may well be the case that the use of the watermaze or similar spatial tasks to assess subicular function (on the presumption that its functions are very similar to the hippocampus proper) has obscured rather than revealed neurocognitive functions of subiculum. The anatomy of subiculum suggests it participates in a rather subtle fashion in a very broad range of functions, rather than in a relatively more isolated fashion in a narrower range of functions, as might be the case for "earlier" components of hippocampal circuitry, such as the CA1 and CA3 subfields. Overall, there appears to a strong dorso-ventral segregation of function within subiculum, with the dorsal subiculum relatively more concerned with space and memory, and the ventral hippocampus concerned with stress, anxiety and reward. Finally, it may be the case that the whole subiculum participates in the temporal control of reinforced behaviour, although further experimentation is required to clarify this hypothesis.


Subject(s)
Hippocampus/physiology , Motivation , Space Perception/physiology , Spatial Behavior/physiology , Animals , Humans , Memory , Nerve Net/physiology , Neuronal Plasticity/physiology , Time Factors
14.
Behav Brain Res ; 193(2): 230-4, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18585407

ABSTRACT

Converging lines of evidence suggest that oligomers of amyloid-beta play a role in the cognitive impairment characteristic of Alzheimer's disease, but only three studies have provided experimental evidence of such impairment. To provide additional information about the effects of these oligomers on memory, the present study examined the memory of groups of rats exposed to ICV injections of the culture media (CM) of Chinese Hamster Ovary cells that were (7PA2) and were not (CHO-) transfected with a human mutation of amyloid precursor protein that appears to cause early-onset Alzheimer's disease. The 7PA2 CM, which contained concentrations of soluble amyloid-beta oligomers physiologically relevant to those found in human brain, significantly disrupted working memory in rats tested in a radial-arm maze. In contrast, CHO- CM, which did not contain such oligomers, had no effect on memory. The disruptive effects of 7PA2-derived amyloid-beta oligomers, evident 2h after exposure, disappeared within a day. These findings are compared to results from 7PA2 CM tested under a complex procedure thought to measure aspects of executive function. The results confirm the disruptive effects of low-n amyloid-beta oligomers and extend them to a well-established rat model of memory.


Subject(s)
Amyloid beta-Peptides/toxicity , Behavior, Animal/drug effects , Maze Learning/drug effects , Memory/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal/physiology , Blotting, Western , Brain/drug effects , Brain/physiopathology , CHO Cells , Cricetinae , Cricetulus , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/toxicity , Female , Humans , Injections, Intraventricular , Male , Molecular Weight , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Rats , Rats, Sprague-Dawley , Transfection
15.
Physiol Behav ; 91(2-3): 299-303, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17445845

ABSTRACT

This study examined the effects of thyroxine (T(4)) treatment on spatial learning and memory in congenitally hypothyroid (CH) rats. Forty CH male offspring of methimazole-treated dams were randomly divided into three groups: no T(4) (vehicle) treatment (n=12), T(4) treatment commencing on postnatal day (P-) 7 (n=14), and T(4) treatment commencing on P-21 (n=14). Normal male rats were used as a control group (n=14). T(4) was administered daily (sc, 0.02 microg/g) to the treatment groups for 30 days. A water-maze was used to assess behaviour at 42, 70 and 98 days of age. T(4) treatment beginning at P-7 improved learning and memory associated with CH at 70 and 98 days of age but T(4) treatment beginning at P-21 did not improve CH-impaired learning and memory.


Subject(s)
Congenital Hypothyroidism/therapy , Critical Period, Psychological , Hormone Replacement Therapy , Maze Learning/physiology , Thyroxine/therapeutic use , Age Factors , Analysis of Variance , Animals , Congenital Hypothyroidism/chemically induced , Congenital Hypothyroidism/physiopathology , Discrimination Learning/physiology , Disease Models, Animal , Drug Administration Schedule , Female , Male , Methimazole , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Space Perception/physiology , Thyroid Hormones/administration & dosage , Thyroid Hormones/physiology , Thyroxine/physiology
16.
Ann Neurol ; 60(6): 668-76, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17192927

ABSTRACT

OBJECTIVE: Despite progress in defining a pathogenic role for amyloid beta protein (Abeta) in Alzheimer's disease, orally bioavailable compounds that prevent its effects on hippocampal synaptic plasticity and cognitive function have not yet emerged. A particularly attractive therapeutic strategy is to selectively neutralize small, soluble Abeta oligomers that have recently been shown to mediate synaptic dysfunction. METHODS: Using electrophysiological, biochemical, and behavioral assays, we studied how scyllo-inositol (AZD-103; molecular weight, 180) neutralizes the acutely toxic effects of Abeta on synaptic function and memory recall. RESULTS: Scyllo-inositol, but not its stereoisomer, chiro-inositol, dose-dependently rescued long-term potentiation in mouse hippocampus from the inhibitory effects of soluble oligomers of cell-derived human Abeta. Cerebroventricular injection into rats of the soluble Abeta oligomers interfered with learned performance on a complex lever-pressing task, but administration of scyllo-inositol via the drinking water fully prevented oligomer-induced errors. INTERPRETATION: A small, orally available natural product penetrates into the brain in vivo to rescue the memory impairment produced by soluble Abeta oligomers through a mechanism that restores hippocampal synaptic plasticity.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Brain/drug effects , Inositol/pharmacology , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Injections, Intraventricular , Inositol/therapeutic use , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Neuroprotective Agents/therapeutic use , Organ Culture Techniques , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Rats , Stereoisomerism , Time Factors
17.
Brain Res ; 1029(1): 135-9, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15533326

ABSTRACT

The central nucleus of the amygdala (CeA) and the nucleus of the accumbens shell (NAc) have been shown to be involved in opioid-mediated feeding behavior. The present study examined whether mu-opioid signalling between the CeA and NAc affected feeding. Male Sprague-Dawley rats were fitted with one cannula placed in the CeA and two cannulae placed in the NAc, which allowed for coadministration of the mu-opioid receptor agonist [D-Ala(2), NMe-Phe(4), Gly-ol(5)]-enkephalin (DAMGO) in one site and the opioid antagonist naltrexone (NTX) in the other site. Single injection of DAMGO (2.4 nmol) into the CeA and bilateral injections of DAMGO (2.4 nmol) into the NAc stimulated feeding (P<0.05). The DAMGO-induced increase of food intake following injection into the CeA was decreased by bilateral injection of NTX (13.2 and 26.5 nmol) into the NAc at 2- and 4-h postinjections (P<0.05). In the reverse situation, the DAMGO-induced increase of food intake following injection into the NAc was decreased by injection of NTX (13.2 and 26.5 nmol) into the CeA at 1-, 2-, and 4-h postinjections (P<0.05). These results suggest that a bi-directional mu-opioid-opioid signalling pathway exists between the CeA and the NAc, which influences feeding.


Subject(s)
Amygdala/physiology , Feeding Behavior/physiology , Neural Pathways/physiology , Nucleus Accumbens/physiology , Receptors, Opioid, mu/physiology , Amygdala/drug effects , Animals , Eating/drug effects , Eating/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Feeding Behavior/drug effects , Male , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neural Pathways/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists
18.
Brain Res ; 991(1-2): 206-11, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14575893

ABSTRACT

The paraventricular nucleus (PVN) and the ventral tegmental area (VTA) have been shown to be involved in opioid mediated feeding behavior. The present study examined whether mu-opioid signalling between the PVN and VTA affected feeding behavior. Male Sprague-Dawley rats were cannulated with one cannula placed in the PVN and two cannulae placed in the VTA, which allowed for co-administration of the mu-opioid receptor agonist [D-Ala(2), NMe-Phe(4), Gly-ol(5)]-enkephalin (DAMGO) in one site and the opioid antagonist naltrexone (NTX) in the other site. Bilateral administration of DAMGO (1.2, 2.4 and 4.9 nmol) into the VTA stimulated feeding dose dependently at 2.4 and 4.9 nmol (P<0.05). The DAMGO (2.4 nmol)-induced increase of food intake following injection into the PVN was blocked by bilateral injection of NTX (6.6, 13.2 and 26.5 nmol) into the VTA at 2 and 4 h (P<0.05). In the reverse situation, the DAMGO (2.4 nmol)-induced increase of food intake following injection into the VTA was blocked by injection of NTX (13.2 and 26.5 nmol) into the PVN at 2 and 4 h (P<0.05). The present study suggests that a bidirectional mu-opioid-opioid signalling pathway exists between the PVN and the VTA which influences feeding.


Subject(s)
Feeding Behavior/physiology , Paraventricular Hypothalamic Nucleus/physiology , Ventral Tegmental Area/physiology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Feeding Behavior/drug effects , Injections, Intraventricular , Male , Naltrexone/administration & dosage , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/drug effects
19.
Neuroreport ; 13(4): 497-500, 2002 Mar 25.
Article in English | MEDLINE | ID: mdl-11930169

ABSTRACT

beta-Endorphin (beta-END) and alpha-melanocyte stimulating hormone (alpha-MSH), neuropeptides derived from proopiomelanocortin (POMC), have opposite effects on eating behavior. We injected rats with alpha-MSH (0.6 nmol) or beta-END (1 nmol) into the PVN (three times in a 26 h period). These doses of alpha-MSH and beta-END decreased and increased feeding respectively. Following alpha-MSH administration into the PVN, mRNA levels of POMC decreased by 17%, whereas there was no significant change in gene expression of either proDynorphin or proEnkephalin. PVN injection of beta-END failed to alter gene expression of POMC, proDynorphin or proEnkephalin. These data suggest that a feedback pathway exists between the PVN and ARC for alpha-MSH and POMC, but not for beta-END and POMC.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Gene Expression Regulation/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics , alpha-MSH/administration & dosage , beta-Endorphin/administration & dosage , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Eating/drug effects , Eating/genetics , Eating/physiology , Injections, Intraventricular , Male , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , alpha-MSH/physiology , beta-Endorphin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...