Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Mol Sci ; 24(13)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37446337

ABSTRACT

The introduction of anti-amyloid monoclonal antibodies against Alzheimer's disease (AD) is of high importance. However, even though treated patients show very little amyloid pathology, there is only a modest effect on the rate of cognitive decline. Although this effect can possibly increase over time, there is still a need for alternative treatments that will improve cognitive function in patients with AD. Therefore, the purpose of this study was to characterize the triazinetrione ACD856, a novel pan-Trk positive allosteric modulator, in multiple models to address its neuroprotective and potential disease-modifying effects. The pharmacological effect of ACD856 was tested in recombinant cell lines, primary cortical neurons, or animals. We demonstrate that ACD856 enhanced NGF-induced neurite outgrowth, increased the levels of the pre-synaptic protein SNAP25 in PC12 cells, and increased the degree of phosphorylated TrkB in SH-SY5Y cells. In primary cortical neurons, ACD856 led to increased levels of phospho-ERK1/2, showed a neuroprotective effect against amyloid-beta or energy-deprivation-induced neurotoxicity, and increased the levels of brain-derived neurotrophic factor (BDNF). Consequently, administration of ACD856 resulted in a significant increase in BDNF in the brains of 21 months old mice. Furthermore, repeated administration of ACD856 resulted in a sustained anti-depressant effect, which lasted up to seven days, suggesting effects that go beyond merely symptomatic effects. In conclusion, the results confirm ACD856 as a cognitive enhancer, but more importantly, they provide substantial in vitro and in vivo evidence of neuroprotective and long-term effects that contribute to neurotrophic support and increased neuroplasticity. Presumably, the described effects of ACD856 may improve cognition, increase resilience, and promote neurorestorative processes, thereby leading to a healthier brain in patients with AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Neuroblastoma , Neuroprotective Agents , Rats , Mice , Humans , Animals , Alzheimer Disease/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neuroblastoma/drug therapy , Amyloid beta-Peptides/metabolism , PC12 Cells , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology
2.
Paediatr Anaesth ; 29(11): 1122-1127, 2019 11.
Article in English | MEDLINE | ID: mdl-31536668

ABSTRACT

BACKGROUND: Advances in perioperative pediatric care have resulted in an increased number of procedures requiring anesthesia. During anesthesia and surgery, the patient is subjected to factors that affect the circulatory homeostasis, which can influence oxygenation of the brain. Near-infrared spectroscopy (NIRS) is an easy applicable noninvasive method for monitoring of regional tissue oxygenation (rScO2%). Alternate placements for NIRS have been investigated; however, no alternative cranial placements have been explored. AIM: To evaluate the agreement between frontal and occipital recordings of rScO2% in infants using INVOSTM during surgery and general anesthesia. METHOD: A standard frontal monitoring of rScO2% with NIRS was compared with occipital rScO2% measurements in fifteen children at an age <1 year, ASA 1-2, undergoing cleft lip and/or palate surgery during general anesthesia with sevoflurane. An agreement analysis was performed according to Bland and Altman. RESULTS: Mean values of frontal and occipital rScO2% at baseline were largely similar (70.7 ± 4.77% and 69.40 ± 5.04%, respectively). In the majority of the patients, the frontal and occipital recordings of rScO2 changed in parallel. There was a moderate positive correlation between frontal and occipital rScO2% INVOS™ readings (rho[ρ]: 0.513, P < .01). The difference between frontal and occipital rScO2 ranged from -31 to 28 with a mean difference (bias) of -0.15%. The 95% limit of agreement was -18.04%-17.74%. The error between frontal and occipital rScO2 recordings was 23%. CONCLUSION: The agreement between frontal and occipital recordings of brain rScO2% in infants using INVOSTM during surgery and general anesthesia was acceptable. In surgical procedures where the frontal region of the head is not available for monitoring, occipital recordings of rScO2% could be an option for monitoring.


Subject(s)
Anesthesia, General/methods , Frontal Lobe/metabolism , Occipital Lobe/metabolism , Oxygen/metabolism , Female , Humans , Infant , Male , Oxygen Consumption , Surgical Procedures, Operative/methods
3.
J Neurochem ; 125(3): 446-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23410232

ABSTRACT

Abnormal tau phosphorylation resulting in detachment of tau from microtubules and aggregation are critical events in neuronal dysfunction, degeneration, and neurofibrillary pathology seen in Alzheimer's disease. Glycogen synthase kinase-3ß (GSK3ß) is a key target for drug discovery in the treatment of Alzheimer's disease and related tauopathies because of its potential to abnormally phosphorylate proteins and contribute to synaptic degeneration. We report the discovery of AZD1080, a potent and selective GSK3 inhibitor that demonstrates peripheral target engagement in Phase 1 clinical studies. AZD1080 inhibits tau phosphorylation in cells expressing human tau and in intact rat brain. Interestingly, subchronic but not acute administration with AZD1080 reverses MK-801-induced deficits, measured by long-term potentiation in hippocampal slices and in a cognitive test in mice, suggesting that reversal of synaptic plasticity deficits in dysfunctional systems requires longer term modifications of proteins downstream of GSK3ß signaling. The inhibitory pattern on tau phosphorylation reveals a prolonged pharmacodynamic effect predicting less frequent dosing in humans. Consistent with the preclinical data, in multiple ascending dose studies in healthy volunteers, a prolonged suppression of glycogen synthase activity was observed in blood mononuclear cells providing evidence of peripheral target engagement with a selective GSK3 inhibitor in humans.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Long-Term Potentiation/drug effects , tau Proteins/metabolism , Animals , Cell Line, Transformed , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Crystallography , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Double-Blind Method , Electric Stimulation , Enzyme Inhibitors/chemistry , Excitatory Amino Acid Antagonists/toxicity , Excitatory Postsynaptic Potentials/drug effects , Glycogen Synthase/metabolism , Glycogen Synthase Kinase 3/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Humans , In Vitro Techniques , Indoles/pharmacology , Indoles/therapeutic use , Leukocytes, Mononuclear/drug effects , Long-Term Potentiation/physiology , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinases/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
4.
Front Mol Neurosci ; 16: 1279740, 2023.
Article in English | MEDLINE | ID: mdl-37908487

ABSTRACT

Recent clinical data with three therapeutic anti-Aß antibodies have demonstrated that removal of Aß-amyloid plaques in early Alzheimer's disease (AD) can attenuate disease progression. This ground-breaking progress in AD medicine has validated both the amyloid cascade hypothesis and Aß-amyloid as therapeutic targets. These results also strongly support therapeutic approaches that aim to reduce the production of amyloidogenic Aß to prevent the formation of Aß-pathology. One such strategy, so-called gamma-secretase modulators (GSM), has been thoroughly explored in preclinical settings but has yet to be fully tested in clinical trials. Recent scientific progress has shed new light on the role of Aß in Alzheimer's disease and suggests that GSMs exhibit specific pharmacological features that hold great promise for the prevention and treatment of Alzheimer's disease. In this short review, we discuss the data that support why it is important to continue to progress in this class of compounds.

5.
Psychopharmacology (Berl) ; 240(8): 1789-1804, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37394539

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is defined as a complex mental disorder which is characterized by a pervasive low mood and aversion to activity. Several types of neurotransmitter systems e.g. serotonergic, glutamatergic and noradrenergic systems have been suggested to play an important role in the origination of depression, but neurotrophins such as brain derived neurotrophic factor (BDNF) have also been implicated in the disease process. OBJECTIVES: The purpose of this study was to examine the effects of a newly developed class of molecules, characterized as positive allosteric modulators of neurotrophin/Trk receptor mediated signaling (Trk-PAM), on neurotransmitter release and depression-like behavior in vivo. METHODS: The effect of and possible interaction of neurotrophin/Trk signaling pathways with serotonergic and glutamatergic systems in the modulation of depression-related responses was studied using newly developed Trk-PAM compounds (ACD855, ACD856 and AC26845), as well as ketamine and fluoxetine in the forced swim test (FST) in rodents. Moreover, in vivo microdialysis in freely moving rats was used to assess changes in neurotransmitter levels in the rat. RESULTS: The results from the study show that several different compounds, which all potentiate Trk-receptor mediated signaling, display antidepressant-like activity in the FST. Moreover, the data also indicate that the effects of both fluoxetine and ketamine in the FST, both used in clinical practice, are mediated via BDNF/TrkB signaling, which could have implications for novel therapies in MDD. CONCLUSIONS: Trk-PAMs could provide an interesting avenue for the development of novel therapeutics in this area.


Subject(s)
Depressive Disorder, Major , Ketamine , Rats , Animals , Fluoxetine/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major/drug therapy , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Receptor, trkB/metabolism
6.
EMBO Mol Med ; 15(2): e16556, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36524456

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic form of familial small vessel disease; no preventive or curative therapy is available. CADASIL is caused by mutations in the NOTCH3 gene, resulting in a mutated NOTCH3 receptor, with aggregation of the NOTCH3 extracellular domain (ECD) around vascular smooth muscle cells. In this study, we have developed a novel active immunization therapy specifically targeting CADASIL-like aggregated NOTCH3 ECD. Immunizing CADASIL TgN3R182C150 mice with aggregates composed of CADASIL-R133C mutated and wild-type EGF1-5 repeats for a total of 4 months resulted in a marked reduction (38-48%) in NOTCH3 deposition around brain capillaries, increased microglia activation and lowered serum levels of NOTCH3 ECD. Active immunization did not impact body weight, general behavior, the number and integrity of vascular smooth muscle cells in the retina, neuronal survival, or inflammation or the renal system, suggesting that the therapy is tolerable. This is the first therapeutic study reporting a successful reduction of NOTCH3 accumulation in a CADASIL mouse model supporting further development towards clinical application for the benefit of CADASIL patients.


Subject(s)
CADASIL , Animals , Mice , Brain/metabolism , CADASIL/genetics , CADASIL/therapy , Capillaries/metabolism , Disease Models, Animal , Immunotherapy, Active , Mutation , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptors, Notch/metabolism
7.
Eur J Neurosci ; 36(9): 3165-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22958226

ABSTRACT

Neuropil deposition of beta-amyloid (Aß) peptides is believed to be a key event in the neurodegenerative process of Alzheimer's disease (AD). An early and consistent clinical finding in AD is olfactory dysfunction with associated pathology. Interestingly, transgenic amyloid precursor protein (Tg2576) mice also show early amyloid pathology in olfactory regions. Moreover, a recent study indicates that axonal transport is compromised in the olfactory system of Tg2576 mice, as measured by manganese-enhanced magnetic resonance imaging (MEMRI). Here we tested whether the putative axonal transport deficit in the Tg2576 mouse model improves in response to a selective gamma-secretase inhibitor, N-[cis-4-[(4-chlorophenyl)-sulfonyl]-4-(2,5-difluorophenyl)cyclohexyl]-1,1,1-trifluoromethanesulfonamide (MRK-560). Tg2576 mice or wild-type (WT) littermates were treated daily with MRK-560 (30 µmol/kg) or vehicle for 4 (acute) or 29 days (chronic). The subsequent MEMRI analysis revealed a distinct axonal transport dysfunction in the Tg2576 mice compared with its littermate controls. Interestingly, the impairment of axonal transport could be fully reversed by chronic administration of MRK-560, in line with the significantly lowered levels of both soluble and insoluble forms of Aß found in the brain and olfactory bulbs (OBs) following treatment. However, no improvement of axonal transport was observed after acute treatment with MRK-560, where soluble but not insoluble forms of Aß were reduced in the brain and OBs. The present results show that axonal transport is impaired in Tg2576 mice compared with WT controls, as measured by MEMRI. Chronic treatment in vivo with a gamma-secretase inhibitor, MRK-560, significantly reduces soluble and insoluble forms of Aß, and fully reverses the axonal transport dysfunction.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Axonal Transport/drug effects , Sulfonamides/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Axonal Transport/genetics , Brain/metabolism , Magnetic Resonance Imaging , Manganese , Mice , Mice, Transgenic , Olfactory Bulb/metabolism
8.
Drug Discov Today ; 27(10): 103318, 2022 10.
Article in English | MEDLINE | ID: mdl-35850433

ABSTRACT

Neurotrophins, such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), are small proteins expressed in the brain and peripheral tissues, which regulate several key aspects of neuronal function, including neurogenesis, synaptic plasticity and neuroprotection, but also programmed cell death. This broad range of effects is a result of a complex downstream signaling pathway, with differential spatial and temporal activation patterns further diversifying their physiological effects. Alterations in neurotrophin levels, or known polymorphisms in neurotrophin genes, have been linked to a variety of disorders, including depression and Alzheimer's disease (AD). Historically, their therapeutic potential in these disorders has been hampered by the lack of suitable tool molecules for clinical studies. However, recent advancements have led to the development of new therapeutic candidates, which are now in clinical testing.


Subject(s)
Brain-Derived Neurotrophic Factor , Nerve Growth Factor , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Receptors, Nerve Growth Factor/genetics , Signal Transduction/physiology
9.
Cells ; 10(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34440640

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and results in severe neurodegeneration and progressive cognitive decline. Neurotrophins are growth factors involved in the development and survival of neurons, but also in underlying mechanisms for memory formation such as hippocampal long-term potentiation. Our aim was to identify small molecules with stimulatory effects on the signaling of two neurotrophins, the nerve growth factor (NGF) and the brain derived neurotrophic factor (BDNF). To identify molecules that could potentiate neurotrophin signaling, 25,000 molecules were screened, which led to the identification of the triazinetrione derivatives ACD855 (Ponazuril) and later on ACD856, as positive allosteric modulators of tropomyosin related kinase (Trk) receptors. ACD855 or ACD856 potentiated the cellular signaling of the neurotrophin receptors with EC50 values of 1.9 and 3.2 or 0.38 and 0.30 µM, respectively, for TrkA or TrkB. ACD855 increased acetylcholine levels in the hippocampus by 40% and facilitated long term potentiation in rat brain slices. The compounds acted as cognitive enhancers in a TrkB-dependent manner in several different behavioral models. Finally, the age-induced cognitive dysfunction in 18-month-old mice could be restored to the same level as found in 2-month-old mice after a single treatment of ACD856. We have identified a novel mechanism to modulate the activity of the Trk-receptors. The identification of the positive allosteric modulators of the Trk-receptors might have implications for the treatment of Alzheimer's diseases and other diseases characterized by cognitive impairment.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Nootropic Agents/pharmacology , Receptors, Nerve Growth Factor/agonists , Age Factors , Animals , Brain/enzymology , Brain/physiopathology , Cell Line, Tumor , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Membrane Glycoproteins , Mice, Inbred C57BL , Motor Activity/drug effects , Protein-Tyrosine Kinases , Rats, Sprague-Dawley , Receptor, trkA/agonists , Receptor, trkA/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Small Molecule Libraries , Triazines/pharmacology
10.
Hippocampus ; 19(12): 1187-98, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19309036

ABSTRACT

Cholinergic and GABAergic neurons in the medial septum/vertical limb of the diagonal band of Broca (MS/vDB) projecting to the hippocampus, constitute the septohippocampal projection, which is important for hippocampal-dependent learning and memory. There is also evidence for an extrinsic as well as an intrinsic glutamatergic network within the MS/vDB. GABAergic and cholinergic septohippocampal neurons express the serotonergic 5-HT(1A) receptor and most likely also glutamatergic NMDA receptors. The aim of the present study was to examine whether septal 5-HT(1A) receptors are important for hippocampal-dependent long-term memory and whether these receptors interact with glutamatergic NMDA receptor transmission in a manner important for hippocampal-dependent spatial memory. Intraseptal infusion of the 5-HT(1A) receptor agonist (R)-8-OH-DPAT (1 or 4 microg/rat) did not affect spatial learning in the water maze task but impaired emotional memory in the passive avoidance task at the higher dose tested (4 microg/rat). While intraseptal administration of (R)-8-OH-DPAT (4 microg) combined with a subthreshold dose of the NMDA receptor antagonist D-AP5 (1 microg) only marginally affected spatial acquisition, it produced a profound impairment in spatial memory. In conclusion, septal 5-HT(1A) receptors appears to play a more prominent role in emotional than in spatial memory. Importantly, septal 5-HT(1A) and NMDA receptors appear to interact in a manner, which is particularly critical for the expression or retrieval of hippocampal-dependent long-term spatial memory. It is proposed that NMDA receptor hypofunction in the septal area may unmask a negative effect of 5-HT(1A) receptor activation on memory, which may be clinically relevant.


Subject(s)
Hippocampus/physiology , Learning/physiology , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Septum of Brain/physiology , Space Perception/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Dose-Response Relationship, Drug , Emotions/drug effects , Emotions/physiology , Excitatory Amino Acid Antagonists/pharmacology , Learning/drug effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Septum of Brain/drug effects , Serotonin 5-HT1 Receptor Agonists , Serotonin Receptor Agonists/administration & dosage , Serotonin Receptor Agonists/pharmacology , Space Perception/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors
11.
Learn Mem ; 15(9): 625-32, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18772249

ABSTRACT

Two experiments were conducted to investigate the possibility of faster forgetting by PDAPP mice (a well-established model of Alzheimer's disease as reported by Games and colleagues in an earlier paper). Experiment 1, using mice aged 13-16 mo, confirmed the presence of a deficit in a spatial reference memory task in the water maze by hemizygous PDAPP mice relative to littermate controls. However, after overtraining to a criterion of equivalent navigational performance, a series of memory retention tests revealed faster forgetting in the PDAPP group. Very limited retraining was sufficient to reinstate good memory in both groups, indicating that their faster forgetting may be due to retrieval failure rather than trace decay. In Experiment 2, 6-mo-old PDAPP and controls were required to learn each of a series of spatial locations to criterion with their memory assessed 10 min after learning each location. No memory deficit was apparent in the PDAPP mice initially, but a deficit built up through the series of locations suggestive of increased sensitivity to interference. Faster forgetting and increased interference may each reflect a difficulty in accessing memory traces. This interpretation of one aspect of the cognitive deficit in human mutant APP mice has parallels to deficits observed in patients with Alzheimer's disease, further supporting the validity of transgenic models of the disease.


Subject(s)
Alzheimer Disease/physiopathology , Memory/physiology , Spatial Behavior/physiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Disease Models, Animal , Humans , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Transgenic
12.
Eur Neuropsychopharmacol ; 27(12): 1298-1307, 2017 12.
Article in English | MEDLINE | ID: mdl-29102248

ABSTRACT

The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.


Subject(s)
Avoidance Learning/physiology , Opioid Peptides/metabolism , Receptors, Opioid/deficiency , Animals , Association Learning/drug effects , Avoidance Learning/drug effects , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Retention, Psychology/drug effects , Spiro Compounds/pharmacology , Nociceptin Receptor , Nociceptin
13.
Neuropharmacology ; 123: 233-241, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28623171

ABSTRACT

Although the muscarinic receptor family has long been a source of potentially compelling targets for small molecule drug discovery, it was difficult to achieve agonist selectivity within the family. A new class of M1 muscarinic agonists has emerged, and these compounds have been characterized as agonists that activate the receptor at an allosteric site. Members of this class of M1 agonists have been shown to be selective across the muscarinic receptors. However, upon introduction of a novel pharmacologic mechanism, it is prudent to ensure that no new off-target activities have arisen, particularly within the context of in vivo experiments. Reported here, is the in vitro and in vivo characterization of a novel M1 agonist tool compound, PPBI, and demonstrations that the primary biological effects of PPBI are mediated through M1. PPBI reverses d-amphetamine locomotor activity, but fails to do so in transgenic mice that do not express M1. PPBI also reverses a natural deficit in a rat cognition model at a level of exposure which also activates cortical circuitry. Most notably, PPBI is analgesic in a variety of rat and mouse models and the analgesic effect of PPBI is reversed by an M1-preferring antagonist and an M1-selective toxin. Finally, the pharmacokinetic/pharmacodynamic measures of PPBI are compared across multiple endpoints which highlights that activity in models of psychosis and pain require higher exposures than that required in the cognition model.


Subject(s)
Analgesics/pharmacology , Benzimidazoles/pharmacology , Muscarinic Agonists/pharmacology , Nootropic Agents/pharmacology , Piperidines/pharmacology , Pyrrolidines/pharmacology , Receptor, Muscarinic M1/agonists , Amphetamine/pharmacology , Analgesics/chemistry , Analgesics/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , CHO Cells , Central Nervous System Stimulants/pharmacology , Cognition/drug effects , Cricetulus , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Motor Activity/drug effects , Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacokinetics , Nootropic Agents/chemistry , Nootropic Agents/pharmacokinetics , Pain/drug therapy , Random Allocation , Rats, Sprague-Dawley , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism , Transfection
14.
Behav Brain Res ; 166(2): 204-10, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16159675

ABSTRACT

The pyramidal CA1 neurons of the hippocampus are critically involved in spatial learning and memory. These neurons are especially vulnerable to cerebral ischemia, but in spite of this, it has been consistently difficult to show any learning and memory deficits in two-vessel occlusion models of global ischemia. Transient global ischemia was induced in adult male rats under general anaesthesia administered by artificial respiration to prevent respiratory arrest. Systemic blood pressure was reduced to below 50 mmHg by instant adjustments of the halothane concentration, before and during bilateral occlusion of the carotid arteries. Cerebral blood flow was monitored by laser-Doppler flowmetry. Dying neurons were detected by TUNEL at 14 days after ischemia and surviving neurons by NeuN at 14 and 125 days after ischemia. Learning and memory was assessed in a novel water maze with three successive left-right choices. Transient global ischemia produced a profound and selective degeneration of CA1 neurons at 14 days after ischemia. This degeneration was associated with severe impairments in learning at 13 days after ischemia and in memory, as tested 24 h afterwards. At 125 days after ischemia, there was no significant learning and memory impairment, whereas the number of CA1 neurons was increased. These results show that transient global ischemia induced by two-vessel occlusion may lead to severe, but transient, impairments in learning and memory using a novel water maze, and that restored learning and memory is associated with an increased number of CA1 neurons.


Subject(s)
Brain Ischemia/complications , Learning Disabilities/etiology , Maze Learning/physiology , Memory Disorders/etiology , Analysis of Variance , Animals , Behavior, Animal , Body Weight/physiology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death/physiology , Cerebrovascular Circulation/physiology , Functional Laterality/physiology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , Laser-Doppler Flowmetry/methods , Learning Disabilities/metabolism , Learning Disabilities/pathology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
15.
J Cereb Blood Flow Metab ; 25(12): 1586-95, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15917746

ABSTRACT

The pyramidal neurons of the hippocampal CA1 region are essential for cognitive functions such as spatial learning and memory, and are selectively destroyed after cerebral ischemia. To analyze whether degenerated CA1 neurons are replaced by new neurons and whether such regeneration is associated with amelioration in learning and memory deficits, we have used a rat global ischemia model that provides an almost complete disappearance (to approximately 3% of control) of CA1 neurons associated with a robust impairment in spatial learning and memory at two weeks after ischemia. We found that transient cerebral ischemia can evoke a massive formation of new neurons in the CA1 region, reaching approximately 40% of the original number of neurons at 90 days after ischemia (DAI). Co-localization of the mature neuronal marker neuronal nuclei with 5-bromo-2'-deoxyuridine in CA1 confirmed that neurogenesis indeed had occurred after the ischemic insult. Furthermore, we found increased numbers of cells expressing the immature neuron marker polysialic acid neuronal cell adhesion molecule in the adjacent lateral periventricular region, suggesting that the newly formed neurons derive from this region. The reappearance of CA1 neurons was associated with a recovery of ischemia-induced impairments in spatial learning and memory at 90 DAI, suggesting that the newly formed CA1 neurons restore hippocampal CA1 function. In conclusion, these results show that the brain has an endogenous capacity to form new nerve cells after injury, which correlates with a restoration of cognitive functions of the brain.


Subject(s)
Hippocampus/cytology , Ischemic Attack, Transient/physiopathology , Nerve Regeneration/physiology , Pyramidal Cells/cytology , Recovery of Function/physiology , Animals , Cell Division/physiology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Hippocampus/physiology , Ischemic Attack, Transient/pathology , Lateral Ventricles/cytology , Male , Maze Learning/physiology , Memory/physiology , Memory Disorders/pathology , Memory Disorders/physiopathology , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Space Perception/physiology
16.
Neuropsychopharmacology ; 28(9): 1642-55, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12838273

ABSTRACT

The present study examined the role of the 5-HT1B receptor in learning and memory. The ability of the 5-HT1B receptor agonist anpirtoline and the selective 5-HT1B receptor antagonist NAS-181 to affect spatial learning in the water maze (WM) and aversive learning in the passive avoidance (PA) task were examined in the rat. Anpirtoline (0.1-1.0 mg/kg, s.c.) caused a dose-dependent impairment of learning and memory in both the WM and PA tasks. NAS-181 (1.0-10 mg/kg, s.c.) failed to alter performance of the WM task, but produced a dose-dependent (0.1-20 mg/kg) facilitation of PA retention. Furthermore, treatment with NAS-181 (10 mg/kg) fully blocked the impairment of the WM and PA performance caused by anpirtoline (1.0 mg/kg). In contrast, NAS-181 (3.0-10 mg/kg) did not attenuate the spatial learning deficit and the impairment of PA retention caused by scopolamine (0.1 mg/kg in WM task, 0.3 mg/kg in PA task, s.c.), a nonselective muscarinic antagonist. Moreover, a subthreshold dose of scopolamine (0.1 mg/kg) blocked the facilitation of PA retention induced by NAS-181 (1.0-10 mg/kg). In addition, the behavioral disturbances (eg thigmotaxic swimming and platform deflections) induced by anpirtoline and scopolamine were analyzed in the WM task and correlated with WM performance. These results indicate that: (1) 5-HT1B receptor stimulation and blockade result in opposite effects in two types of cognitive tasks in the rat, and that (2) the 5-HT1B antagonist NAS-181 can facilitate some aspects of cognitive function, most likely via an increase of cholinergic transmission. These results suggest that 5-HT1B receptor antagonists may have a potential in the treatment of cognitive deficits resulting from loss of cholinergic transmission.


Subject(s)
Avoidance Learning/physiology , Receptors, Serotonin/physiology , Space Perception/physiology , Animals , Behavior, Animal , Benzopyrans/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Escape Reaction/drug effects , Male , Maze Learning/drug effects , Morpholines/pharmacology , Motor Activity/drug effects , Muscarinic Antagonists/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B , Receptors, Serotonin/drug effects , Retention, Psychology/drug effects , Scopolamine/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Swimming
17.
Br J Pharmacol ; 141(1): 132-40, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14662736

ABSTRACT

1. The ORL1 agonists nociceptin and Ro 64-6198 were compared in their ability to modify spontaneous locomotor activity in male NMRI mice not habituated to the test environment. 2. Higher doses of nociceptin (>5 nmol i.c.v.) reduced whereas lower doses (<1 nmol i.c.v.) stimulated locomotor activity. Both effects were blocked by the putative ORL1 antagonists [NPhe1]nociceptin(1-13)NH2 (10 nmol i.c.v.) and UFP101 (10 nmol, i.c.v.). The effects were also blocked by naloxone benzoylhydrazone (1 mg x kg(-1) s.c.), but not by the nonselective opioid antagonist naloxone (1 mg x kg(-1) s.c.). 3 In contrast to nociceptin, the synthetic ORL1 agonist Ro 64-6198 (0.01-1.0 mg x kg(-1) i.p.) produced monophasic inhibition of locomotor activity, which was insensitive to the treatment with [NPhe1]nociceptin(1-13)NH2 or naloxone benzoylhydrazone. Treatment with UFP101 abolished the locomotor inhibition induced by Ro 64-6198 (1.0 mg x kg(-1)), whereas naloxone (1.0 mg x kg(-1), s.c.) further increased the locomotor-inhibitory effects. 4. Naloxone benzoylhydrazone (0.3; 1.0 and 3.0 mg x kg(-1) s.c.) increased locomotor activity, although the effect was statistically significant only with the highest dose used. 5. Pretreatment with the tyrosine hydroxylase inhibitor H44-68 totally eliminated the motor-stimulatory effects of low doses of nociceptin, probably via dopamine depletion. 6. The results suggest that nociceptin stimulates locomotor activity at low doses if dopamine activity is intact. High doses of nociceptin and all the tested doses of Ro 64-6198 seem to interact with a functionally different subset of ORL1 receptors. In addition, the effects of Ro 64-6198 are modulated by tonic opioid receptor activity.


Subject(s)
Motor Activity/physiology , Naloxone/analogs & derivatives , Receptors, Opioid/physiology , Animals , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/physiology , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Imidazoles/administration & dosage , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacokinetics , Injections, Intraperitoneal , Injections, Intraventricular , Injections, Subcutaneous , Male , Methods , Methyltyrosines/administration & dosage , Methyltyrosines/pharmacokinetics , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Naloxone/administration & dosage , Naloxone/pharmacokinetics , Narcotic Antagonists , Opioid Peptides/administration & dosage , Opioid Peptides/antagonists & inhibitors , Opioid Peptides/chemistry , Opioid Peptides/pharmacokinetics , Receptors, Opioid/administration & dosage , Spiro Compounds/administration & dosage , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacokinetics , Tyrosine 3-Monooxygenase/administration & dosage , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/pharmacokinetics , Nociceptin Receptor , Nociceptin
18.
Brain Res ; 997(2): 222-33, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14706874

ABSTRACT

The endogenous peptide nociceptin (orphanin FQ) plays a role in several important physiological functions in the CNS such as pain, anxiety and locomotion. It has previously been found that injection of 10 nmol nociceptin into the CA3 region of the hippocampus markedly impairs spatial learning and memory in the rat. The present study examined the effects of lower doses of nociceptin (3.3, 1, 0.33 and 0.1 nmol/rat) on spatial learning. The 3.3 nmol dose impaired spatial learning over the 5 days of training although the effect was not as strong as with 10 nmol. In contrast, the two lower doses, 1 and 0.33 nmol/rat, improved spatial learning whereas the lowest dose, 0.1 nmol/rat, had no significant effect. Both the impairing and facilitating effect of nociceptin could be blocked by an ORL-1 receptor antagonist, [Phe1Psi(CH(2)-NH)Gly2]NC(1-13)NH2 (10 nmol/rat), indicating that both effects are ORL-1 receptor-mediated. The 3.3 nmol dose of nociceptin did not impair the performance in the visual platform task and did not alter swim speed or motor activity indicating no effects on motivation or motor performance. Taken together, these results show that nociceptin has a biphasic dose-effect curve and provide further evidence for a role of this neuropeptide in cognitive processes in the hippocampus.


Subject(s)
Hippocampus/physiology , Learning/physiology , Opioid Peptides/physiology , Receptors, Opioid/physiology , Spatial Behavior/physiology , Animals , Male , Narcotic Antagonists , Opioid Peptides/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Nociceptin Receptor , Nociceptin
19.
Front Pharmacol ; 5: 102, 2014.
Article in English | MEDLINE | ID: mdl-24847271

ABSTRACT

Alzheimer's Disease (AD) is the most common form of dementia, affecting approximately 36 million people worldwide. To date there is no preventive or curative treatment available for AD, and in absence of major progress in therapeutic development, AD manifests a concrete socioeconomic threat. The awareness of the growing problem of AD is increasing, exemplified by the recent G8 Dementia Summit, a meeting held in order to set the stage and steer the compass for the future. Simultaneously, and paradoxically, we have seen key players in the pharmaceutical industry that have recently closed or significantly decreased their R&D spending on AD and other CNS disorders. Given the pressing need for new treatments in this area, other actors need to step-in and enter this drug discovery arena complementing the industrial efforts, in order to turn biological and technological progress into novel therapeutics. In this article, we present an example of a novel drug discovery initiative that in a non-profit setting, aims to integrate with both preclinical and clinical academic groups and pharmaceutical industry to explore the therapeutic potential of new concepts in patients, using novel biology, state of the art technologies and rapid concept testing.

20.
J Alzheimers Dis ; 26(1): 143-55, 2011.
Article in English | MEDLINE | ID: mdl-21593569

ABSTRACT

Similar to patients with Alzheimer's disease (AD), dogs exhibit age-dependent cognitive decline, amyloid-ß (Aß) pathology, and evidence of cholinergic hypofunction. The present study sought to further investigate the role of cholinergic hypofunction in the canine model by examining the effect of the cholinesterase inhibitors phenserine and donepezil on performance of two tasks, a delayed non-matching-to-position task (DNMP) designed to assess working memory, and an oddity discrimination learning task designed to assess complex learning, in aged dogs. Phenserine (0.5 mg/kg; PO) significantly improved performance on the DNMP at the longest delay compared to wash-out and partially attenuated scopolamine-induced deficits (15 µg/kg; SC). Phenserine also improved learning on a difficult version of an oddity discrimination task compared to placebo, but had no effect on an easier version. We also examined the effects of three doses of donepezil (0.75, 1.5, and 6 mg/kg; PO) on performance of the DNMP. Similar to the results with phenserine, 1.5 mg/kg of donepezil improved performance at the longest delay compared to baseline and wash-out, indicative of memory enhancement. These results further extend the findings of cholinergic hypofunction in aged dogs and provide pharmacological validation of the canine model with a cholinesterase inhibitor approved for use in AD. Collectively, these studies support utilizing the aged dog in future screening of therapeutics for AD, as well as for investigating the links among cholinergic function, Aß pathology, and cognitive decline.


Subject(s)
Aging , Cholinesterase Inhibitors/therapeutic use , Indans/therapeutic use , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Physostigmine/analogs & derivatives , Piperidines/therapeutic use , Analysis of Variance , Animals , Behavior, Animal/drug effects , Cholinergic Antagonists/toxicity , Discrimination, Psychological/drug effects , Disease Models, Animal , Dogs , Donepezil , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Learning Disabilities/chemically induced , Male , Memory Disorders/chemically induced , Neuropsychological Tests , Odorants , Physostigmine/therapeutic use , Scopolamine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL