Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Transl Stroke Res ; 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37326791

ABSTRACT

Repulsive guidance molecule A (RGMa) is an inhibitor of neuronal growth and survival which is upregulated in the damaged central nervous system following acute spinal cord injury (SCI), traumatic brain injury, acute ischemic stroke (AIS), and other neuropathological conditions. Neutralization of RGMa is neuroprotective and promotes neuroplasticity in several preclinical models of neurodegeneration and injury including multiple sclerosis, AIS, and SCI. Given the limitations of current treatments for AIS due to narrow time windows to intervention (TTI), and restrictive patient selection criteria, there is significant unmet need for therapeutic agents that enable tissue survival and repair following acute ischemic damage for a broader population of stroke patients. In this preclinical study, we evaluated whether elezanumab, a human anti-RGMa monoclonal antibody, could improve neuromotor function and modulate neuroinflammatory cell activation following AIS with delayed intervention times up to 24 h using a rabbit embolic permanent middle cerebral artery occlusion model (pMCAO). In two replicate 28-day pMCAO studies, weekly intravenous infusions of elezanumab, over a range of doses and TTIs of 6 and 24 h after stroke, significantly improved neuromotor function in both pMCAO studies when first administered 6 h after stroke. All elezanumab treatment groups, including the 24 h TTI group, had significantly less neuroinflammation as assessed by microglial and astrocyte activation. The novel mechanism of action and potential for expanding TTI in human AIS make elezanumab distinct from current acute reperfusion therapies, and support evaluation in clinical trials of acute CNS damage to determine optimal dose and TTI in humans. A: Ramified/resting astrocytes and microglia in a normal, uninjured rabbit brain. B: Rabbit pMCAO brain illustrating lesion on right side of brain (red), surrounded by penumbra (pink) during acute phase post stroke, with minimal injury to left brain hemisphere. Penumbra characterized by activated astrocytes and microglia (region in crosshair within circle), with upregulation of free and bound RGMa. C: Elezanumab binds to both free and bound RGMa, preventing full activation of astrocytes and microglia. D: Elezanumab is efficacious in rabbit pMCAO with a 4 × larger TTI window vs. tPA (6 vs. 1.5 h, respectively). In human AIS, tPA is approved for a TTI of 3-4.5 h. Elezanumab is currently being evaluated in a clinical Ph2 study of AIS to determine the optimal dose and TTI (NCT04309474).

2.
Adv Drug Deliv Rev ; 173: 20-59, 2021 06.
Article in English | MEDLINE | ID: mdl-33705875

ABSTRACT

Initially thought to be useful only to reach tissues in the immediate vicinity of the CSF circulatory system, CSF circulation is now increasingly viewed as a viable pathway to deliver certain therapeutics deeper into brain tissues. There is emerging evidence that this goal is achievable in the case of large therapeutic proteins, provided conditions are met that are described herein. We show how fluid dynamic modeling helps predict infusion rate and duration to overcome high CSF turnover. We posit that despite model limitations and controversies, fluid dynamic models, pharmacokinetic models, preclinical testing, and a qualitative understanding of the glymphatic system circulation can be used to estimate drug penetration in brain tissues. Lastly, in addition to highlighting landmark scientific and medical literature, we provide practical advice on formulation development, device selection, and pharmacokinetic modeling. Our review of clinical studies suggests a growing interest for intra-CSF delivery, particularly for targeted proteins.


Subject(s)
Brain/metabolism , Cerebrospinal Fluid/metabolism , Drug Delivery Systems , Pharmaceutical Preparations/metabolism , Cerebrospinal Fluid/chemistry , Humans , Pharmaceutical Preparations/chemistry
3.
Bioorg Med Chem Lett ; 29(15): 1968-1973, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31133534

ABSTRACT

Aberrant activation of calpain has been observed in various pathophysiological disorders including neurodegenerative diseases such as Alzheimer's Disease. Here we describe our efforts on ketoamide-based 1-benzyl-5-oxopyrrolidine-2-carboxamides as a novel series of highly selective calpain inhibitors mitigating the metabolic liability of carbonyl reduction. The most advanced compound from this new series, namely A-1212805 (ABT-957, Alicapistat) proceeded to clinical phase I studies.


Subject(s)
Glycoproteins/therapeutic use , Pyrrolidines/metabolism , Glycoproteins/pharmacology , Humans , Structure-Activity Relationship
4.
J Med Chem ; 60(16): 7123-7138, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28759231

ABSTRACT

Calpain overactivation has been implicated in a variety of pathological disorders including ischemia/reperfusion injury, cataract formation, and neurodegenerative diseases such as Alzheimer's disease (AD). Herein we describe our efforts leading to the identification of ketoamide-based 2-(3-phenyl-1H-pyrazol-1-yl)nicotinamides as potent and reversible inhibitors of calpain with high selectivity versus related cysteine protease cathepsins, other proteases, and receptors. Broad efficacy in a set of preclinical models relevant to AD suggests that inhibition of calpain represents an attractive approach with potential benefit for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Aminobutyrates/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pyrazoles/pharmacology , Aminobutyrates/chemical synthesis , Aminobutyrates/pharmacokinetics , Animals , Cathepsins , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacokinetics , Dogs , Hippocampus/metabolism , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Male , Microsomes, Liver/metabolism , Niacinamide/chemical synthesis , Niacinamide/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Sleep, REM/drug effects , Spectrin/metabolism , Stereoisomerism , Structure-Activity Relationship
5.
Neuropharmacology ; 101: 358-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26471422

ABSTRACT

The GABA(B) receptor has been indicated as a promising target for multiple CNS-related disorders. Baclofen, a prototypical orthosteric agonist, is used clinically for the treatment of spastic movement disorders, but is associated with unwanted side-effects, such as sedation and motor impairment. Positive allosteric modulators (PAM), which bind to a topographically-distinct site apart from the orthosteric binding pocket, may provide an improved side-effect profile while maintaining baclofen-like efficacy. GABA, the major inhibitory neurotransmitter in the CNS, plays an important role in the etiology and treatment of seizure disorders. Baclofen is known to produce anticonvulsant effects in the DBA/2J mouse audiogenic seizure test (AGS), suggesting it may be a suitable assay for assessing pharmacodynamic effects. Little is known about the effects of GABA(B) PAMs, however. The studies presented here sought to investigate the AGS test as a pharmacodynamic (PD) screening model for GABA(B) PAMs by comparing the profile of structurally diverse PAMs to baclofen. GS39783, rac-BHFF, CMPPE, A-1295120 (N-(3-(4-(4-chloro-3-fluorobenzyl)-6-methoxy-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide), and A-1474713 (N-(3-(4-(4-chlorobenzyl)-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide) all produced robust, dose-dependent anticonvulsant effects; a similar profile was observed with baclofen. Pre-treatment with the GABA(B) antagonist SCH50911 completely blocked the anticonvulsant effects of baclofen and CMPPE in the AGS test, indicating such effects are likely mediated by the GABA(B) receptor. In addition to the standard anticonvulsant endpoint of the AGS test, video tracking software was employed to assess potential drug-induced motor side-effects during the acclimation period of the test. This analysis was sensitive to detecting drug-induced changes in total distance traveled, which was used to establish a therapeutic index (TI = hypoactivity/anticonvulsant effects). Calculated TIs for A-1295120, CMPPE, rac-BHFF, GS39783, and A-1474713 were 5.31x, 5.00x, 4.74x, 3.41x, and 1.83x, respectively, whereas baclofen was <1. The results presented here suggest the DBA/2J mouse AGS test is a potentially useful screening model for detecting PD effects of GABA(B) PAMs and can provide an initial read-out on target-related motor side-effects. Furthermore, an improved TI was observed for PAMs compared to baclofen, indicating the PAM approach may be a viable therapeutic alternative to baclofen.


Subject(s)
Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Baclofen/therapeutic use , Seizures/drug therapy , Acoustic Stimulation/adverse effects , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Animals , Animals, Newborn , Cyclopentanes/pharmacology , Drug Interactions , GABA Agonists/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Male , Mice , Mice, Inbred DBA , Morpholines/pharmacology , Motor Activity/drug effects , Protein Binding/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Seizures/etiology , Sulfur Isotopes/pharmacokinetics
6.
Neuromolecular Med ; 17(3): 270-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25920466

ABSTRACT

Cognitive and behavioral acts go along with highly coordinated spatiotemporal activity patterns in neuronal networks. Most of these patterns are synchronized by coherent membrane potential oscillations within and between local networks. By entraining multiple neurons into a common time regime, such network oscillations form a critical interface between cellular activity and large-scale systemic functions. Synaptic integrity is altered in neurodegenerative diseases, and it is likely that this goes along with characteristic changes of coordinated network activity. This notion is supported by EEG recordings from human patients and from different animal models of such disorders. However, our knowledge about the pathophysiology of network oscillations in neurodegenerative diseases is surprisingly incomplete, and increased research efforts are urgently needed. One complicating factor is the pronounced diversity of network oscillations between different brain regions and functional states. Pathological changes must, therefore, be analyzed separately in each condition and affected area. However, cumulative evidence from different diseases may result, in the future, in more unifying "oscillopathy" concepts of neurodegenerative diseases. In this review, we report present evidence for pathological changes of network oscillations in Alzheimer's disease (AD), one of the most prominent and challenging neurodegenerative disorders. The heterogeneous findings from AD are contrasted to Parkinson's disease, where motor-related changes in specific frequency bands do already fulfill criteria of a valid biomarker.


Subject(s)
Brain Waves/physiology , Membrane Potentials/physiology , Nerve Net/physiopathology , Neurodegenerative Diseases/physiopathology , Alzheimer Disease/physiopathology , Animals , Basal Ganglia/physiopathology , Cerebral Cortex/physiopathology , Cognition Disorders/physiopathology , Computer Simulation , Cortical Synchronization/physiology , Disease Models, Animal , Electroencephalography , Epilepsy/physiopathology , Frontotemporal Dementia/physiopathology , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/physiopathology , Haplorhini , Hippocampus/physiopathology , Humans , Huntington Disease/physiopathology , Interneurons/physiology , Parkinson Disease/physiopathology , Rodentia , Synaptic Transmission/physiology , Tremor/physiopathology
7.
Eur J Pharmacol ; 702(1-3): 44-55, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23376566

ABSTRACT

Alzheimer's disease is accompanied by increased brain levels of soluble amyloid-ß (Aß) oligomers. It has been suggested that oligomers directly impair synaptic function, thereby causing cognitive deficits in Alzheimer's disease patients. Recently, it has been shown that synthetic Aß oligomers directly modulate P/Q-type calcium channels, possibly leading to excitotoxic cascades and subsequent synaptic decline. Using whole-cell recordings we studied the modulation of recombinant presynaptic calcium channels in HEK293 cells after application of a stable Aß oligomer preparation (Aß1-42 globulomer). Aß globulomer shifted the half-activation voltage of P/Q-type and N-type calcium channels to more hyperpolarized values (by 11.5 and 7.5 mV). Application of non-aggregated Aß peptides had no effect. We then analyzed the potential of calcium channel blockers to prevent Aß globulomer-induced synaptic decline in hippocampal slice cultures. Specific block of P/Q-type or N-type calcium channels with peptide toxins completely reversed Aß globulomer-induced deficits in glutamatergic neurotransmission. Two state-dependent low molecular weight P/Q-type and N-type calcium channel blockers also protected neurons from Aß-induced alterations. On the contrary, inhibition of L-type calcium channels failed to reverse the deficit. Our data show that Aß globulomer directly modulates recombinant P/Q-type and N-type calcium channels in HEK293 cells. Block of presynaptic calcium channels with both state-dependent and state-independent modulators can reverse Aß-induced functional deficits in synaptic transmission. These findings indicate that presynaptic calcium channel blockers may be a therapeutic strategy for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/physiology , Calcium Channels/physiology , Peptide Fragments/pharmacology , Synapses/drug effects , Animals , Calcium/physiology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Rats , Rats, Wistar , Synapses/physiology , omega-Agatoxin IVA/pharmacology , omega-Conotoxins/pharmacology
8.
Comb Chem High Throughput Screen ; 16(3): 233-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23228050

ABSTRACT

Development of calcium channel blockers is attractive, but has in the past been hampered by lack of high throughput electrophysiological technology. This limitation has been overcome by the implementation of automated patch clamp systems that allow identification of state-dependent compounds, which preferentially target pathologically overactive channels. We recently presented a fluorescence-based high-throughput screen for P/Q-type calcium channels followed by automated electrophysiology. Here, we provide a detailed description of the development of the secondary screen, and show the full analysis of the inactivation kinetics of the recombinant P/Q channel that served as a basis for the automated patch clamp protocol. Increasing the length of pre-depolarization shifted the inactivation to more hyperpolarized potentials. No steady-state inactivation was reached up to pre-depolarization durations of 3 min, while stability of the recordings progressively declined. As a compromise, a 3s pre-depolarization protocol was proposed for functional screening. In order to validate the electrophysiological screening, we compared kinetics and pharmacology of recombinant P/Q-type channels between automated and manual patch clamp measurements. Channel activation was similar under both conditions. By contrast, inactivation occurred at more hyperpolarized potentials in the automated system. Therefore, P/Q-type calcium channel inactivation is sensitive to the applied technological platform and needs to be adjusted when performing automated patch clamp recordings. Our results indicate that a thorough analysis of the inactivation kinetics is mandatory, when establishing an electrophysiological screening protocol for calcium channel blockers. As some data obtained by automated recordings may not be identical to manual patch clamp analysis, we recommend a proper initial validation of the screening assay and--if necessary--a posthoc adjustment of automated patch clamp values. The protocol presented here supports hit-to-lead and lead optimization efforts during the development of novel P/Q-type calcium channel blockers, and may be valuable for the generation of assays in other ion channel programs.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Drug Evaluation, Preclinical/methods , Cell Line , Humans , Patch-Clamp Techniques/methods , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
9.
Comb Chem High Throughput Screen ; 15(5): 372-85, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22272661

ABSTRACT

Dysfunction of P/Q-type calcium channels is thought to underlie a variety of neurological diseases. There is evidence that migraine, Alzheimer's disease, and epilepsy involve a gain-of-function of the channel, leading to abnormal presynaptic vesicle release. P/Q-channel blockers may normalize current flow and consequently lead to an alleviation of disease symptoms. Although the medical need is high, there are no such compounds on the market. Here we describe a high throughput screen (HTS) for P/Q-type calcium channel blockers and the confirmation of hits by automated electrophysiology. We generated a HEK293 cell line stably expressing the α1A subunit of the P/Q-type calcium channel under control of a tetracycline (Tet) promoter. The accessory ß1.1 and α2δ1 subunits were co-expressed constitutively. The cell line was pharmacologically characterized by ion channel specific modulators, and revealed functional P/Q-type calcium currents. Using a fluorescence imaging plate reader (FLIPR), an assay for P/Q-type calcium channels was established based on a calcium sensitive dye. HTS of a 150,000 compound-containing sub-library led to the identification of 3262 hits that inhibited the fluorescence signal with potencies below 10 µM. Hit-to-lead (HTL) efforts identified 12,400 analogues. Compounds were clustered into 37 series, and 8 series of interest were prioritized. An electrophysiological secondary screen, providing a more direct measure of channel function, was implemented into the HTL process. 27 selected exemplars of different chemotypes were validated by automated whole-cell patch clamp analysis at inactivated channel state. The discovery of P/Q-channel blockers may foster the development of new therapeutics for a variety of neurological diseases.


Subject(s)
Calcium Channel Blockers/analysis , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , High-Throughput Screening Assays/methods , Calcium Channel Blockers/pharmacology , Cell Line , Electrophysiology , HEK293 Cells , Humans , Patch-Clamp Techniques/methods , Spectrometry, Fluorescence/methods , Transfection
10.
J Neurosci ; 30(31): 10369-79, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20685980

ABSTRACT

Oligomers of the beta-amyloid (Abeta) peptide have been indicated in early neuropathologic changes in Alzheimer's disease. Here, we present a synthetic Abeta(20-42) oligomer (named globulomer) with a different conformation to monomeric and fibrillar Abeta peptide, enabling the generation of highly Abeta oligomer-specific monoclonal antibodies. The globulomer-derived antibodies specifically detect oligomeric but not monomeric or fibrillar Abeta in various Abeta preparations. The globulomer-specific antibody A-887755 was able to prevent Abeta oligomer binding and dynamin cleavage in primary hippocampal neurons and to reverse globulomer-induced reduced synaptic transmission. In amyloid precursor protein (APP) transgenic mice, vaccination with Abeta globulomer and treatment with A-887755 improved novel object recognition. The cognitive improvement is likely attributable to reversing a deficit in hippocampal synaptic spine density in APP transgenic mice as observed after treatment with A-887755. Our findings demonstrate that selective reduction of Abeta oligomers by immunotherapy is sufficient to normalize cognitive behavior and synaptic deficits in APP transgenic mice.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Antibodies, Monoclonal/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Analysis of Variance , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/cytology , Hippocampus/immunology , Immunoprecipitation , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/immunology , Rats , Rats, Wistar , Recognition, Psychology
11.
Neuropharmacology ; 59(4-5): 334-42, 2010.
Article in English | MEDLINE | ID: mdl-20650285

ABSTRACT

Amyloid-beta (Abeta) is toxic to neurons and such toxicity is - at least in part - mediated via the NMDA receptor. Calpain, a calcium dependent cystein protease, is part of the NMDA receptor-induced neurodegeneration pathway, and we previously reported that inhibition of calpain prevents excitotoxic lesions of the cholinergic nucleus basalis magnocellularis of Meynert. The present study reveals that inhibition of calpain is also neuroprotective in an in vivo model of Abeta oligomer-induced neurodegeneration in rats. Abeta-induced lesions of the nucleus basalis induced a significant decrease in the number of cholinergic neurons and their projecting fibers, as determined by analysis of choline-acetyltransferase in the nucleus basalis magnocellularis and cortical mantle of the lesioned animals. Treatment with the calpain inhibitor A-705253 significantly attenuated cholinergic neurodegeneration in a dose-dependent manner. Calpain inhibition also significantly diminished the accompanying neuroinflammatory response, as determined by immunohistochemical analysis of microglia activation. Administration of beta-amyloid markedly impaired performance in the novel object recognition test. Treatment with the calpain inhibitor, A-705253, dose-dependently prevented this behavioral deficit. In order to determine whether pre-treatment with the calpain inhibitor is necessary to exhibit its full protective effect on neurons we induced Abeta toxicity in primary neuronal cultures and administered A-705253 at various time points before and after Abeta oligomer application. Although the protective effect was higher when A-705253 was applied before induction of Abeta toxicity, calpain inhibition was still beneficial when applied up to 1h post-treatment. We conclude that inhibition of calpains may represent a valuable strategy for the prevention of Abeta oligomer-induced neuronal decline and associated cognitive deterioration.


Subject(s)
Amyloid beta-Peptides/toxicity , Benzamides/therapeutic use , Calpain/antagonists & inhibitors , Exploratory Behavior/physiology , Nerve Degeneration/enzymology , Nerve Degeneration/prevention & control , Peptide Fragments/toxicity , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Benzamides/pharmacology , Calpain/physiology , Cells, Cultured , Exploratory Behavior/drug effects , Female , Glycoproteins/pharmacology , Glycoproteins/therapeutic use , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Wistar
12.
Rev Neurosci ; 20(1): 1-12, 2009.
Article in English | MEDLINE | ID: mdl-19526730

ABSTRACT

Since Alois Alzheimer first described morphological alterations associated with his patient's dementia more than 100 years ago, Alzheimer's disease (AD) was defined as neurodegenerative disease caused by extracellular deposits of misfolded proteins. These amyloid plaques and neurofibrillary tangles have been unambiguously considered as hallmarks of this ailment, accompanied by devastating brain atrophy and cell loss. When a 40-42 amino acid peptide, called beta-amyloid (Abeta), was identified as a main component of amyloid plaques and a few genetic cases of AD were linked to Abeta metabolism, the Abeta hypothesis of AD was proposed. It was initially thought that an increase in Abeta42 precipitates plaque formation, which causes the generation of neurofibrillary tangles and ultimately the death of neurons. However, during the last decade it became apparent that soluble rather than deposited Abeta is associated with dementia. Among the constituents of soluble Abeta, small oligomeric forms were increasingly associated with neuropathology. There is now ample evidence that Abeta oligomers do not affect neuronal viability in general, but interfere specifically with synaptic function. Long-term neurophysiological impairment ultimately causes degeneration of synapses, which becomes most apparent on the morphological level by retraction of dendritic spines. Loss of meaningful synaptic connections in the brain of patients with AD will shatter their capacity to encode and retrieve memories. The precise molecular mechanism of Abeta oligomer-induced impairment of synaptic transmission is not fully understood, but there are several independent observations that oligomers interfere with the vesicular release machinery at the presynaptic terminal. While this hypothesis offers a promising avenue to understand the underlying cause of cognition and memory deficits in the AD brain, it also opens a possibility to address new mechanisms for preventing and ultimately curing AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Brain/pathology , Brain/physiopathology , Calcium Channels/metabolism , Calcium Signaling/physiology , Humans , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Synapses/pathology
13.
J Pharmacol Exp Ther ; 327(2): 343-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18701765

ABSTRACT

N-Methyl-D-aspartate (NMDA) receptor-mediated excitotoxicity is thought to underlie a variety of neurological disorders, and inhibition of either the NMDA receptor itself, or molecules of the intracellular cascade, may attenuate neurodegeneration in these diseases. Calpain, a calcium-dependent cysteine protease, has been identified as part of such an NMDA receptor-induced excitotoxic signaling pathway. The present study addressed the question of whether inhibition of calpain can prevent neuronal cell death and associated behavioral deficits in a disease-relevant animal model, which is based on excitotoxic lesions of the cholinergic nucleus basalis magnocellularis of Meynert. Excitotoxic lesions of the nucleus basalis with NMDA induced a markedly impaired performance in the novel object recognition test. Treatment with the calpain inhibitor, N-(1-benzyl-2-carbamoyl-2-oxoethyl)-2-[E-2-(4-diethlyaminomethylphenyl) ethen-1-yl]benzamide (A-705253), dose-dependently prevented the behavioral deficit. Subsequent analysis of choline acetyltransferase in the cortical mantle of the lesioned animals revealed that application of A-705253 dose-dependently and significantly attenuated cholinergic neurodegeneration. Calpain inhibition also significantly diminished the accompanying gliosis, as determined by immunohistochemical analysis of microglia activation. Finally, inhibition of calpain by A-705253 and the peptidic calpain inhibitor N-acetyl-Leu-Leu-Nle-CHO did not impair long-term potentiation in hippocampal slices, indicating that calpain inhibition interrupts NMDA excitotoxicity pathways without interfering with NMDA receptor-mediated signaling involved in cognition. We conclude that inhibition of calpains may represent a valuable strategy for the prevention of excitotoxicity-induced neuronal decline without interfering with the physiological neuronal functions associated with learning and memory processes. Thus, calpain inhibition may be a promising and novel approach for the treatment of various neurodegenerative disorders.


Subject(s)
Basal Nucleus of Meynert/drug effects , Benzamides/pharmacology , Calpain/antagonists & inhibitors , N-Methylaspartate/toxicity , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Animals , Basal Nucleus of Meynert/pathology , Cognition/drug effects , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/physiology , Long-Term Potentiation/drug effects , Male , Microglia/drug effects , Microglia/physiology , Motor Activity/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/physiology
14.
J Neurosci ; 28(4): 788-97, 2008 Jan 23.
Article in English | MEDLINE | ID: mdl-18216187

ABSTRACT

Abnormal accumulation of soluble oligomers of amyloid beta (Abeta) is believed to cause malfunctioning of neurons in Alzheimer's disease. It has been shown that Abeta oligomers impair synaptic plasticity, thereby altering the ability of the neuron to store information. We examined the underlying cellular mechanism of Abeta oligomer-induced synaptic modifications by using a recently described stable oligomeric Abeta preparation called "Abeta(1-42) globulomer." Synthetically prepared Abeta(1-42) globulomer has been shown to localize to neurons and impairs long-term potentiation (Barghorn et al., 2005). Here, we demonstrate that Abeta(1-42) globulomer does not affect intrinsic neuronal properties, as assessed by measuring input resistance and discharge characteristics, excluding an unspecific alteration of membrane properties. We provide evidence that Abeta(1-42) globulomer, at concentrations as low as 8 nM, specifically suppresses spontaneous synaptic activity resulting from a reduction of vesicular release at terminals of both GABAergic and glutamatergic synapses. EPSCs and IPSCs were primarily unaffected. A detailed search for the precise molecular target of Abeta(1-42) globulomer revealed a specific inhibition of presynaptic P/Q calcium currents, whereas other voltage-activated calcium currents remained unaltered. Because intact P/Q calcium currents are needed for synaptic plasticity, the disruption of such currents by Abeta(1-42) globulomer may cause deficits in cellular mechanisms of information storage in brains of Alzheimer's disease patients. The inhibitory effect of Abeta(1-42) globulomer on synaptic vesicle release could be reversed by roscovitine, a specific enhancer of P/Q currents. Selective enhancement of the P/Q calcium current may provide a promising strategy in the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Neural Inhibition/physiology , Peptide Fragments/chemistry , Synaptic Transmission/physiology , Amyloid beta-Peptides/physiology , Animals , Cells, Cultured , Glutamic Acid/physiology , Hippocampus/drug effects , Hippocampus/physiology , Peptide Fragments/physiology , Rats , Rats, Wistar , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/physiology
15.
J Neurochem ; 95(3): 834-47, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16135089

ABSTRACT

Amyloid beta-peptide (Abeta)(1-42) oligomers have recently been discussed as intermediate toxic species in Alzheimer's disease (AD) pathology. Here we describe a new and highly stable Abeta(1-42) oligomer species which can easily be prepared in vitro and is present in the brains of patients with AD and Abeta(1-42)-overproducing transgenic mice. Physicochemical characterization reveals a pure, highly water-soluble globular 60-kDa oligomer which we named 'Abeta(1-42) globulomer'. Our data indicate that Abeta(1-42) globulomer is a persistent structural entity formed independently of the fibrillar aggregation pathway. It is a potent antigen in mice and rabbits eliciting generation of Abeta(1-42) globulomer-specific antibodies that do not cross-react with amyloid precursor protein, Abeta(1-40) and Abeta(1-42) monomers and Abeta fibrils. Abeta(1-42) globulomer binds specifically to dendritic processes of neurons but not glia in hippocampal cell cultures and completely blocks long-term potentiation in rat hippocampal slices. Our data suggest that Abeta(1-42) globulomer represents a basic pathogenic structural principle also present to a minor extent in previously described oligomer preparations and that its formation is an early pathological event in AD. Selective neutralization of the Abeta globulomer structure epitope is expected to have a high potential for treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Animals , Antibodies , Antibody Specificity , Cells, Cultured , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Fatty Acids , Hippocampus/cytology , Humans , Long-Term Potentiation , Male , Mice , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Conformation , Rabbits , Rats , Rats, Sprague-Dawley , Solubility , Water/metabolism
16.
Brain Res Mol Brain Res ; 133(1): 110-8, 2005 Jan 05.
Article in English | MEDLINE | ID: mdl-15661371

ABSTRACT

Local protein synthesis in dendrites is thought to provide a mechanism for long-lasting modifications of synapses in response to physiological activity and behavioral experience. New synthesis of dendritic proteins may be triggered by various paradigms, including induction of epileptiform activity. Prerequisite for such modulated synthesis is a mechanism that limits translation of synaptodendritic mRNAs to times of demand. Recently identified as a translational repressor that is localized to dendrites, small untranslated BC1 RNA has been implicated in the regulation of postsynaptic protein synthesis. Here we show that translational repressor BC1 RNA is itself undergoing modulation as a result of neuronal stimulation. Induction of hippocampal epileptiform activity resulted in a significant decrease of BC1 RNA in the CA3 region over several hours after excitation. The observed decrease was cell-wide, thus indicating reduced expression rather than intracellular redistribution. We suggest that a downregulation of the translational repressor BC1 RNA serves to modulate postsynaptic protein complements in response to the induction of epileptiform activity. Such increased protein synthesis in dendrites may be required for the consolidation of enduring epileptogenic mechanisms.


Subject(s)
Dendrites/metabolism , Gene Expression Regulation/physiology , Kindling, Neurologic/metabolism , RNA, Small Cytoplasmic/metabolism , Analysis of Variance , Animals , Autoradiography/methods , Brain/cytology , Brain/physiology , Electric Stimulation , Electroencephalography/methods , Immunohistochemistry/methods , In Situ Hybridization/methods , Kindling, Neurologic/physiology , Male , RNA, Small Cytoplasmic/genetics , Rats , Rats, Sprague-Dawley
17.
J Physiol ; 563(Pt 3): 663-70, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15661820

ABSTRACT

The characteristic, behaviour-related network oscillations of the mammalian hippocampus (, gamma and ripples) are accompanied by strongly phase-coupled action potentials in specific subsets of GABAergic interneurones. It has been suggested that the resulting phasic, repetitive inhibition shapes rhythmic coherent activity of the neuronal network. Here, we examined whether synaptic inhibition entrains approximately 200 Hz network ripples by applying the GABA(A) receptor antagonist gabazine to CA1 minislices of mouse hippocampus. Gabazine blocked spontaneously occurring sharp wave-ripple (SPW-R) activity. However, local application of KCl to the dendritic layer elicited excitatory sharp waves on which approximately 200 Hz ripple oscillations were superimposed with equal temporal properties of native SPW-R. The activity also persisted in the additional presence of blockers of glutamatergic synaptic transmission. In contrast, synchrony was largely abolished after addition of gap junction blockers. Thus, GABAergic transmission appears to be involved in the generation of sharp waves but phasic inhibition is no prerequisite for the precise synchronization of hippocampal neurones during high-frequency oscillations at approximately 200 Hz. Gap junctions on the other hand seem to be necessary to orchestrate coordinated activity within the ripple frequency domain.


Subject(s)
Biological Clocks/physiology , Hippocampus/physiology , Interneurons/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Potassium Chloride/pharmacology , Pyridazines/pharmacology , Synaptic Transmission/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biological Clocks/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Hippocampus/drug effects , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Neural Inhibition/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
18.
J Biol Chem ; 279(50): 52613-22, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15371429

ABSTRACT

Protein kinase Mzeta (PKMzeta) is an atypical protein kinase C isoform that has been implicated in the protein synthesis-dependent maintenance of long term potentiation and memory storage in the brain. Synapse-associated kinases are uniquely positioned to promote enduring consolidation of structural and functional modifications at the synapse, provided that kinase mRNA is available on site for local input-specific translation. We now report that the mRNA encoding PKMzeta is rapidly transported and specifically localized to synaptodendritic neuronal domains. Transport of PKMzeta mRNA is specified by two cis-acting dendritic targeting elements (Mzeta DTEs). Mzeta DTE1, located at the interface of the 5'-untranslated region and the open reading frame, directs somato-dendritic export of the mRNA. Mzeta DTE2, in contrast, is located in the 3'-untranslated region and is required for delivery of the mRNA to distal dendritic segments. Colocalization with translational repressor BC1 RNA in hippocampal dendrites suggests that PKMzeta mRNA may be subject to translational control in local domains. Dendritic localization of PKMzeta mRNA provides a molecular basis for the functional integration of synaptic signal transduction and translational control pathways.


Subject(s)
Dendrites/metabolism , Memory/physiology , Protein Kinase C/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Base Sequence , Biological Transport, Active , Cells, Cultured , Hippocampus/metabolism , Nucleic Acid Conformation , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , Rats , Signal Transduction
19.
J Physiol ; 550(Pt 3): 873-87, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12807984

ABSTRACT

The mammalian hippocampus displays a peculiar pattern of fast (approximately 200 Hz) network oscillations superimposed on slower sharp waves. Such sharp wave-ripple complexes (SPW-R) have been implicated in memory consolidation. We have recently described a novel and unique method for studying SPW-R in naive slices of murine hippocampus. Here, we used this model to analyse network and cellular mechanisms of this type of network activity. SPW-R are usually generated within area CA3 but can also originate within the isolated CA1 region. Cellular synchronisation during SPW-R requires both excitatory and inhibitory synaptic transmission as well as electrical coupling, the latter being particularly important for the high-frequency component. Extracellular and intracellular recordings revealed a surprisingly strong inhibition of most CA1 pyramidal cells during SPW-R. A minority of active cells, however, increases action potential frequency and fires in strict synchrony with the field ripples. This strong separation between members and non-members of the network may serve to ensure a high signal-to-noise ratio in information processing during sharp wave-ripple complexes.


Subject(s)
Hippocampus/physiology , Nerve Net/physiology , Neurons/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Electric Stimulation , Electroencephalography , Electrophysiology , Extracellular Space/physiology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Gap Junctions/physiology , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Models, Neurological , Nerve Net/cytology , Pyramidal Cells/physiology , Pyridazines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Synapses/physiology , Uncoupling Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...