Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nature ; 560(7720): 589-594, 2018 08.
Article in English | MEDLINE | ID: mdl-30089910

ABSTRACT

Dysfunction in prosocial interactions is a core symptom of autism spectrum disorder. However, the neural mechanisms that underlie sociability are poorly understood, limiting the rational development of therapies to treat social deficits. Here we show in mice that bidirectional modulation of the release of serotonin (5-HT) from dorsal raphe neurons in the nucleus accumbens bidirectionally modifies sociability. In a mouse model of a common genetic cause of autism spectrum disorder-a copy number variation on chromosome 16p11.2-genetic deletion of the syntenic region from 5-HT neurons induces deficits in social behaviour and decreases dorsal raphe 5-HT neuronal activity. These sociability deficits can be rescued by optogenetic activation of dorsal raphe 5-HT neurons, an effect requiring and mimicked by activation of 5-HT1b receptors in the nucleus accumbens. These results demonstrate an unexpected role for 5-HT action in the nucleus accumbens in social behaviours, and suggest that targeting this mechanism may prove therapeutically beneficial.


Subject(s)
Autism Spectrum Disorder/psychology , Autism Spectrum Disorder/therapy , Nucleus Accumbens/metabolism , Serotonin/metabolism , Social Behavior , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Chromosomes, Mammalian/genetics , Disease Models, Animal , Dorsal Raphe Nucleus/cytology , Dorsal Raphe Nucleus/metabolism , Humans , Male , Mice , Neural Pathways , Nucleus Accumbens/cytology , Optogenetics , Synteny/genetics
2.
Acta Neuropathol Commun ; 6(1): 57, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976255

ABSTRACT

Parkinson's disease is diagnosed upon the presentation of motor symptoms, resulting from substantial degeneration of dopaminergic neurons in the midbrain. Prior to diagnosis, there is a lengthy prodromal stage in which non-motor symptoms, including olfactory deficits (hyposmia), develop. There is limited information about non-motor impairments and there is a need for directed research into these early pathogenic cellular pathways that precede extensive dopaminergic death in the midbrain. The protein tau has been identified as a genetic risk factor in the development of sporadic PD. Tau knockout mice have been reported as an age-dependent model of PD, and this study has demonstrated that they develop motor deficits at 15-months-old. We have shown that at 7-month-old tau knockout mice present with an overt hyposmic phenotype. This olfactory deficit correlates with an accumulation of α-synuclein, as well as autophagic impairment, in the olfactory bulb. This pathological feature becomes apparent in the striatum and substantia nigra of 15-month-old tau knockout mice, suggesting the potential for a spread of disease. Initial primary cell culture experiments have demonstrated that ablation of tau results in the release of α-synuclein enriched exosomes, providing a potential mechanism for disease spread. These alterations in α-synuclein level as well as a marked autophagy impairment in the tau knockout primary cells recapitulate results seen in the animal model. These data implicate a pathological role for tau in early Parkinson's disease.


Subject(s)
Olfaction Disorders/etiology , Olfaction Disorders/genetics , Parkinson Disease/complications , tau Proteins/deficiency , Age Factors , Animals , Autophagy , Brain/metabolism , Brain/pathology , Disease Models, Animal , Exosomes/metabolism , Exosomes/pathology , Exosomes/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Odorants , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Parkinson Disease/pathology , Psychomotor Performance/physiology , Sequestosome-1 Protein/metabolism , alpha-Synuclein/metabolism , tau Proteins/genetics
3.
ACS Chem Neurosci ; 9(11): 2731-2740, 2018 11 21.
Article in English | MEDLINE | ID: mdl-29920069

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia worldwide accounting for around 70% of all cases. There is currently no treatment for AD beyond symptom management and attempts at developing disease-modifying therapies have yielded very little. These strategies have traditionally targeted the peptide Aß, which is thought to drive pathology. However, the lack of clinical translation of these Aß-centric strategies underscores the need for diverse treatment strategies targeting other aspects of the disease. Metal dyshomeostasis is a common feature of several neurodegenerative diseases such as AD, Parkinson's disease, and frontotemporal dementia, and manipulation of metal homeostasis has been explored as a potential therapeutic avenue for these diseases. The copper ionophore glyoxalbis-[N4-methylthiosemicarbazonato]Cu(II) (CuII(gtsm)) has previously been shown to improve the cognitive deficits seen in an AD animal model; however, the molecular mechanism remained unclear. Here we report that the treatment of two animal tauopathy models (APP/PS1 and rTg4510) with CuII(gtsm) recovers the cognitive deficits seen in both neurodegenerative models. In both models, markers of tau pathology were significantly reduced with CuII(gtsm) treatment, and in the APP/PS1 model, the levels of Aß remained unchanged. Analysis of tau kinases (GSK3ß and CDK5) revealed no drug induced changes; however, both models exhibited a significant increase in the levels of the structural subunit of the tau phosphatase, PP2A. These findings suggest that targeting the tau phosphatase PP2A has therapeutic potential for preventing memory impairments and reducing the tau pathology seen in AD and other tauopathies.


Subject(s)
Cognition/drug effects , Organometallic Compounds/pharmacology , Protein Phosphatase 2/drug effects , Spatial Memory/drug effects , Tauopathies/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Cyclin-Dependent Kinase 5/drug effects , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/psychology , Glycogen Synthase Kinase 3 beta/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Mutation , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/pathology , Presenilin-1/genetics , Protein Phosphatase 2/metabolism , Tauopathies/metabolism , Tauopathies/psychology , tau Proteins/genetics
4.
Science ; 357(6358): 1406-1411, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963257

ABSTRACT

The reward generated by social interactions is critical for promoting prosocial behaviors. Here we present evidence that oxytocin (OXT) release in the ventral tegmental area (VTA), a key node of the brain's reward circuitry, is necessary to elicit social reward. During social interactions, activity in paraventricular nucleus (PVN) OXT neurons increased. Direct activation of these neurons in the PVN or their terminals in the VTA enhanced prosocial behaviors. Conversely, inhibition of PVN OXT axon terminals in the VTA decreased social interactions. OXT increased excitatory drive onto reward-specific VTA dopamine (DA) neurons. These results demonstrate that OXT promotes prosocial behavior through direct effects on VTA DA neurons, thus providing mechanistic insight into how social interactions can generate rewarding experiences.


Subject(s)
Dopaminergic Neurons/physiology , Interpersonal Relations , Oxytocin/metabolism , Reward , Social Behavior , Ventral Tegmental Area/metabolism , Animals , Integrases , Mice , Mice, Knockout , Oxytocin/genetics , Paraventricular Hypothalamic Nucleus/cytology , Presynaptic Terminals/physiology
5.
Sci Rep ; 6: 22398, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928495

ABSTRACT

The administration of MPTP selectively targets the dopaminergic system resulting in Parkinsonism-like symptoms and is commonly used as a mice model of Parkinson's disease. We previously demonstrated that the neuroprotective compound Cu(II)(atsm) rescues nigral cell loss and improves dopamine metabolism in the MPTP model. The mechanism of action of Cu(II)(atsm) needs to be further defined to understand how the compound promotes neuronal survival. Whole genome transcriptomic profiling has become a popular method to examine the relationship between gene expression and function. Substantia nigra samples from MPTP-lesioned mice were evaluated using whole transcriptome sequencing to investigate the genes altered upon Cu(II)(atsm) treatment. We identified 143 genes affected by MPTP lesioning that are associated with biological processes related to brain and cognitive development, dopamine synthesis and perturbed synaptic neurotransmission. Upon Cu(II)(atsm) treatment, the expression of 40 genes involved in promoting dopamine synthesis, calcium signaling and synaptic plasticity were restored which were validated by qRT-PCR. The study provides the first detailed whole transcriptomic analysis of pathways involved in MPTP-induced Parkinsonism. In addition, we identify key therapeutic pathways targeted by a potentially new class of neuroprotective agents which may provide therapeutic benefits for other neurodegenerative disorders.


Subject(s)
Dopamine/biosynthesis , MPTP Poisoning/pathology , Neuroprotective Agents/therapeutic use , Organometallic Compounds/therapeutic use , Parkinson Disease, Secondary/genetics , Parkinson Disease/genetics , Substantia Nigra/metabolism , Synaptic Transmission/genetics , Thiosemicarbazones/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Calcium Signaling/genetics , Coordination Complexes , Disease Models, Animal , Dopamine/genetics , Dopaminergic Neurons/metabolism , MPTP Poisoning/genetics , Mice , Mice, Inbred C57BL , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Substantia Nigra/drug effects
6.
J Alzheimers Dis ; 44(1): 69-78, 2015.
Article in English | MEDLINE | ID: mdl-25182735

ABSTRACT

Oligomeric forms of amyloid-ß (Aß) are thought to be responsible for the pathogenesis of Alzheimer's disease. While many oligomers of Aß are thought to be naturally occurring in the brain of humans and/or transgenic animals, it is well known that Aß oligomers are also readily produced in vitro in the laboratory. In recent studies, we discovered that synthetic monomeric Aß (4.7 kDa) could be transformed by microdialysis to higher molecular weight species (approximately 56 kDa, by western blot). Surface-enhanced laser desorption/ionization mass spectrometry and electron microscopy further identified these species' as potential Aß oligomers. The production of similar species could also be produced by centrifugal filtration and this formation was concentration and pore-size dependent. These higher order species of Aß were resistant to dissolution in NaOH, HFIP, formic acid, urea, and guanidine. We postulate that we have identified a novel way of producing a high order species of oligomeric Aß and we provide evidence to suggest that Aß oligomers can quite easily be a product of normal laboratory practices. These data suggest that the experimental detection of higher order oligomers in tissues derived from Alzheimer's disease brains or from animal models of disease could, in some cases, be a product the method of analysis.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Brain/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/ultrastructure , Amyloid beta-Protein Precursor/genetics , Animals , Brain/ultrastructure , Dialysis , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Molecular Weight , Mutation/genetics , Peptide Fragments/chemistry , Presenilin-1/genetics , Silver Staining , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Dalton Trans ; 44(11): 4933-44, 2015 Mar 21.
Article in English | MEDLINE | ID: mdl-25515141

ABSTRACT

Alzheimer's disease is associated with the presence of insoluble protein deposits in the brain called amyloid plaques. The major constituent of these deposits is aggregated amyloid-ß peptide. Technetium-99m complexes that bind to amyloid-ß plaques could provide important diagnostic information on amyloid-ß plaque burden using Single Photon Emission Computed Tomography (SPECT). Tridentate ligands with a stilbene functional group were used to form complexes with the fac-[M(I)(CO)3](+) (M = Re or (99m)Tc) core. The rhenium carbonyl complexes with tridentate co-ligands that included a stilbene functional group and a dimethylamino substituent bound to amyloid-ß present in human frontal cortex brain tissue from subjects with Alzheimer's disease. This chemistry was extended to make the analogous [(99m)Tc(I)(CO)3](+) complexes and the complexes were sufficiently stable in human serum. Whilst the lipophilicity (log D7.4) of the technetium complexes appeared ideally suited for penetration of the blood-brain barrier, preliminary biodistribution studies in an AD mouse model (APP/PS1) revealed relatively low brain uptake (0.24% ID g(-1) at 2 min post injection).


Subject(s)
Amyloid beta-Peptides/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Rhenium/chemistry , Technetium/chemistry , Animals , Brain/diagnostic imaging , Brain/metabolism , Drug Stability , Humans , Mice , Organometallic Compounds/pharmacokinetics , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/metabolism , Protein Binding , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
8.
J Neurosci ; 34(23): 8021-31, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24899723

ABSTRACT

Mutations in the metallo-protein Cu/Zn-superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS) in humans and an expression level-dependent phenotype in transgenic rodents. We show that oral treatment with the therapeutic agent diacetyl-bis(4-methylthiosemicarbazonato)copper(II) [Cu(II)(atsm)] increased the concentration of mutant SOD1 (SOD1G37R) in ALS model mice, but paradoxically improved locomotor function and survival of the mice. To determine why the mice with increased levels of mutant SOD1 had an improved phenotype, we analyzed tissues by mass spectrometry. These analyses revealed most SOD1 in the spinal cord tissue of the SOD1G37R mice was Cu deficient. Treating with Cu(II)(atsm) decreased the pool of Cu-deficient SOD1 and increased the pool of fully metallated (holo) SOD1. Tracking isotopically enriched (65)Cu(II)(atsm) confirmed the increase in holo-SOD1 involved transfer of Cu from Cu(II)(atsm) to SOD1, suggesting the improved locomotor function and survival of the Cu(II)(atsm)-treated SOD1G37R mice involved, at least in part, the ability of the compound to improve the Cu content of the mutant SOD1. This was supported by improved survival of SOD1G37R mice that expressed the human gene for the Cu uptake protein CTR1. Improving the metal content of mutant SOD1 in vivo with Cu(II)(atsm) did not decrease levels of misfolded SOD1. These outcomes indicate the metal content of SOD1 may be a greater determinant of the toxicity of the protein in mutant SOD1-associated forms of ALS than the mutations themselves. Improving the metal content of SOD1 therefore represents a valid therapeutic strategy for treating ALS caused by SOD1.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neurons/drug effects , Mutation/genetics , Organometallic Compounds/administration & dosage , Superoxide Dismutase/genetics , Thiosemicarbazones/administration & dosage , Administration, Oral , Age Factors , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/pathology , Animals , Cation Transport Proteins/genetics , Chromatography, Gel , Coordination Complexes , Copper Transporter 1 , Disease Models, Animal , Humans , Locomotion/drug effects , Locomotion/genetics , Mice , Mice, Transgenic , Phenotype , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
9.
Hum Mol Genet ; 23(15): 4051-63, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24634145

ABSTRACT

All cases of Huntington's disease (HD) are caused by mutant huntingtin protein (mhtt), yet the molecular mechanisms that link mhtt to disease symptoms are not fully elucidated. Given glycogen synthase kinase-3 (GSK3) is implicated in several neurodegenerative diseases as a molecular mediator of neuronal decline and widely touted as a therapeutic target, we investigated GSK3 in cells expressing mhtt, brains of R6/1 HD mice and post-mortem human brain samples. Consistency in data across the two models and the human brain samples indicate decreased GSK3 signalling contributes to neuronal dysfunction in HD. Inhibitory phosphorylation of GSK3 (pGSK3) was elevated in mhtt cells and this appeared related to an overall energy metabolism deficit as the mhtt cells had less ATP and inhibiting ATP production in control cells expressing non-pathogenic htt with paraquat also increased pGSK3. pGSK3 was increased and ATP levels decreased in the frontal cortex and striatum of R6/1 mice and levels of cortical pGSK3 inversely correlated with cognitive function of the mice. Consistent with decreased GSK3 activity in the R6/1 mouse brain, ß-catenin levels were increased and phosphorylation of collapsin response mediator protein-2 (CRMP2) decreased in the frontal cortex where inhibitory phosphorylation of GSK3 was the greatest. pGSK3 was predominantly undetectable in HD and healthy control human brain samples, but levels of total GSK3 were decreased in the HD-affected frontal cortex and this correlated with decreased pCRMP2. Thus, disruptions to cortical GSK3 signalling, possibly due to localized energy metabolism deficits, appear to contribute to the cognitive symptoms of HD.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Glycogen Synthase Kinase 3/genetics , Huntington Disease/genetics , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Adenosine Triphosphate/biosynthesis , Aged , Aged, 80 and over , Animals , Cell Line , Cerebral Cortex/pathology , Corpus Striatum/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Paraquat/pharmacology , Phosphorylation , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
11.
Future Med Chem ; 4(8): 955-69, 2012 May.
Article in English | MEDLINE | ID: mdl-22650238

ABSTRACT

In 1906, Alois Alzheimer first characterized the disease that bears his name. Despite intensive research, which has led to a better understanding of the pathology, there is no effective treatment for this disease. Of the drugs approved by the US FDA, none are disease modifying, only symptomatic. Unfortunately, there have been a number of failed clinical trials in the past 10 years where studies show either no cognitive improvement or, worse, serious side effects associated with treatment. Hence, there is a need for the field to look at alternative approaches to therapy. In this review, we will discuss how metal dyshomeostasis occurs in aging and Alzheimer's disease. Concomitantly, we will discuss how targeting this dyshomeostasis offers an effective and novel therapeutic approach. Thus far, compounds that mediate these effects have shown great potential in both preclinical animal studies as well as in early-stage clinical trials.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Clioquinol/therapeutic use , Metals/metabolism , Molecular Targeted Therapy/methods , Organometallic Compounds/therapeutic use , Thiosemicarbazones/therapeutic use , Amyloid beta-Peptides/metabolism , Animals , Clioquinol/chemistry , Clioquinol/pharmacology , Copper/chemistry , Copper/pharmacology , Copper/therapeutic use , Humans , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Transition Elements/metabolism
12.
J Exp Med ; 209(4): 837-54, 2012 Apr 09.
Article in English | MEDLINE | ID: mdl-22473957

ABSTRACT

Parkinson's disease (PD) is a progressive, chronic disease characterized by dyskinesia, rigidity, instability, and tremors. The disease is defined by the presence of Lewy bodies, which primarily consist of aggregated α-synuclein protein, and is accompanied by the loss of monoaminergic neurons. Current therapeutic strategies only give symptomatic relief of motor impairment and do not address the underlying neurodegeneration. Hence, we have identified Cu(II)(atsm) as a potential therapeutic for PD. Drug administration to four different animal models of PD resulted in improved motor and cognition function, rescued nigral cell loss, and improved dopamine metabolism. In vitro, this compound is able to inhibit the effects of peroxynitrite-driven toxicity, including the formation of nitrated α-synuclein oligomers. Our results show that Cu(II)(atsm) is effective in reversing parkinsonian defects in animal models and has the potential to be a successful treatment of PD.


Subject(s)
Cognition/drug effects , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Organometallic Compounds/therapeutic use , Parkinson Disease/drug therapy , Radiopharmaceuticals/therapeutic use , Thiosemicarbazones/therapeutic use , Animals , Cell Line, Tumor , Coordination Complexes , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neuroprotective Agents/pharmacology , Organometallic Compounds/pharmacology , Parkinson Disease/psychology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacology , Rats , Rats, Sprague-Dawley , Thiosemicarbazones/pharmacology , alpha-Synuclein/chemistry
13.
J Biol Chem ; 286(51): 44035-44044, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22033929

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive paralyzing disease characterized by tissue oxidative damage and motor neuron degeneration. This study investigated the in vivo effect of diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuII(atsm)), which is an orally bioavailable, blood-brain barrier-permeable complex. In vitro the compound inhibits the action of peroxynitrite on Cu,Zn-superoxide dismutase (SOD1) and subsequent nitration of cellular proteins. Oral treatment of transgenic SOD1G93A mice with CuII(atsm) at presymptomatic and symptomatic ages was performed. The mice were examined for improvement in lifespan and motor function, as well as histological and biochemical changes to key disease markers. Systemic treatment of SOD1G93A mice significantly delayed onset of paralysis and prolonged lifespan, even when administered to symptomatic animals. Consistent with the properties of this compound, treated mice had reduced protein nitration and carbonylation, as well as increased antioxidant activity in spinal cord. Treatment also significantly preserved motor neurons and attenuated astrocyte and microglial activation in mice. Furthermore, CuII(atsm) prevented the accumulation of abnormally phosphorylated and fragmented TAR DNA-binding protein-43 (TDP-43) in spinal cord, a protein pivotal to the development of ALS. CuII(atsm) therefore represents a potential new class of neuroprotective agents targeting multiple major disease pathways of motor neurons with therapeutic potential for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Organometallic Compounds/chemistry , Peroxynitrous Acid/metabolism , Superoxide Dismutase/genetics , Thiosemicarbazones/chemistry , Animals , Antioxidants/chemistry , Astrocytes/cytology , Coordination Complexes , Copper/chemistry , DNA-Binding Proteins/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Neurodegenerative Diseases/embryology , Neurons/metabolism , Oxidative Stress , Oxygen/chemistry , Spinal Cord/pathology , Superoxide Dismutase-1 , Transgenes
14.
J Neurochem ; 119(1): 220-30, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21797865

ABSTRACT

Impaired metal ion homeostasis causes synaptic dysfunction and treatments for Alzheimer's disease (AD) that target metal ions have therefore been developed. The leading compound in this class of therapeutic, PBT2, improved cognition in a clinical trial with AD patients. The aim of the present study was to examine the cellular mechanism of action for PBT2. We show PBT2 induces inhibitory phosphorylation of the α- and ß-isoforms of glycogen synthase kinase 3 and that this activity is dependent on PBT2 translocating extracellular Zn and Cu into cells. This activity is supported when Aß:Zn aggregates are the source of extracellular Zn and adding PBT2 to Aß:Zn preparations promotes Aß degradation by matrix metalloprotease 2. PBT2-induced glycogen synthase kinase 3 phosphorylation appears to involve inhibition of the phosphatase calcineurin. Consistent with this, PBT2 increased phosphorylation of other calcineurin substrates, including cAMP response element binding protein and Ca²âº/calmodulin-dependent protein kinase. These data demonstrate PBT2 can decrease Aß levels by sequestering the Zn that promotes extracellular formation of protease resistant Aß:Zn aggregates, and that subsequent intracellular translocation of the Zn by PBT2 induces cellular responses with synapto-trophic potential. Intracellular translocation of Zn and Cu via the metal chaperone activity of PBT2 may be an important mechanism by which PBT2 improves cognitive function in people with AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Clioquinol/analogs & derivatives , Glycogen Synthase Kinase 3/metabolism , Metals/metabolism , Molecular Chaperones/metabolism , Alzheimer Disease/drug therapy , Blotting, Western , Calcineurin/metabolism , Calcineurin Inhibitors , Caspase 3/metabolism , Cell Line, Tumor , Clioquinol/pharmacology , Copper/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mass Spectrometry , Matrix Metalloproteinase 2/metabolism , Peptide Hydrolases/metabolism , Phosphorylation/drug effects , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...