Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 11(1): 200, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38111057

ABSTRACT

TAR DNA-binding protein 43 (TDP-43) and Fused in Sarcoma/Translocated in Sarcoma (FUS) are ribonucleoproteins associated with pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Under physiological conditions, TDP-43 and FUS are predominantly localized in the nucleus, where they participate in transcriptional regulation, RNA splicing and metabolism. In disease, however, they are typically mislocalized to the cytoplasm where they form aggregated inclusions. A number of shared cellular pathways have been identified that contribute to TDP-43 and FUS toxicity in neurodegeneration. In the present study, we report a novel pathogenic mechanism shared by these two proteins. We found that pathological FUS co-aggregates with a ribosomal protein, the Receptor for Activated C-Kinase 1 (RACK1), in the cytoplasm of spinal cord motor neurons of ALS, as previously reported for pathological TDP-43. In HEK293T cells transiently transfected with TDP-43 or FUS mutant lacking a functional nuclear localization signal (NLS; TDP-43ΔNLS and FUSΔNLS), cytoplasmic TDP-43 and FUS induced co-aggregation with endogenous RACK1. These co-aggregates sequestered the translational machinery through interaction with the polyribosome, accompanied by a significant reduction of global protein translation. RACK1 knockdown decreased cytoplasmic aggregation of TDP-43ΔNLS or FUSΔNLS and alleviated associated global translational suppression. Surprisingly, RACK1 knockdown also led to partial nuclear localization of TDP-43ΔNLS and FUSΔNLS in some transfected cells, despite the absence of NLS. In vivo, RACK1 knockdown alleviated retinal neuronal degeneration in transgenic Drosophila melanogaster expressing hTDP-43WT or hTDP-43Q331K and improved motor function of hTDP-43WT flies, with no observed adverse effects on neuronal health in control knockdown flies. In conclusion, our results revealed a novel shared mechanism of pathogenesis for misfolded aggregates of TDP-43 and FUS mediated by interference with protein translation in a RACK1-dependent manner. We provide proof-of-concept evidence for targeting RACK1 as a potential therapeutic approach for TDP-43 or FUS proteinopathy associated with ALS and FTLD.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Sarcoma , Animals , Humans , Amyotrophic Lateral Sclerosis/pathology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HEK293 Cells , Motor Neurons/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/pathology , Protein Biosynthesis , Sarcoma/metabolism , Sarcoma/pathology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Receptors for Activated C Kinase/genetics , Receptors for Activated C Kinase/metabolism , Neoplasm Proteins/genetics
2.
J Neurochem ; 157(3): 684-694, 2021 05.
Article in English | MEDLINE | ID: mdl-33251603

ABSTRACT

Various lines of evidence implicate oxidative stress in the pathogenic mechanism(s) underpinning tauopathies. Consequently, antioxidant therapies have been considered in clinical practice for the treatment of tauopathies such as Alzheimer's disease (AD), but with mixed results. We and others have previously reported increased protein oxidation upon expression of both human 0N3R (hTau0N3R ) and 0N4R (hTau0N4R ) tau in vivo. Building on these studies, we demonstrate here the suppression of hTau0N3R associated phenotypes in Drosophila melanogaster after treatment with vitamin C or vitamin E. Curiously the rescue of phenotype was seen without alteration in total tau level or alteration in phosphorylation at a number of disease-associated sites. Moreover, treatment with paraquat, a pro-oxidant drug, did not exacerbate the hTau0N3R phenotypes. This result following paraquat treatment is reminiscent of our previous findings with hTau0N4R which also causes greater oxidative stress when compared to hTau0N3R but has a milder phenotype. Collectively our data imply that the role of oxidative stress in tau-mediated toxicity is not straight forward and there may be isoform-specific effects as well as contribution of other factors. This may explain the ambiguous effects of anti-oxidant treatments on clinical outcome in dementia patients.


Subject(s)
Antioxidants/pharmacology , Vitamin E/pharmacology , tau Proteins/antagonists & inhibitors , tau Proteins/toxicity , Animals , Ascorbic Acid/pharmacology , Drosophila melanogaster , Larva , Locomotion/drug effects , Male , Oxidants/toxicity , Oxidative Stress/drug effects , Paraquat/toxicity , Phosphorylation/drug effects
3.
J Biol Chem ; 294(10): 3744-3759, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30635404

ABSTRACT

Extracellular vesicles (EVs) are secreted by myriad cells in culture and also by unicellular organisms, and their identification in mammalian fluids suggests that EV release also occurs at the organism level. However, although it is clearly important to better understand EVs' roles in organismal biology, EVs in solid tissues have received little attention. Here, we modified a protocol for EV isolation from primary neural cell culture to collect EVs from frozen whole murine and human neural tissues by serial centrifugation and purification on a sucrose gradient. Quantitative proteomics comparing brain-derived EVs from nontransgenic (NTg) and a transgenic amyotrophic lateral sclerosis (ALS) mouse model, superoxide dismutase 1 (SOD1)G93A, revealed that these EVs contain canonical exosomal markers and are enriched in synaptic and RNA-binding proteins. The compiled brain EV proteome contained numerous proteins implicated in ALS, and EVs from SOD1G93A mice were significantly depleted in myelin-oligodendrocyte glycoprotein compared with those from NTg animals. We observed that brain- and spinal cord-derived EVs, from NTg and SOD1G93A mice, are positive for the astrocyte marker GLAST and the synaptic marker SNAP25, whereas CD11b, a microglial marker, was largely absent. EVs from brains and spinal cords of the SOD1G93A ALS mouse model, as well as from human SOD1 familial ALS patient spinal cord, contained abundant misfolded and nonnative disulfide-cross-linked aggregated SOD1. Our results indicate that CNS-derived EVs from an ALS animal model contain pathogenic disease-causing proteins and suggest that brain astrocytes and neurons, but not microglia, are the main EV source.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Astrocytes/pathology , Extracellular Vesicles/enzymology , Neurons/pathology , Protein Folding , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/pathology , Glycoproteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Proteomics , Spinal Cord/pathology , Superoxide Dismutase-1/metabolism
4.
ACS Chem Neurosci ; 9(7): 1591-1606, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29614860

ABSTRACT

Oligomers of amyloid-ß (AßO) are deemed key in synaptotoxicity and amyloid seeding of Alzheimer's disease (AD). However, the heterogeneous and dynamic nature of AßO and inadequate markers for AßO subtypes have stymied effective AßO identification and therapeutic targeting in vivo. We identified an AßO-subclass epitope defined by differential solvent orientation of the lysine 28 side chain in a constrained loop of serine-asparagine-lysine (cSNK), rarely displayed in molecular dynamics simulations of monomer and fibril ensembles. A mouse monoclonal antibody targeting AßOcSNK recognizes ∼50-60 kDa SDS-resistant soluble Aß assemblages in AD brain and prolongs the lag phase of Aß aggregation in vitro. Acute peripheral infusion of a murine IgG1 anti-AßOcSNK in two AD mouse models reduced soluble brain Aß aggregates by 20-30%. Chronic cSNK peptide immunization of APP/PS1 mice engendered an anti-AßOcSNK IgG1 response without epitope spreading to Aß monomers or fibrils and was accompanied by preservation of global PSD95 expression and improved cued fear memory. Our data indicate that the oligomer subtype AßOcSNK participates in synaptotoxicity and propagation of Aß aggregation in vitro and in vivo.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Epitopes , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Brain/immunology , Brain/pathology , Brain Chemistry , Disease Models, Animal , Female , Humans , Male , Memory/physiology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Molecular Dynamics Simulation , Plaque, Amyloid/chemistry , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Protein Aggregation, Pathological , Protein Conformation , Protein Multimerization
5.
Mol Neurodegener ; 12(1): 60, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28830501

ABSTRACT

BACKGROUND: Alzheimer's Disease (AD), characterized by accumulation of beta-amyloid (Aß) plaques in the brain, can be caused by age-related failures to clear Aß from the brain through pathways that involve the cerebrovasculature. Vascular risk factors are known to increase AD risk, but less is known about potential protective factors. We hypothesize that high-density lipoproteins (HDL) may protect against AD, as HDL have vasoprotective properties that are well described for peripheral vessels. Epidemiological studies suggest that HDL is associated with reduced AD risk, and animal model studies support a beneficial role for HDL in selectively reducing cerebrovascular amyloid deposition and neuroinflammation. However, the mechanism by which HDL may protect the cerebrovascular endothelium in the context of AD is not understood. METHODS: We used peripheral blood mononuclear cell adhesion assays in both a highly novel three dimensional (3D) biomimetic model of the human vasculature composed of primary human endothelial cells (EC) and smooth muscle cells cultured under flow conditions, as well as in monolayer cultures of ECs, to study how HDL protects ECs from the detrimental effects of Aß. RESULTS: Following Aß addition to the abluminal (brain) side of the vessel, we demonstrate that HDL circulated within the lumen attenuates monocyte adhesion to ECs in this biofidelic vascular model. The mechanism by which HDL suppresses Aß-mediated monocyte adhesion to ECs was investigated using monotypic EC cultures. We show that HDL reduces Aß-induced PBMC adhesion to ECs independent of nitric oxide (NO) production, miR-233 and changes in adhesion molecule expression. Rather, HDL acts through scavenger receptor (SR)-BI to block Aß uptake into ECs and, in cell-free assays, can maintain Aß in a soluble state. We confirm the role of SR-BI in our bioengineered human vessel. CONCLUSION: Our results define a novel activity of HDL that suppresses Aß-mediated monocyte adhesion to the cerebrovascular endothelium.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Leukocytes, Mononuclear/metabolism , Lipoproteins, HDL/metabolism , Alzheimer Disease/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Monocytes/metabolism , Plaque, Amyloid/metabolism
6.
Neurobiol Dis ; 105: 74-83, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28502805

ABSTRACT

Tau exists as six closely related protein isoforms in the adult human brain. These are generated from alternative splicing of a single mRNA transcript and they differ in the absence or presence of two N-terminal and three or four microtubule binding domains. Typically all six isoforms have been considered functionally similar. However, their differential involvement in particular tauopathies raises the possibility that there may be isoform-specific differences in physiological function and pathological role. To explore this, we have compared the phenotypes induced by the 0N3R and 0N4R isoforms in Drosophila. Expression of the 3R isoform causes more profound axonal transport defects and locomotor impairments, culminating in a shorter lifespan than the 4R isoform. In contrast, the 4R isoform leads to greater neurodegeneration and impairments in learning and memory. Furthermore, the phosphorylation patterns of the two isoforms are distinct, as is their ability to induce oxidative stress. These differences are not consequent to different expression levels and are suggestive of bona fide physiological differences in isoform biology and pathological potential. They may therefore explain isoform-specific mechanisms of tau-toxicity and the differential susceptibility of brain regions to different tauopathies.


Subject(s)
Disease Models, Animal , Tandem Repeat Sequences/genetics , Tauopathies/genetics , Tauopathies/physiopathology , tau Proteins/genetics , Age Factors , Animals , Animals, Genetically Modified , Axonal Transport , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Humans , Larva/genetics , Learning/physiology , Locomotion/genetics , Male , Memory/physiology , Phenotype , Phosphorylation , Protein Isoforms/metabolism , Tauopathies/mortality , Tauopathies/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Visual Pathways/metabolism , tau Proteins/metabolism
7.
Nat Neurosci ; 20(4): 540-549, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28192395

ABSTRACT

Drugs of abuse alter synaptic connections in the reward circuitry of the brain, which leads to long-lasting behavioral changes that underlie addiction. Here we show that cadherin adhesion molecules play a critical role in mediating synaptic plasticity and behavioral changes driven by cocaine. We demonstrate that cadherin is essential for long-term potentiation in the ventral tegmental area and is recruited to the synaptic membranes of excitatory synapses onto dopaminergic neurons following cocaine-mediated behavioral conditioning. Furthermore, we show that stabilization of cadherin at the membrane of these synapses blocks cocaine-induced synaptic plasticity, leading to a reduction in conditioned place preference induced by cocaine. Our findings identify cadherins and associated molecules as targets of interest for understanding pathological plasticity associated with addiction.


Subject(s)
Cadherins/physiology , Cocaine/pharmacology , Conditioning, Psychological/physiology , Neuronal Plasticity/physiology , Ventral Tegmental Area/physiology , Animals , Cadherins/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Mice, Transgenic , Neuronal Plasticity/drug effects , Receptors, AMPA/metabolism , Synapses/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
8.
Sci Rep ; 5: 17191, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26608845

ABSTRACT

Aggregation of highly phosphorylated tau is a hallmark of Alzheimer's disease and other tauopathies. Nevertheless, animal models demonstrate that tau-mediated dysfunction/toxicity may not require large tau aggregates but instead may be caused by soluble hyper-phosphorylated tau or by small tau oligomers. Challenging this widely held view, we use multiple techniques to show that insoluble tau oligomers form in conditions where tau-mediated dysfunction is rescued in vivo. This shows that tau oligomers are not necessarily always toxic. Furthermore, their formation correlates with increased tau levels, caused intriguingly, by either pharmacological or genetic inhibition of tau kinase glycogen-synthase-kinase-3beta (GSK-3ß). Moreover, contrary to common belief, these tau oligomers were neither highly phosphorylated, and nor did they contain beta-pleated sheet structure. This may explain their lack of toxicity. Our study makes the novel observation that tau also forms non-toxic insoluble oligomers in vivo in addition to toxic oligomers, which have been reported by others. Whether these are inert or actively protective remains to be established. Nevertheless, this has wide implications for emerging therapeutic strategies such as those that target dissolution of tau oligomers as they may be ineffective or even counterproductive unless they act on the relevant toxic oligomeric tau species.


Subject(s)
Neurons/pathology , Protein Multimerization , tau Proteins/metabolism , Animals , Drosophila melanogaster/metabolism , Drosophila melanogaster/ultrastructure , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Microtubules/drug effects , Microtubules/metabolism , Neurons/drug effects , Neurons/ultrastructure , Phenotype , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , Protein Structure, Secondary , Solubility , Spectrum Analysis, Raman , tau Proteins/chemistry
9.
Front Neurol ; 4: 114, 2013.
Article in English | MEDLINE | ID: mdl-23964266

ABSTRACT

Aggregation of highly phosphorylated tau into aggregated forms such as filaments and neurofibrillary tangles is one of the defining pathological hallmarks of Alzheimer's disease and other tauopathies. Hence therapeutic strategies have focused on inhibition of tau phosphorylation or disruption of aggregation. However, animal models imply that tau-mediated dysfunction and toxicity do not require aggregation but instead are caused by soluble hyper-phosphorylated tau. Over the years, our findings from a Drosophila model of tauopathy have reinforced this. We have shown that highly phosphorylated wild-type human tau causes behavioral deficits resulting from synaptic dysfunction, axonal transport disruption, and cytoskeletal destabilization in vivo. These deficits are evident in the absence of neuronal death or filament/tangle formation. Unsurprisingly, both pharmacological and genetic inhibition of GSK-3ß rescue these tau phenotypes. However, GSK-3ß inhibition also unexpectedly increases tau protein levels, and produces insoluble granular tau oligomers. As well as underlining the growing consensus that tau toxicity is mediated by a highly phosphorylated soluble tau species, our findings further show that not all insoluble tau aggregates are toxic. Some tau aggregates, in particular tau oligomers, are non-toxic, and may even be protective against tau toxicity in vivo. This has serious implications for emerging therapeutic strategies to dissolve tau aggregates, which might be ineffective or even counter-productive. In light of this, it is imperative to identify the key toxic tau species and to understand how it mediates dysfunction and degeneration so that the effective disease-modifying therapies can be developed.

10.
Biochem Soc Trans ; 40(4): 693-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22817718

ABSTRACT

Insoluble aggregates of the microtubule-associated protein tau characterize a number of neurodegenerative diseases collectively termed tauopathies. These aggregates comprise abnormally hyperphosphorylated and misfolded tau proteins. Research in this field has traditionally focused on understanding how hyperphosphorylated and aggregated tau mediates dysfunction and toxicity in tauopathies. Recent findings from both Drosophila and rodent models of tauopathy suggest that large insoluble aggregates such as tau filaments and tangles may not be the key toxic species in these diseases. Thus some investigators have shifted their focus to study pre-filament tau species such as tau oligomers and hyperphosphorylated tau monomers. Interestingly, tau oligomers can exist in a variety of states including hyperphosphorylated and unphosphorylated forms, which can be both soluble and insoluble. It remains to be determined which of these oligomeric states of tau are causally involved in neurodegeneration and which signal the beginning of the formation of inert/protective filaments. It will be important to better understand this so that tau-based therapeutic interventions can target the most toxic tau species.


Subject(s)
tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Humans , Phosphorylation , Tauopathies/metabolism
11.
Biochem Biophys Res Commun ; 422(3): 351-7, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22580275

ABSTRACT

NELL-1 is a potent osteoinductive molecule that enhances bone formation in multiple animal models through currently unidentified pathways. In the present manuscript, we hypothesized that NELL-1 may regulate osteogenic differentiation accompanied by alteration of inorganic phosphate (Pi) entry into the osteoblast via sodium dependent phosphate (NaPi) transporters. To determine this, MC3T3-E1 pre-osteoblasts were cultured in the presence of recombinant human (rh)NELL-1 or rhBMP-2. Analysis was performed for intracellular Pi levels through malachite green staining, Pit-1 and Pit-2 expression, and forced upregulation of Pit-1 and Pit-2. Results showed rhNELL-1 to increase MC3T3-E1 matrix mineralization and Pi influx associated with activation of both Pit-1 and Pit-2 channels, with significantly increased Pit-2 production. In contrast, Pi transport elicited by rhBMP-2 showed to be associated with increased Pit-1 production only. Next, neutralizing antibodies against Pit-1 and Pit-2 completely abrogated the Pi influx effect of rhNELL-1, suggesting rhNELL-1 is dependent on both transporters. These results identify one potential mechanism of action for rhNELL-1 induced osteogenesis and highlight a fundamental difference between NELL-1 and BMP-2 signaling.


Subject(s)
Calcification, Physiologic/physiology , Nerve Tissue Proteins/metabolism , Osteoblasts/physiology , Osteogenesis/physiology , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/drug effects , Calcium-Binding Proteins , Cell Line , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Signal Transduction
12.
Int J Alzheimers Dis ; 2011: 598157, 2011.
Article in English | MEDLINE | ID: mdl-22254145

ABSTRACT

Drosophila melanogaster is an experimentally tractable model organism that has been used successfully to model aspects of many human neurodegenerative diseases. Drosophila models of tauopathy have provided valuable insights into tau-mediated mechanisms of neuronal dysfunction and death. Here we review the findings from Drosophila models of tauopathy reported over the past ten years and discuss how they have furthered our understanding of the pathogenesis of tauopathies. We also discuss the multitude of technical advantages that Drosophila offers, which make it highly attractive as a model for such studies.

13.
Acta Neuropathol ; 120(5): 593-604, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20617325

ABSTRACT

It has been hypothesised that tau protein, when hyper-phosphorylated as in Alzheimer's disease (AD), does not bind effectively to microtubules and is no longer able to stabilise them; thus microtubules break down, and axonal transport can no longer proceed efficiently in affected brain regions in AD and related tauopathies (tau-microtubule hypothesis). We have used Drosophila models of tauopathy to test all components of this hypothesis in vivo. We have previously shown that upon expression of human 0N3R tau in Drosophila motor neurons it becomes highly phosphorylated, resulting in disruptions to both axonal transport and synaptic function which culminate in behavioural phenotypes. We now show that the mechanism by which the human tau mediates these effects is twofold: first, as predicted by the tau-microtubule hypothesis, the highly phosphorylated tau exhibits significantly reduced binding to microtubules; and second, it participates in a pathogenic interaction with the endogenous normal Drosophila tau and sequesters it away from microtubules. This causes disruption of the microtubular cytoskeleton as evidenced by a reduction in the numbers of intact correctly-aligned microtubules and the appearance of microtubules that are not correctly oriented within the axon. These deleterious effects of human tau are phosphorylation dependent because treatment with LiCl to suppress tau phosphorylation increases microtubule binding of both human and Drosophila tau and restores cytoskeletal integrity. Notably, all these phospho-tau-mediated phenotypes occur in the absence of tau filament/neurofibrillary tangle formation or neuronal death, and may thus constitute the mechanism by which hyper-phosphorylated tau disrupts neuronal function and contributes to cognitive impairment prior to neuronal death in the early stages of tauopathies.


Subject(s)
Microtubules/metabolism , Microtubules/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , tau Proteins/metabolism , Animals , Blotting, Western , Drosophila , Humans , Immunohistochemistry , Immunoprecipitation , Microscopy, Electron, Transmission , Phosphorylation , Tauopathies/metabolism , Tauopathies/pathology
14.
Biochem Soc Trans ; 38(4): 988-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20658990

ABSTRACT

AD (Alzheimer's disease) is a neurodegenerative disorder characterized by the abnormal hyperphosphorylation and aggregation of the microtubule-associated protein tau and the misfolding and deposition of Abeta peptide. The mechanisms by which tau and Abeta become abnormal is not clearly understood, neither is it known what role either protein plays in the neurodegenerative process underlying AD. We have modelled aspects of AD in Drosophila melanogaster to shed light on these processes and to further our understanding of the relationship between tau and amyloid in this disease.


Subject(s)
Alzheimer Disease/pathology , Disease Models, Animal , Drosophila melanogaster , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Humans , Models, Biological , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Peptide Hydrolases/metabolism , Phosphorylation , Synaptic Transmission/physiology , tau Proteins/metabolism , tau Proteins/physiology
15.
Biochem Soc Trans ; 38(2): 564-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20298222

ABSTRACT

Axonal microtubules are essential for transport of materials to the synapse. Compromised microtubules and synaptic loss have been demonstrated in AD (Alzheimer's disease), which is believed to contribute to cognitive dysfunction before neuronal death in the early stages of the disease. The mechanism by which hyperphosphorylated tau, the building block of neurofibrillary tangles, one of the pathological hallmarks of AD, disrupts neuronal and synaptic function is unclear. There is a theory that hyperphosphorylated tau does not bind effectively to microtubules and is no longer able to function in stabilizing them, thus axonal transport can no longer proceed efficiently. This leads to synaptic dysfunction. We have tested this theory in a Drosophila model of tauopathies in which we expressed human tau (h-tau). Using this model, we have tested all aspects of this hypothesis and have demonstrated that axonal transport does become compromised in the presence of hyperphosphorylated h-tau and this leads to synaptic and behavioural defects. We are currently investigating the mechanism by which hyperphosphorylated h-tau mediates this effect and are preliminary data indicate that this entails phospho-tau-mediated effects that are predicted by the tau-microtubule hypothesis, as well as novel effects. These deleterious effects of h-tau occur in the absence of tau filaments and before neuronal death. This sequence of pathogenic events may constitute the mechanism by which abnormal tau disrupts neuronal and synaptic function and contributes to cognitive impairment before neuronal death in the early stages of tauopathies such as AD.


Subject(s)
Disease Models, Animal , Drosophila , Neurons/drug effects , Tauopathies/pathology , tau Proteins/pharmacology , Animals , Drosophila/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Models, Biological , Neurons/physiology , Phosphorylation/physiology , Solubility , Tauopathies/metabolism , Tauopathies/physiopathology , Up-Regulation/physiology , tau Proteins/metabolism
16.
J Oral Maxillofac Surg ; 68(2): 300-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20116699

ABSTRACT

PURPOSE: Most craniofacial birth defects contain skeletal components that require bone grafting. Although many growth factors have shown potential for use in bone regeneration, bone morphogenetic proteins (BMPs) are the most osteoinductive. However, supraphysiologic doses, high cost, and potential adverse effects stimulate clinicians and researchers to identify complementary molecules that allow a reduction in dose of BMP-2. Because NELL1 plays a key role as a regulator of craniofacial skeletal morphogenesis, especially in committed chondrogenic and osteogenic differentiation, and a previous synergistic mechanism has been identified, NELL1 is an ideal molecule for combination with BMP-2 in calvarial defect regeneration. We investigated the effect of NELL1 and BMP-2 on bone regeneration in vivo. MATERIALS AND METHODS: BMP-2 doses of 589 and 1,178 ng were grafted into 5-mm critical-sized rat calvarial defects, as compared with 589 ng of NELL1 plus 589 ng of BMP-2 and 1,178 ng of NELL1 plus 1,178 ng of BMP-2, and bone regeneration was analyzed. RESULTS: Live micro-computed tomography data showed increased bone formation throughout 4 to 8 weeks in all groups but a significant improvement when the lower doses of each molecule were combined. High-resolution micro-computed tomography and histology showed more mature and complete defect healing when the combination of NELL1 plus BMP-2 was compared with BMP-2 alone at lower doses. CONCLUSION: The observed potential synergy has significant value in the future treatment of patients with craniofacial defects requiring extensive bone grafting that would normally entail extraoral autogenous bone grafts or doses of BMP-2 in milligrams.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Bone Regeneration/drug effects , Nerve Tissue Proteins/pharmacology , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/administration & dosage , Dose-Response Relationship, Drug , Drug Carriers , Drug Synergism , Humans , Lactic Acid , Male , Nerve Tissue Proteins/administration & dosage , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Skull/surgery , Transforming Growth Factor beta/administration & dosage , X-Ray Microtomography
17.
Exp Neurol ; 223(2): 401-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19782075

ABSTRACT

Alzheimer's disease (AD) is characterised by neurofibrillary tangles composed of hyper-phosphorylated tau, and neuritic plaques composed of misfolded amyloid peptide (Abeta(42)). It is generally believed that the hyper-phosphorylated tau and oligomeric Abeta(42) are responsible for the neuronal dysfunction and cognitive impairments that underlie the early stages of AD, but the mechanism by which they interact in the pathogenic process is not clear. Mounting evidence suggests that Abeta(42) pathology lies upstream of hyper-phosphorylated tau pathology. Similarly much is being learnt about how each protein affects neuronal function. However, the impact that either pathological protein has on neuronal dysfunction caused by the other is not extensively studied. We have investigated this in a Drosophila model of AD in which we express both phosphorylated human tau (tau(wt)) and oligomeric Abeta(42). We find that expression of tau(wt) causes neuronal dysfunction by disrupting axonal transport and synaptic structure, and that this leads to behavioural impairments and reduced lifespan. Co-expression of Abeta(42) with tau(wt) increases tau phosphorylation and exacerbates all these tau-mediated phenotypes. Treatment of tau(wt)/Abeta(42) and flies with LiCl ameliorates the exacerbating effect of Abeta(42), suggesting that GSK-3beta may be involved in the mechanism by which Abeta(42) and tau(wt) interact to cause neuronal dysfunction. Conversely to the effect of Abeta(42), mimicking the wingless signalling pathway by co-expression of dishevelled with tau(wt) reduces tau phosphorylation and suppresses the tau-mediated phenotypes. It is therefore possible to speculate that the mechanism by which Abeta(42) interacts with tau in the pathogenesis of AD is by down-regulating endogenous wnt signalling.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Axonal Transport/physiology , Drosophila/physiology , Peptide Fragments/genetics , tau Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Down-Regulation/physiology , Drosophila/genetics , Drosophila Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Locomotion/physiology , Longevity/physiology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiology , Neurons/pathology , Neurons/physiology , Peptide Fragments/metabolism , Phenotype , Phosphorylation/physiology , Signal Transduction/physiology , Synapses/pathology , Synapses/physiology , Wnt1 Protein/metabolism , tau Proteins/metabolism
18.
J Neurosci ; 29(35): 10928-38, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19726651

ABSTRACT

Evidence suggests that NMDA-type glutamate receptors contribute to degeneration of striatal medium-sized spiny neurons (MSNs) in Huntington's disease (HD). Previously, we demonstrated that NMDA receptor (NMDAR)-mediated current and/or toxicity is increased in MSNs from the yeast artificial chromosome (YAC) transgenic mouse model expressing polyglutamine (polyQ)-expanded (mutant) full-length human huntingtin (htt). Others have shown that membrane-associated guanylate kinases (MAGUKs), such as PSD-95 and SAP102, modulate NMDAR surface expression and excitotoxicity in hippocampal and cortical neurons and that htt interacts with PSD-95. Here, we tested the hypothesis that an altered association between MAGUKs and NMDARs in mutant huntingtin-expressing cells contributes to increased susceptibility to excitotoxicity. We show that htt coimmunoprecipitated with SAP102 in HEK293T cells and striatal tissue from wild-type and YAC transgenic mice; however, the association of SAP102 with htt or the NMDAR NR2B subunit was unaffected by htt polyQ length, whereas association of PSD-95 with NR2B in striatal tissue was enhanced by increased htt polyQ length. Treatment of cultured MSNs with Tat-NR2B9c peptide blocked binding of NR2B with SAP102 and PSD-95 and reduced NMDAR surface expression by 20% in both YAC transgenic and wild-type MSNs, and also restored susceptibility to NMDAR excitoxicity in YAC HD MSNs to levels observed in wild-type MSNs; a similar effect on excitotoxicity was observed after knockdown of PSD-95 by small interfering RNA. Unlike previous findings in cortical and hippocampal neurons, rescue of NMDA toxicity by Tat-NR2B9c occurred independently of any effect on neuronal nitric oxide synthase activity. Our results elucidate further the mechanisms underlying enhanced excitotoxicity in HD.


Subject(s)
Chromosomes, Artificial, Yeast/metabolism , Disease Models, Animal , Excitatory Amino Acid Agents/metabolism , Huntington Disease/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cell Line , Cells, Cultured , Chromosomes, Artificial, Yeast/genetics , Disks Large Homolog 4 Protein , Genetic Predisposition to Disease , Guanylate Kinases , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics
19.
Cell Transplant ; 18(10): 1111-21, 2009.
Article in English | MEDLINE | ID: mdl-19650972

ABSTRACT

Human mesenchymal stem cells (hMSC) are being administered by direct intramyocardial (IM) injection into patients with myocardial dysfunction with an objective to improve clinical status. However, surprisingly little attention has been directed to qualifying hMSC functionality beyond simple viability. In particular, the transit of hMSCs through a small-caliber needle lumen, the final fluidic pathway for all IM injection devices, may be especially prone to inducing unwarranted effects on cell function. This study evaluated the changes in clonogenicity, gene expression, and cytokine secretion that may be induced in hMSC (20 million/ml) by injection through a 26-gauge Nitinol needle at two different flow rates compared to noninjected control samples. Results indicated that hMSC viability and colony forming unit (CFU) formation was not altered by changes in injection rate, although a trend toward lower titers was noted at the higher flow rate, for the specific batch of hMSCs studied. The gene expression and cytokine analysis data suggest that delivering a suspension of MSCs through narrow lumen needles may marginally alter certain gene expression programs, but that such in vitro effects are transient and not translated into measurable differences in protein production. Gene expression levels of four cytokines (bFGF, SDF-1, SCF, VEGF) were significantly different at 400 microl/min, and that of all cytokines were significantly different at 1600 microl/min when compared to controls (p < 0.05). These changes were less pronounced (statistically insignificant for most cases, p > 0.05) and, in certain instances directionally opposite, at 72 h. However, no differences in the amounts of secreted bFGF, VEGF, or TGF-beta were detectable at either of the two time points or flow rates. We infer that intramyocardial administration by transcatheter techniques is unlikely to interfere with the machinery required for cell replication or secretion of regulatory and other growth factors, which are the mainstays of MSC contribution to cardiac tissue repair and regeneration.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Catheterization , Cell Survival , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Compressive Strength , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Injections , Mesenchymal Stem Cells/metabolism , Shear Strength , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Biol Proced Online ; 11: 161-9, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19495917

ABSTRACT

The dissociation of adherent mesenchymal stem cell (MSC) monolayers with trypsin and enzyme-free dissociation buffer was compared. A significantly lower proportion of viable cells were obtained with enzyme-free dissociation buffers compared to trypsin. Subsequently, the dissociated cells were re-seeded on new cell culture dishes and were subjected to the MTT assay 24 h later. The proportion of viable cells that reattached was significantly lower for cells obtained by dissociation with enzyme-free dissociation buffer compared to trypsin. Frozen-thawed MSC displayed a similar trend, yielding consistently higher cell viability and reattachment rates when dissociated with trypsin compared to enzyme-free dissociation buffer. It was also demonstrated that exposure of trypsin-dissociated MSC to enzyme-free dissociation buffer for 1 h had no significant detrimental effect on cell viability.

SELECTION OF CITATIONS
SEARCH DETAIL