Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katrina M. Laughton is active.

Publication


Featured researches published by Katrina M. Laughton.


Neuron | 2008

Rapid Restoration of Cognition in Alzheimer's Transgenic Mice with 8-Hydroxy Quinoline Analogs Is Associated with Decreased Interstitial Aβ

Paul A. Adlard; Robert A. Cherny; David Finkelstein; Elisabeth Gautier; Elysia Robb; Mikhalina Cortes; Irene Volitakis; Xiang Liu; Jeffrey P. Smith; Keyla Perez; Katrina M. Laughton; Qiao-Xin Li; Susan A. Charman; Joseph A. Nicolazzo; Simon Wilkins; Karolina Deleva; Toni Lynch; Gaik Beng Kok; Craig W. Ritchie; Rudolph E. Tanzi; Roberto Cappai; Colin L. Masters; Kevin J. Barnham; Ashley I. Bush

As a disease-modifying approach for Alzheimers disease (AD), clioquinol (CQ) targets beta-amyloid (Abeta) reactions with synaptic Zn and Cu yet promotes metal uptake. Here we characterize the second-generation 8-hydroxy quinoline analog PBT2, which also targets metal-induced aggregation of Abeta, but is more effective as a Zn/Cu ionophore and has greater blood-brain barrier permeability. Given orally to two types of amyloid-bearing transgenic mouse models of AD, PBT2 outperformed CQ by markedly decreasing soluble interstitial brain Abeta within hours and improving cognitive performance to exceed that of normal littermate controls within days. Nontransgenic mice were unaffected by PBT2. The current data demonstrate that ionophore activity, inhibition of in vitro metal-mediated Abeta reactions, and blood-brain barrier permeability are indices that predict a potential disease-modifying drug for AD. The speed of recovery of the animals underscores the acutely reversible nature of the cognitive deficits associated with transgenic models of AD.


Journal of Biological Chemistry | 2006

Degradation of the Alzheimer Disease Amyloid β-Peptide by Metal-dependent Up-regulation of Metalloprotease Activity

Anthony R. White; Tai Du; Katrina M. Laughton; Irene Volitakis; Robyn A. Sharples; Michel Xilinas; David E. Hoke; R. M. Damian Holsinger; Genevieve Evin; Robert A. Cherny; Andrew F. Hill; Kevin J. Barnham; Qiao-Xin Li; Ashley I. Bush; Colin L. Masters

Biometals play an important role in Alzheimer disease, and recent reports have described the development of potential therapeutic agents based on modulation of metal bioavailability. The metal ligand clioquinol (CQ) has shown promising results in animal models and small phase clinical trials; however, the actual mode of action in vivo has not been determined. We now report a novel effect of CQ on amyloid β-peptide (Aβ) metabolism in cell culture. Treatment of Chinese hamster ovary cells overexpressing amyloid precursor protein with CQ and Cu2+ or Zn2+ resulted in an ∼85–90% reduction of secreted Aβ-(1–40) and Aβ-(1–42) compared with untreated controls. Analogous effects were seen in amyloid precursor protein-overexpressing neuroblastoma cells. The secreted Aβ was rapidly degraded through up-regulation of matrix metalloprotease (MMP)-2 and MMP-3 after addition of CQ and Cu2+. MMP activity was increased through activation of phosphoinositol 3-kinase and JNK. CQ and Cu2+ also promoted phosphorylation of glycogen synthase kinase-3, and this potentiated activation of JNK and loss of Aβ-(1–40). Our findings identify an alternative mechanism of action for CQ in the reduction of Aβ deposition in the brains of CQ-treated animals and potentially in Alzheimer disease patients.


PLOS ONE | 2007

Mitochondrial oxidative stress causes hyperphosphorylation of tau.

Simon Melov; Paul A. Adlard; Karl Morten; Felicity Johnson; Tamara R. Golden; Doug Hinerfeld; Birgit Schilling; Christine Mavros; Colin L. Masters; Irene Volitakis; Qiao-Xin Li; Katrina M. Laughton; Alan Hubbard; Robert A. Cherny; Brad Gibson; Ashley I. Bush

Age-related neurodegenerative disease has been mechanistically linked with mitochondrial dysfunction via damage from reactive oxygen species produced within the cell. We determined whether increased mitochondrial oxidative stress could modulate or regulate two of the key neurochemical hallmarks of Alzheimers disease (AD): tau phosphorylation, and ß-amyloid deposition. Mice lacking superoxide dismutase 2 (SOD2) die within the first week of life, and develop a complex heterogeneous phenotype arising from mitochondrial dysfunction and oxidative stress. Treatment of these mice with catalytic antioxidants increases their lifespan and rescues the peripheral phenotypes, while uncovering central nervous system pathology. We examined sod2 null mice differentially treated with high and low doses of a catalytic antioxidant and observed striking elevations in the levels of tau phosphorylation (at Ser-396 and other phospho-epitopes of tau) in the low-dose antioxidant treated mice at AD-associated residues. This hyperphosphorylation of tau was prevented with an increased dose of the antioxidant, previously reported to be sufficient to prevent neuropathology. We then genetically combined a well-characterized mouse model of AD (Tg2576) with heterozygous sod2 knockout mice to study the interactions between mitochondrial oxidative stress and cerebral Aß load. We found that mitochondrial SOD2 deficiency exacerbates amyloid burden and significantly reduces metal levels in the brain, while increasing levels of Ser-396 phosphorylated tau. These findings mechanistically link mitochondrial oxidative stress with the pathological features of AD.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Increasing Cu bioavailability inhibits Aβ oligomers and tau phosphorylation

Peter J. Crouch; Lin Wai Hung; Paul A. Adlard; Mikhalina Cortes; Varsha Lal; Gulay Filiz; Keyla Perez; Milawaty Nurjono; Aphrodite Caragounis; Tai Du; Katrina M. Laughton; Irene Volitakis; Ashley I. Bush; Qiao-Xin Li; Colin L. Masters; Roberto Cappai; Robert A. Cherny; Paul S. Donnelly; Anthony R. White; Kevin J. Barnham

Cognitive decline in Alzheimers disease (AD) involves pathological accumulation of synaptotoxic amyloid-β (Aβ) oligomers and hyperphosphorylated tau. Because recent evidence indicates that glycogen synthase kinase 3β (GSK3β) activity regulates these neurotoxic pathways, we developed an AD therapeutic strategy to target GSK3β. The strategy involves the use of copper-bis(thiosemicarbazonoto) complexes to increase intracellular copper bioavailability and inhibit GSK3β through activation of an Akt signaling pathway. Our lead compound CuII(gtsm) significantly inhibited GSK3β in the brains of APP/PS1 transgenic AD model mice. CuII(gtsm) also decreased the abundance of Aβ trimers and phosphorylated tau, and restored performance of AD mice in the Y-maze test to levels expected for cognitively normal animals. Improvement in the Y-maze correlated directly with decreased Aβ trimer levels. This study demonstrates that increasing intracellular copper bioavailability can restore cognitive function by inhibiting the accumulation of neurotoxic Aβ trimers and phosphorylated tau.


The Journal of Neuroscience | 2007

In Vitro Characterization of Pittsburgh Compound-B Binding to Lewy Bodies

Michelle Fodero-Tavoletti; David P. Smith; Catriona McLean; Paul A. Adlard; Kevin J. Barnham; Lisa Foster; Laura Leone; Keyla Perez; Mikhalina Cortes; Janetta G. Culvenor; Qiao-Xin Li; Katrina M. Laughton; Christopher C. Rowe; Colin L. Masters; Roberto Cappai; Victor L. Villemagne

Dementia with Lewy bodies (DLB) is pathologically characterized by the presence of α-synuclein-containing Lewy bodies within the neocortical, limbic, and paralimbic regions. Like Alzheimers disease (AD), Aβ plaques are also present in most DLB cases. The contribution of Aβ to the development of DLB is unclear. [11C]-Pittsburgh compound B ([11C]-PIB) is a thioflavin-T derivative that has allowed in vivo Aβ burden to be quantified using positron emission tomography (PET). [11C]-PIB PET studies have shown similar high cortical [11C]-PIB binding in AD and DLB subjects. To establish the potential binding of PIB to α-synuclein in DLB patients, we characterized the in vitro binding of PIB to recombinant human α-synuclein and DLB brain homogenates. Analysis of the in vitro binding studies indicated that [3H]-PIB binds to α-synuclein fibrils but with lower affinity than that demonstrated/reported for Aβ1–42 fibrils. Furthermore, [3H]-PIB was observed to bind to Aβ plaque-containing DLB brain homogenates but failed to bind to DLB homogenates that were Aβ plaque-free (“pure DLB”). Positive PIB fluorescence staining of DLB brain sections colocalized with immunoreactive Aβ plaques but failed to stain Lewy bodies. Moreover, image quantification analysis suggested that given the small size and low density of Lewy bodies within the brains of DLB subjects, any contribution of Lewy bodies to the [11C]-PIB PET signal would be negligible. These studies indicate that PIB retention observed within the cortical gray matter regions of DLB subjects in [11C]-PIB PET studies is largely attributable to PIB binding to Aβ plaques and not Lewy bodies.


Journal of Biological Chemistry | 2008

Selective Intracellular Release of Copper and Zinc Ions from Bis(thiosemicarbazonato) Complexes Reduces Levels of Alzheimer Disease Amyloid-β Peptide

Paul S. Donnelly; Aphrodite Caragounis; Tai Du; Katrina M. Laughton; Irene Volitakis; Robert A. Cherny; Robyn A. Sharples; Andrew F. Hill; Qiao-Xin Li; Colin L. Masters; Kevin J. Barnham; Anthony R. White

Copper and zinc play important roles in Alzheimer disease pathology with recent reports describing potential therapeutics based on modulation of metal bioavailability. We examined the ability of a range of metal bis(thiosemicarbazonato) complexes (MII(btsc), where M = CuII or ZnII) to increase intracellular metal levels in Chinese hamster ovary cells overexpressing amyloid precursor protein (APP-CHO) and the subsequent effect on extracellular levels of amyloid-β peptide (Aβ). The CuII(btsc) complexes were engineered to be either stable to both a change in oxidation state and dissociation of metal or susceptible to intracellular reduction and dissociation of metal. Treatment of APP-CHO cells with stable complexes resulted in elevated levels of intracellular copper with no effect on the detected levels of Aβ. Treatment with complexes susceptible to intracellular reduction increased intracellular copper levels but also resulted in a dose-dependent reduction in the levels of monomeric Aβ. Treatment with less stable ZnII(btsc) complexes increased intracellular zinc levels with a subsequent dose-dependent depletion of monomeric Aβ levels. The increased levels of intracellular bioavailable copper and zinc initiated a signaling cascade involving activation of phosphoinositol 3-kinase and c-Jun N-terminal kinase. Inhibition of these enzymes prevented Aβ depletion induced by the MII(btsc) complexes. Inhibition of metalloproteases also partially restored Aβ levels, implicating metal-driven metalloprotease activation in the extracellular monomeric Aβ depletion. However, a role for alternative metal-induced Aβ metabolism has not been ruled out. These studies demonstrate that MII(btsc) complexes have potential for Alzheimer disease therapy.


Journal of Alzheimer's Disease | 2010

Plasma Amyloid-β as a Biomarker in Alzheimer's Disease: The AIBL Study of Aging

James Lui; Simon M. Laws; Qiao-Xin Li; Victor L. Villemagne; David Ames; Belinda M. Brown; Ashley I. Bush; Karl De Ruyck; Jasmin Dromey; K. Ellis; Noel G. Faux; Jonathan K. Foster; Chris Fowler; Veer Gupta; Peter J. Hudson; Katrina M. Laughton; Colin L. Masters; Kelly K. Pertile; Alan Rembach; Mira Rimajova; Mark Rodrigues; Christopher C. Rowe; Rebecca L. Rumble; Cassandra Szoeke; Kevin Taddei; Tania Taddei; Brett Trounson; Vanessa Ward; Ralph N. Martins

Amyloid-beta (Abeta) plays a central role in the pathogenesis of Alzheimers disease (AD) and has been postulated as a potential biomarker for AD. However, there is a lack of consensus as to its suitability as an AD biomarker. The objective of this study was to determine the significance of plasma Abeta as an AD biomarker and its relationship with Abeta load and to determine the effect of different assay methods on the interpretation of Abeta levels. Plasma Abeta1-40, Abeta1-42, and N-terminal cleaved fragments were measured using both a commercial multiplex assay and a well-documented ELISA in 1032 individuals drawn from the well-characterized Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Further, Abeta levels were compared to Abeta load derived from positron-emission tomography (PET) with the Pittsburgh compound B (PiB). Lower Abeta1-42 and Abeta1-42/1-40 ratio were observed in patients with AD and inversely correlated with PiB-PET derived Abeta load. However, assay methodology significantly impacted the interpretation of data. The cross-sectional analysis of plasma Abeta isoforms suggests that they may not be sufficient per se to diagnose AD. The value of their measurement in prognosis and monitoring of AD interventions needs further study, in addition to future longitudinal comparisons together with other predictors, which will determine whether plasma Abeta has diagnostic value in a panel of biomarkers.


Neurobiology of Disease | 2004

APP intracellular domain is increased and soluble Aβ is reduced with diet-induced hypercholesterolemia in a transgenic mouse model of Alzheimer disease

Amee J. George; R. M. Damian Holsinger; Catriona McLean; Katrina M. Laughton; Konrad Beyreuther; Genevieve Evin; Colin L. Masters; Qiao-Xin Li

Cholesterol is one of multiple factors, other than familial genetic mutations, that can influence amyloid-beta peptide (Abeta) metabolism and accumulation in Alzheimer disease (AD). The effect of a high-cholesterol diet on amyloid precursor protein (APP) processing in brain has not been thoroughly studied. This study was designed to further investigate the role of cholesterol in the production of Abeta and APP intracellular domain (AICD) in 12-month-old Tg2576 transgenic mice. The mice were maintained on a high-cholesterol diet for 6 weeks. We found that diet-induced hypercholesterolemia increased the APP cytosolic fragment AICD and reduced sAPPalpha in the Tg2576 mice compared to the mice on a control basal diet. In addition, the levels of detergent-extracted Abeta40 were reduced, although no change in guanidine-extracted Abeta levels was observed. Full-length APP, alpha/betaC-terminal fragment (alpha/betaCTF), and beta-secretase (BACE) were not different in the cholesterol-fed mice compared to the control diet-fed mice. This study suggests that a high dietary cholesterol in aged mice may not only influence Abeta metabolism, but also regulate the AICD levels. AICD has a proposed role in signal transduction and apoptosis, hence modulation of AICD production could be an alternative mechanism by which cholesterol contributes to AD pathogenesis.


Journal of Biological Chemistry | 2011

Copper Promotes the Trafficking of the Amyloid Precursor Protein

Karla M. Acevedo; Ya Hui Hung; Andrew H. Dalziel; Qiao-Xin Li; Katrina M. Laughton; Krutika Wikhe; Alan Rembach; Blaine R. Roberts; Colin L. Masters; Ashley I. Bush; James Camakaris

Accumulation of the amyloid β peptide in the cortical and hippocampal regions of the brain is a major pathological feature of Alzheimer disease. Amyloid β peptide is generated from the sequential protease cleavage of the amyloid precursor protein (APP). We reported previously that copper increases the level of APP at the cell surface. Here we report that copper, but not iron or zinc, promotes APP trafficking in cultured polarized epithelial cells and neuronal cells. In SH-SY5Y neuronal cells and primary cortical neurons, copper promoted a redistribution of APP from a perinuclear localization to a wider distribution, including neurites. Importantly, a change in APP localization was not attributed to an up-regulation of APP protein synthesis. Using live cell imaging and endocytosis assays, we found that copper promotes an increase in cell surface APP by increasing its exocytosis and reducing its endocytosis, respectively. This study identifies a novel mechanism by which copper regulates the localization and presumably the function of APP, which is of major significance for understanding the role of APP in copper homeostasis and the role of copper in Alzheimer disease.


Journal of Neurochemistry | 2009

Restored degradation of the Alzheimer's amyloid-β peptide by targeting amyloid formation

Peter J. Crouch; Deborah J. Tew; Tai Du; Diem Nguyen; Aphrodite Caragounis; Gulay Filiz; Rachel E. Blake; Ian A. Trounce; Cynthia P.W. Soon; Katrina M. Laughton; Keyla Perez; Qiao-Xin Li; Robert A. Cherny; Colin L. Masters; Kevin J. Barnham; Anthony R. White

Accumulation of neurotoxic amyloid‐β (Aβ) is central to the pathology of Alzheimer’s disease (AD). Elucidating the mechanisms of Aβ accumulation will therefore expedite the development of Aβ‐targeting AD therapeutics. We examined activity of an Aβ‐degrading protease (matrix metalloprotease 2) to investigate whether biochemical factors consistent with conditions in the AD brain contribute to Aβ accumulation by altering Aβ sensitivity to proteolytic degradation. An Aβ amino acid mutation found in familial AD, Aβ interactions with zinc (Zn), and increased Aβ hydrophobicity all strongly prevented Aβ degradation. Consistent to all of these factors is the promotion of specific Aβ aggregates where the protease cleavage site, confirmed by mass spectrometry, is inaccessible within an amyloid structure. These data indicate decreased degradation due to amyloid formation initiates Aβ accumulation by preventing normal protease activity. Zn also prevented Aβ degradation by the proteases neprilysin and insulin degrading enzyme. Treating Zn‐induced Aβ amyloid with the metal‐protein attenuating compound clioquinol reversed amyloid formation and restored the peptide’s sensitivity to degradation by matrix metalloprotease 2. This provides new data indicating that therapeutic compounds designed to modulate Aβ‐metal interactions can inhibit Aβ accumulation by restoring the catalytic potential of Aβ‐degrading proteases.

Collaboration


Dive into the Katrina M. Laughton's collaboration.

Top Co-Authors

Avatar

Qiao-Xin Li

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irene Volitakis

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Robert A. Cherny

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Ashley I. Bush

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony R. White

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul A. Adlard

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge