Network


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

Hotspot


Dive into the research topics where Paul S. Donnelly is active.

Publication


Featured researches published by Paul S. Donnelly.


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.


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.


Bioconjugate Chemistry | 2012

Comparison of 64Cu-Complexing Bifunctional Chelators for Radioimmunoconjugation: Labeling Efficiency, Specific Activity, and in Vitro/in Vivo Stability

Maggie S. Cooper; Michelle T. Ma; Kavitha Sunassee; Karen Shaw; Jennifer Williams; Rowena L. Paul; Paul S. Donnelly; Philip J. Blower

High radiolabeling efficiency, preferably to high specific activity, and good stability of the radioimmunoconjugate are essential features for a successful immunoconjugate for imaging or therapy. In this study, the radiolabeling efficiency, in vitro stability, and biodistribution of immunoconjugates with eight different bifunctional chelators labeled with (64)Cu were compared. The anti-CD20 antibody, rituximab, was conjugated to four macrocyclic bifunctional chelators (p-SCN-Bn-DOTA, p-SCN-Bn-Oxo-DO3A, p-SCN-NOTA, and p-SCN-PCTA), three DTPA derivatives (p-SCN-Bn-DTPA, p-SCN-CHX-A″-DTPA, and ITC-2B3M-DTPA), and a macrobicyclic hexamine (sarcophagine) chelator (sar-CO2H) = (1-NH2-8-NHCO(CH2)3CO2H)sar where sar = sarcophagine = 3,6,10,13,16,19-hexaazabicyclo[6.6.6]icosane). Radiolabeling efficiency under various conditions, in vitro stability in serum at 37 °C, and in vivo biodistribution and imaging in normal mice over 48 h were studied. All chelators except sar-CO2H were conjugated to rituximab by thiourea bond formation with an average of 4.9 ± 0.9 chelators per antibody molecule. Sar-CO2H was conjugated to rituximab by amide bond formation with 0.5 chelators per antibody molecule. Efficiencies of (64)Cu radiolabeling were dependent on the concentration of immunoconjugate. Notably, the (64)Cu-NOTA-rituximab conjugate demonstrated the highest radiochemical yield (95%) under very dilute conditions (31 nM NOTA-rituximab conjugate). Similarly, sar-CO-rituximab, containing 1/10th the number of chelators per antibody compared to that of other conjugates, retained high labeling efficiency (98%) at an antibody concentration of 250 nM. In contrast to the radioimmunoconjugates containing DTPA derivatives, which demonstrated poor serum stability, all macrocyclic radioimmunoconjugates were very stable in serum with <6% dissociation of (64)Cu over 48 h. In vivo biodistribution profiles in normal female Balb/C mice were similar for all the macrocyclic radioimmunoconjugates with most of the activity remaining in the blood pool up to 48 h. While all the macrocyclic bifunctional chelators are suitable for molecular imaging using (64)Cu-labeled antibody conjugates, NOTA and sar-CO2H show significant advantages over the others in that they can be radiolabeled rapidly at room temperature, under dilute conditions, resulting in high specific activity.


Journal of Neurochemistry | 2011

The Alzheimer's therapeutic PBT2 promotes amyloid-β degradation and GSK3 phosphorylation via a metal chaperone activity.

Peter J. Crouch; Maria S. Savva; Lin W. Hung; Paul S. Donnelly; Alexandra I. Mot; Sarah J. Parker; Mark Greenough; Irene Volitakis; Paul A. Adlard; Robert A. Cherny; Colin L. Masters; Ashley I. Bush; Kevin J. Barnham; Anthony R. White

J. Neurochem. (2011) 119, 220–230.


PLOS ONE | 2011

Metal Ionophore Treatment Restores Dendritic Spine Density and Synaptic Protein Levels in a Mouse Model of Alzheimer's Disease

Paul A. Adlard; Laura Bica; Anthony R. White; Milawaty Nurjono; Gulay Filiz; Peter J. Crouch; Paul S. Donnelly; Roberto Cappai; David Finkelstein; Ashley I. Bush

We have previously demonstrated that brief treatment of APP transgenic mice with metal ionophores (PBT2, Prana Biotechnology) rapidly and markedly improves learning and memory. To understand the potential mechanisms of action underlying this phenomenon we examined hippocampal dendritic spine density, and the levels of key proteins involved in learning and memory, in young (4 months) and old (14 months) female Tg2576 mice following brief (11 days) oral treatment with PBT2 (30 mg/kg/d). Transgenic mice exhibited deficits in spine density compared to littermate controls that were significantly rescued by PBT2 treatment in both the young (+17%, p<0.001) and old (+32%, p<0.001) animals. There was no effect of PBT2 on spine density in the control animals. In the transgenic animals, PBT2 treatment also resulted in significant increases in brain levels of CamKII (+57%, p = 0.005), spinophilin (+37%, p = 0.04), NMDAR1A (+126%, p = 0.02), NMDAR2A (+70%, p = 0.05), pro-BDNF (+19%, p = 0.02) and BDNF (+19%, p = 0.04). While PBT2-treatment did not significantly alter neurite-length in vivo, it did increase neurite outgrowth (+200%, p = 0.006) in cultured cells, and this was abolished by co-incubation with the transition metal chelator, diamsar. These data suggest that PBT2 may affect multiple aspects of snaptic health/efficacy. In Alzheimers disease therefore, PBT2 may restore the uptake of physiological metal ions trapped within extracellular β-amyloid aggregates that then induce biochemical and anatomical changes to improve cognitive function.


Dalton Transactions | 2003

Structural trends in copper(II) bis(thiosemicarbazone) radiopharmaceuticals

Philip J. Blower; Thomas C. Castle; Andrew R. Cowley; Jonathan R. Dilworth; Paul S. Donnelly; Elena Labisbal; Frank E. Sowrey; Simon J. Teat; Michael J. Went

Redox-related changes in biological properties of copper bis(thiosemicarbazone) radiopharmaceuticals are induced by backbone alkylation. To determine whether these changes are mediated by changes in core structural parameters, eight X-ray structures of variously alkylated complexes were determined. The complexes include the hypoxia tracer diacetylbis(4-methyl-3-thiosemicarbazonato)copper(II) (CuATSM). The structures of the nickel analogue NiATSM and the corresponding free ligand ATSMH2 were also included. Distortions from planarity were slight and only present when there were significant intermolecular interactions (mainly pairs of N–H–N and N–H–S hydrogen bonds). These give rise to cross-linked flat or helical ribbons of complexes. Alkylation at the terminal nitrogen atoms interrupts hydrogen bonding, allowing complexes to become planar, but does not otherwise affect the coordination sphere. Alkylation at the backbone carbon atoms increases the backbone C–C bond length, allowing the metal to fit better into the ligand cavity with shorter Cu–S bonds.


Journal of Experimental Medicine | 2012

The hypoxia imaging agent CuII(atsm) is neuroprotective and improves motor and cognitive functions in multiple animal models of Parkinson’s disease

Lin W. Hung; Victor L. Villemagne; Lesley Cheng; Nicki A. Sherratt; Scott Ayton; Anthony R. White; Peter J. Crouch; SinChun Lim; Su Ling Leong; Simon Wilkins; Jessica L. George; Blaine R. Roberts; Chi L. L. Pham; Xiang Liu; Francis Chi Keung Chiu; David M. Shackleford; Andrew Powell; Colin L. Masters; Ashley I. Bush; Graeme O'Keefe; Janetta G. Culvenor; Roberto Cappai; Robert A. Cherny; Paul S. Donnelly; Andrew F. Hill; David Finkelstein; Kevin J. Barnham

The PET imaging agent CuII(atsm) improves motor and cognitive function in Parkinson’s disease.


Angewandte Chemie | 2012

Conjugation of Transferrin to Azide‐Modified CdSe/ZnS Core–Shell Quantum Dots using Cyclooctyne Click Chemistry

Christine Schieber; Alessandra Bestetti; Jet Phey Lim; Anneke D. Ryan; Tich-Lam Nguyen; Robert James Eldridge; Anthony R. White; Paul A. Gleeson; Paul S. Donnelly; Spencer J. Williams; Paul Mulvaney

Twinkle twinkle quantum dot: Conjugation of biomolecules to azide-modified quantum dots (QDs) through a bifunctional linker, using strain-promoted azide-alkyne cycloaddition with the QD and a squaramide linkage to the biomolecule (see scheme). Transferrin-conjugated QDs were internalized by transferrin-receptor expressing HeLa cells.


Journal of Biological Chemistry | 2011

Diacetylbis(N(4)-methylthiosemicarbazonato) Copper(II) (CuII(atsm)) Protects against Peroxynitrite-induced Nitrosative Damage and Prolongs Survival in Amyotrophic Lateral Sclerosis Mouse Model

Cynthia P.W. Soon; Paul S. Donnelly; Bradley J. Turner; Lin W. Hung; Peter J. Crouch; Nicki A. Sherratt; Jiangli Tan; Nastasia K.-H. Lim; Linh Q. Lam; Laura Bica; SinChun Lim; James L. Hickey; Julia Morizzi; Andrew Powell; David Finkelstein; Janetta G. Culvenor; Colin L. Masters; James A. Duce; Anthony R. White; Kevin J. Barnham; Qiao-Xin Li

Background: CuII(atsm) [(diacetylbis(N(4)-methylthiosemicarbazonato) copper(II)] was orally administrated to transgenic SOD1G93A mice. Results: Treatment significantly prolonged lifespan with preservation of motor neurons. Reduced protein oxidation, attenuated astrocyte, and microglial activation also resulted from treatment. Conclusion: CuII(atsm) is neuroprotective in this model even when treatment begins after the onset of disease symptoms. Significance: The drug has therapeutic potential for amyotrophic lateral sclerosis. 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.


The Journal of Neuroscience | 2014

Oral Treatment with CuII(atsm) Increases Mutant SOD1 In Vivo but Protects Motor Neurons and Improves the Phenotype of a Transgenic Mouse Model of Amyotrophic Lateral Sclerosis

Blaine R. Roberts; Nastasia K.-H. Lim; Erin J. McAllum; Paul S. Donnelly; Dominic J. Hare; Philip Doble; Bradley J. Turner; Katherine A. Price; SinChun Lim; Brett Paterson; James L. Hickey; Tw Rhoads; Williams; Katja M. Kanninen; Lin W. Hung; Liddell; Alexandra Grubman; Jf Monty; Rm Llanos; Kramer; Julian F. B. Mercer; Ashley I. Bush; Colin L. Masters; James A. Duce; Qiao-Xin Li; Joseph S. Beckman; Kevin J. Barnham; Anthony R. White; Peter J. Crouch

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)copperII [CuII(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 CuII(atsm) decreased the pool of Cu-deficient SOD1 and increased the pool of fully metallated (holo) SOD1. Tracking isotopically enriched 65CuII(atsm) confirmed the increase in holo-SOD1 involved transfer of Cu from CuII(atsm) to SOD1, suggesting the improved locomotor function and survival of the CuII(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 CuII(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.

Collaboration


Dive into the Paul S. Donnelly's collaboration.

Top Co-Authors

Avatar

Anthony R. White

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge