Florey Department of Neuroscience and Mental Health - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 10
  • Item
    Thumbnail Image
    The Compound ATH434 Prevents Alpha-Synuclein Toxicity in a Murine Model of Multiple System Atrophy
    Finkelstein, D ; Shukla, JJ ; Cherny, RA ; Billings, JL ; Saleh, E ; Stefanova, N ; Barnham, KJ ; Adlard, PA (IOS PRESS, 2022)
    BACKGROUND: An elevation in iron levels, together with an accumulation of α-synuclein within the oligodendrocytes, are features of the rare atypical parkinsonian disorder, Multiple System Atrophy (MSA). We have previously tested the novel compound ATH434 (formally called PBT434) in preclinical models of Parkinson's disease and shown that it is brain-penetrant, reduces iron accumulation and iron-mediated redox activity, provides neuroprotection, inhibits alpha synuclein aggregation and lowers the tissue levels of alpha synuclein. The compound was also well-tolerated in a first-in-human oral dosing study in healthy and older volunteers with a favorable, dose-dependent pharmacokinetic profile. OBJECTIVE: To evaluate the efficacy of ATH434 in a mouse MSA model. METHODS: The PLP-α-syn transgenic mouse overexpresses α-synuclein, demonstrates oligodendroglial pathology, and manifests motor and non-motor aspects of MSA. Animals were provided ATH434 (3, 10, or 30 mg/kg/day spiked into their food) or control food for 4 months starting at 12 months of age and were culled at 16 months. Western blot was used to assess oligomeric and urea soluble α-synuclein levels in brain homogenates, whilst stereology was used to quantitate the number of nigral neurons and glial cell inclusions (GCIs) present in the substantia nigra pars compacta. RESULTS: ATH434 reduced oligomeric and urea soluble α-synuclein aggregation, reduced the number of GCIs, and preserved SNpc neurons. In vitro experiments suggest that ATH434 prevents the formation of toxic oligomeric "species of synuclein". CONCLUSION: ATH434 is a promising small molecule drug candidate that has potential to move forward to trial for treating MSA.
  • Item
    Thumbnail Image
    ATH434 Reverses Colorectal Dysfunction in the A53T Mouse Model of Parkinson's Disease
    Diwakarla, S ; McQuade, RM ; Constable, R ; Artaiz, O ; Lei, E ; Barnham, KJ ; Adlard, PA ; Cherny, RA ; Di Natale, MR ; Wu, H ; Chai, X-Y ; Lawson, VA ; Finkelstein, D ; Furness, JB (IOS PRESS, 2021)
    BACKGROUND: Gastrointestinal (GI) complications, that severely impact patient quality of life, are a common occurrence in patients with Parkinson's disease (PD). Damage to enteric neurons and the accumulation of alpha-synuclein in the enteric nervous system (ENS) are thought to contribute to this phenotype. Copper or iron chelators, that bind excess or labile metal ions, can prevent aggregation of alpha-synuclein in the brain and alleviate motor-symptoms in preclinical models of PD. OBJECTIVE: We investigated the effect of ATH434 (formally PBT434), a small molecule, orally bioavailable, moderate-affinity iron chelator, on colonic propulsion and whole gut transit in A53T alpha-synuclein transgenic mice. METHODS: Mice were fed ATH434 (30 mg/kg/day) for either 4 months (beginning at ∼15 months of age), after the onset of slowed propulsion ("treatment group"), or for 3 months (beginning at ∼12 months of age), prior to slowed propulsion ("prevention group"). RESULTS: ATH434, given after dysfunction was established, resulted in a reversal of slowed colonic propulsion and gut transit deficits in A53T mice to WT levels. In addition, ATH434 administered from 12 months prevented the slowed bead expulsion at 15 months but did not alter deficits in gut transit time when compared to vehicle-treated A53T mice. The proportion of neurons with nuclear Hu+ translocation, an indicator of neuronal stress in the ENS, was significantly greater in A53T than WT mice, and was reduced in both groups when ATH434 was administered. CONCLUSION: ATH434 can reverse some of the GI deficits and enteric neuropathy that occur in a mouse model of PD, and thus may have potential clinical benefit in alleviating the GI dysfunctions associated with PD.
  • Item
    Thumbnail Image
    The novel compound PBT434 prevents iron mediated neurodegeneration and alpha-synuclein toxicity in multiple models of Parkinson's disease (vol 5, 53, 2017)
    Finkelstein, DI ; Billings, JL ; Adlard, PA ; Ayton, S ; Sedjahtera, A ; Masters, CL ; Wilkins, S ; Shackleford, DM ; Charman, SA ; Bal, W ; Zawisza, IA ; Kurowska, E ; Gundlach, AL ; Ma, S ; Bush, AI ; Hare, DJ ; Doble, PA ; Crawford, S ; Gautier, ECL ; Parsons, J ; Huggins, P ; Barnham, KJ ; Cherny, RA (BMC, 2021-09-29)
  • Item
    Thumbnail Image
    The novel compound PBT434 prevents iron mediated neurodegeneration and alpha-synuclein toxicity in multiple models of Parkinson's disease
    Finkelstein, DI ; Billings, JL ; Adlard, PA ; Ayton, S ; Sedjahtera, A ; Masters, CL ; Wilkins, S ; Shackleford, DM ; Charman, SA ; Bal, W ; Zawisza, IA ; Kurowska, E ; Gundlach, AL ; Ma, S ; Bush, AI ; Hare, DJ ; Doble, PA ; Crawford, S ; Gautier, ECL ; Parsons, J ; Huggins, P ; Barnham, KJ ; Cherny, RA (BMC, 2017-06-28)
    Elevated iron in the SNpc may play a key role in Parkinson's disease (PD) neurodegeneration since drug candidates with high iron affinity rescue PD animal models, and one candidate, deferirpone, has shown efficacy recently in a phase two clinical trial. However, strong iron chelators may perturb essential iron metabolism, and it is not yet known whether the damage associated with iron is mediated by a tightly bound (eg ferritin) or lower-affinity, labile, iron pool. Here we report the preclinical characterization of PBT434, a novel quinazolinone compound bearing a moderate affinity metal-binding motif, which is in development for Parkinsonian conditions. In vitro, PBT434 was far less potent than deferiprone or deferoxamine at lowering cellular iron levels, yet was found to inhibit iron-mediated redox activity and iron-mediated aggregation of α-synuclein, a protein that aggregates in the neuropathology. In vivo, PBT434 did not deplete tissue iron stores in normal rodents, yet prevented loss of substantia nigra pars compacta neurons (SNpc), lowered nigral α-synuclein accumulation, and rescued motor performance in mice exposed to the Parkinsonian toxins 6-OHDA and MPTP, and in a transgenic animal model (hA53T α-synuclein) of PD. These improvements were associated with reduced markers of oxidative damage, and increased levels of ferroportin (an iron exporter) and DJ-1. We conclude that compounds designed to target a pool of pathological iron that is not held in high-affinity complexes in the tissue can maintain the survival of SNpc neurons and could be disease-modifying in PD.
  • Item
    Thumbnail Image
    The effect of paraformaldehyde fixation and sucrose cryoprotection on metal concentration in murine neurological tissue
    Hare, DJ ; George, JL ; Bray, L ; Volitakis, I ; Vais, A ; Ryan, TM ; Cherny, RA ; Bush, AI ; Masters, CL ; Adlard, PA ; Doble, PA ; Finkelstein, DI (ROYAL SOC CHEMISTRY, 2014-03)
  • Item
    Thumbnail Image
    Parkinson's Disease Iron Deposition Caused by Nitric Oxide-Induced Loss of β-Amyloid Precursor Protein
    Ayton, S ; Lei, P ; Hare, DJ ; Duce, JA ; George, JL ; Adlard, PA ; McLean, C ; Rogers, JT ; Cherny, RA ; Finkelstein, DI ; Bush, AI (SOC NEUROSCIENCE, 2015-02-25)
    Elevation of both neuronal iron and nitric oxide (NO) in the substantia nigra are associated with Parkinson's disease (PD) pathogenesis. We reported previously that the Alzheimer-associated β-amyloid precursor protein (APP) facilitates neuronal iron export. Here we report markedly decreased APP expression in dopaminergic neurons of human PD nigra and that APP(-/-) mice develop iron-dependent nigral cell loss. Conversely, APP-overexpressing mice are protected in the MPTP PD model. NO suppresses APP translation in mouse MPTP models, explaining how elevated NO causes iron-dependent neurodegeneration in PD.
  • Item
    Thumbnail Image
    Iron accumulation confers neurotoxicity to a vulnerable population of nigral neurons: implications for Parkinson's disease
    Ayton, S ; Lei, P ; Adlard, PA ; Volitakis, I ; Cherny, RA ; Bush, AI ; Finkelstein, DI (BMC, 2014-07-10)
    BACKGROUND: The substantia nigra (SN) midbrain nucleus is constitutively iron rich. Iron levels elevate further with age, and pathologically in Parkinson's disease (PD). Iron accumulation in PD SN involves dysfunction of ceruloplasmin (CP), which normally promotes iron export. We previously showed that ceruloplasmin knockout (CP KO) mice exhibit Parkinsonian neurodegeneration (~30% nigral loss) by 6 months, which is prevented by iron chelation. Here, we explored whether known iron-stressors of the SN (1) aging and (2) MPTP, would exaggerate the lesion severity of CP KO mice. FINDINGS: We show that while 5 month old CP KO mice exhibited nigral iron elevation and loss of SN neurons, surprisingly, aging CP KO mice to 14 months did not exacerbate iron elevation or SN neuronal loss. Unlike young mice, iron chelation therapy in CP KO mice between 9-14 months did not rescue neuronal loss. MPTP exaggerated iron elevation in young CP KO mice but did not increase cell death when compared to WTs. CONCLUSIONS: We conclude that there may exist a proportion of substantia nigra neurons that depend on CP for protection against iron neurotoxicity and could be protected by iron-based therapeutics. Death of the remaining neurons in Parkinson's disease is likely caused by parallel disease mechanisms, which may call for additional therapeutic options.
  • Item
    Thumbnail Image
    A novel approach to rapidly prevent age-related cognitive decline
    Adlard, PA ; Sedjahtera, A ; Gunawan, L ; Bray, L ; Hare, D ; Lear, J ; Doble, P ; Bush, AI ; Finkelstein, DI ; Cherny, RA (WILEY, 2014-04)
    The loss of cognitive function is a pervasive and often debilitating feature of the aging process for which there are no effective therapeutics. We hypothesized that a novel metal chaperone (PBT2; Prana Biotechnology, Parkville, Victoria, Australia) would enhance cognition in aged rodents. We show here that PBT2 rapidly improves the performance of aged C57Bl/6 mice in the Morris water maze, concomitant with increases in dendritic spine density, hippocampal neuron number and markers of neurogenesis. There were also increased levels of specific glutamate receptors (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and N-methyl-D-aspartate), the glutamate transporter (VGLUT1) and glutamate itself. Markers of synaptic plasticity [calmodulin-dependent protein kinase II (CaMKII) and phosphorylated CaMKII, CREB, synaptophysin] were also increased following PBT2 treatment. We also demonstrate that PBT2 treatment results in a subregion-specific increase in hippocampal zinc, which is increasingly recognized as a potent neuromodulator. These data demonstrate that metal chaperones are a novel approach to the treatment of age-related cognitive decline.
  • Item
    Thumbnail Image
    Mitochondrial Oxidative Stress Causes Hyperphosphorylation of Tau
    Melov, S ; Adlard, PA ; Morten, K ; Johnson, F ; Golden, TR ; Hinerfeld, D ; Schilling, B ; Mavros, C ; Masters, CL ; Volitakis, I ; Li, Q-X ; Laughton, K ; Hubbard, A ; Cherny, RA ; Gibson, B ; Bush, AI ; Khoury, JE (PUBLIC LIBRARY SCIENCE, 2007-06-20)
    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 Alzheimer's disease (AD): tau phosphorylation, and beta-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.
  • Item
    Thumbnail Image
    The effect of dopamine on MPTP-induced rotarod disability
    Ayton, S ; George, JL ; Adlard, PA ; Bush, AI ; Cherny, RA ; Finkelstein, DI (ELSEVIER IRELAND LTD, 2013-05-24)
    UNLABELLED: Dopamine depletion in Parkinson's disease (PD) results in bradykinesia and tremor. Therapeutic administration of the dopamine precursor, l-Dopa, alleviates these symptoms but dyskinesia's can manifest with chronic treatment. In the MPTP toxin mouse model of PD, lesion severity is often assessed by the rotarod behavioral assay. Dopamine depletion by MPTP is thought to induce rotarod behavioral decline. Here we surveyed rotarod behavior and striatal dopamine at timed intervals post-MPTP. Paradoxically, rotarod disability coincided with gradual striatal dopamine restoration. l-Dopa supplementation exacerbated rotarod disability, whereas dopamine antagonism restored performance. CONCLUSION: dopamine restoration, not depletion, precipitates rotarod disability after MPTP intoxication, and caution should be applied when using this assay for MPTP.