Network


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

Hotspot


Dive into the research topics where David Horsley is active.

Publication


Featured researches published by David Horsley.


Journal of Pharmacology and Experimental Therapeutics | 2014

Complex Disposition of Methylthioninium Redox Forms Determines Efficacy in Tau Aggregation Inhibitor Therapy for Alzheimer’s Disease

Thomas C. Baddeley; Jennifer McCaffrey; John M. D. Storey; John Kwok Shing Cheung; Valeria Melis; David Horsley; Charles R. Harrington; Claude M. Wischik

Methylthioninium (MT) is a tau aggregation inhibitor with therapeutic potential in Alzheimer’s disease (AD). MT exists in equilibrium between reduced [leucomethylthioninium (LMT)] and oxidized (MT+) forms; as a chloride salt [methylthioninium chloride (MTC), “methylene blue”], it is stabilized in its MT+ form. Although the results of a phase 2 study of MTC in 321 mild/moderate AD subjects identified a 138-mg MT/day dose as the minimum effective dose on cognitive and imaging end points, further clinical development of MT was delayed pending resolution of the unexpected lack of efficacy of the 228-mg MT/day dose. We hypothesized that the failure of dose response may depend on differences known at the time in dissolution in simulated gastric and intestinal fluids of the 100-mg MTC capsules used to deliver the 228-mg dose and reflect previously unsuspected differences in redox processing of MT at different levels in the gut. The synthesis of a novel chemical entity, LMTX (providing LMT in a stable anhydrous crystalline form), has enabled a systematic comparison of the pharmacokinetic properties of MTC and LMTX in preclinical and clinical studies. The quantity of MT released in water or gastric fluid within 60 minutes proved in retrospect to be an important determinant of clinical efficacy. A further factor was a dose-dependent limitation in the ability to absorb MT in the presence of food when delivered in the MT+ form as MTC. A model is presented to account for the complexity of MT absorption, which may have relevance for other similar redox molecules.


Journal of Biological Chemistry | 2015

Cellular Models of Aggregation-dependent Template-directed Proteolysis to Characterize Tau Aggregation Inhibitors for Treatment of Alzheimer Disease

Charles R. Harrington; John M. D. Storey; Scott Clunas; Kathleen A. Harrington; David Horsley; Ahtsham Ishaq; Steven Kemp; Christopher Paul Larch; Colin Marshall; Sarah Louise Nicoll; Janet Elizabeth Rickard; Michael Simpson; James Peter Sinclair; Lynda J. Storey; Claude M. Wischik

Background: Tau aggregation inhibitors could treat Alzheimer disease. Results: Stable reduced forms of leucomethylthioninium (LMTX®) are active in blocking prion-like Tau aggregation in novel cellular models. Conclusion: The intracellular Ki (0.12 μm) is comparable with brain levels required for clinical benefit. Significance: LMTX® could treat Alzheimer disease. Alzheimer disease (AD) is a degenerative tauopathy characterized by aggregation of Tau protein through the repeat domain to form intraneuronal paired helical filaments (PHFs). We report two cell models in which we control the inherent toxicity of the core Tau fragment. These models demonstrate the properties of prion-like recruitment of full-length Tau into an aggregation pathway in which template-directed, endogenous truncation propagates aggregation through the core Tau binding domain. We use these in combination with dissolution of native PHFs to quantify the activity of Tau aggregation inhibitors (TAIs). We report the synthesis of novel stable crystalline leucomethylthioninium salts (LMTX®), which overcome the pharmacokinetic limitations of methylthioninium chloride. LMTX®, as either a dihydromesylate or a dihydrobromide salt, retains TAI activity in vitro and disrupts PHFs isolated from AD brain tissues at 0.16 μm. The Ki value for intracellular TAI activity, which we have been able to determine for the first time, is 0.12 μm. These values are close to the steady state trough brain concentration of methylthioninium ion (0.18 μm) that is required to arrest progression of AD on clinical and imaging end points and the minimum brain concentration (0.13 μm) required to reverse behavioral deficits and pathology in Tau transgenic mice.


Behavioural Pharmacology | 2015

Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models

Valeria Melis; Mandy Magbagbeolu; Janet Elizabeth Rickard; David Horsley; Kathleen Davidson; Kathleen A. Harrington; Keith Goatman; Elizabeth Anne Goatman; Serena Deiana; Steve P. Close; Claudia Zabke; Karsten Stamer; Silke Dietze; Karima Schwab; John M. D. Storey; Charles R. Harrington; Claude M. Wischik; Franz Theuring; Gernot Riedel

Given the repeated failure of amyloid-based approaches in Alzheimer’s disease, there is increasing interest in tau-based therapeutics. Although methylthioninium (MT) treatment was found to be beneficial in tau transgenic models, the brain concentrations required to inhibit tau aggregation in vivo are unknown. The comparative efficacy of methylthioninium chloride (MTC) and leucomethylthioninium salts (LMTX; 5–75 mg/kg; oral administration for 3–8 weeks) was assessed in two novel transgenic tau mouse lines. Behavioural (spatial water maze, RotaRod motor performance) and histopathological (tau load per brain region) proxies were applied. Both MTC and LMTX dose-dependently rescued the learning impairment and restored behavioural flexibility in a spatial problem-solving water maze task in Line 1 (minimum effective dose: 35 mg MT/kg for MTC, 9 mg MT/kg for LMTX) and corrected motor learning in Line 66 (effective doses: 4 mg MT/kg). Simultaneously, both drugs reduced the number of tau-reactive neurons, particularly in the hippocampus and entorhinal cortex in Line 1 and in a more widespread manner in Line 66. MT levels in the brain followed a sigmoidal concentration–response relationship over a 10-fold range (0.13–1.38 μmol/l). These data establish that diaminophenothiazine compounds, like MT, can reverse both spatial and motor learning deficits and reduce the underlying tau pathology, and therefore offer the potential for treatment of tauopathies.


Cellular and Molecular Life Sciences | 2015

Different pathways of molecular pathophysiology underlie cognitive and motor tauopathy phenotypes in transgenic models for Alzheimer's disease and frontotemporal lobar degeneration.

Valeria Melis; Claudia Zabke; Karsten Stamer; Mandy Magbagbeolu; Karima Schwab; P. Marschall; R. W. Veh; S. Bachmann; S. Deiana; P.-H. Moreau; K. Davidson; Kathleen A. Harrington; Janet Elizabeth Rickard; David Horsley; R. Garman; M. Mazurkiewicz; G. Niewiadomska; Claude M. Wischik; Charles R. Harrington; Gernot Riedel; Franz Theuring

A poorly understood feature of the tauopathies is their very different clinical presentations. The frontotemporal lobar degeneration (FTLD) spectrum is dominated by motor and emotional/psychiatric abnormalities, whereas cognitive and memory deficits are prominent in the early stages of Alzheimer’s disease (AD). We report two novel mouse models overexpressing different human tau protein constructs. One is a full-length tau carrying a double mutation [P301S/G335D; line 66 (L66)] and the second is a truncated 3-repeat tau fragment which constitutes the bulk of the PHF core in AD corresponding to residues 296–390 fused with a signal sequence targeting it to the endoplasmic reticulum membrane (line 1; L1). L66 has abundant tau pathology widely distributed throughout the brain, with particularly high counts of affected neurons in hippocampus and entorhinal cortex. The pathology is neuroanatomically static and declines with age. Behaviourally, the model is devoid of a higher cognitive phenotype but presents with sensorimotor impairments and motor learning phenotypes. L1 displays a much weaker histopathological phenotype, but shows evidence of neuroanatomical spread and amplification with age that resembles the Braak staging of AD. Behaviourally, the model has minimal motor deficits but shows severe cognitive impairments affecting particularly the rodent equivalent of episodic memory which progresses with advancing age. In both models, tau aggregation can be dissociated from abnormal phosphorylation. The two models make possible the demonstration of two distinct but nevertheless convergent pathways of tau molecular pathogenesis. L1 appears to be useful for modelling the cognitive impairment of AD, whereas L66 appears to be more useful for modelling the motor features of the FTLD spectrum. Differences in clinical presentation of AD-like and FTLD syndromes are therefore likely to be inherent to the respective underlying tauopathy, and are not dependent on presence or absence of concomitant APP pathology.


Alzheimers & Dementia | 2008

O1-06-04: Methylthioninium chloride (MTC) acts as a Tau aggregation inhibitor (TAI) in a cellular model and reverses Tau pathology in transgenic mouse models of Alzheimer's disease

Charles R. Harrington; Janet Elizabeth Rickard; David Horsley; Kathleen A. Harrington; Kathleen P. Hindley; Gernot Riedel; Franz Theuring; Kwang Meng Seng; Claude M. Wischik

much is known about different tau kinases, phosphatases and phosphorylation sites the mechanism of neurodegeneration has been elusive. We have previously shown that the cytosolic Alzheimer hyperphosphorylated tau (AD P-tau) sequesters normal tau, MAP1A, MAP1B and MAP2, which results in the inhibition of microtubule assembly and disruption of microtubules. Objective: To study the influence of tau phosphorylation at Ser 199, Thr 212, Thr 231, Ser 262, Ser 396 alone or in combination for tau-tau binding, self-assembly and the effect of its expression on the cells. Methods: Pseudophosphorylated tau was generated by site directed mutagenesis. Pseudophosphorylated tau was transiently expressed in PC12 cells and in CHO cells. The effect of expression of mutated taus on self assembly and binding to microtubules was studied by immunocytochemistry. Binding of normal tau to pseudophosphorylated tau was studied by dot overlay assays. Caspase activation and apoptosis were determined by immunocytochemistry after transient transfection. Results: We found that pseudophosphorylated tau aggregates in cells when Thr 212 is mutated to Glu, suggesting that phosphorylation at this site facilitates tau self-assembly. The expression of tau pseudophosphorylated at Thr212, Thr231, and Ser262 triggers caspase 3 activation in as much as 85% of the transfected cells and apoptosis to a lesser degree. Conclusions: These findings suggest that tau phosphorylation at Thr 212 facilitates tau self-aggregation, and that the combination of phosphorylation at Thr212, 231 and Ser262 in the same tau molecule can trigger toxic reaction. Pseudophosphorylated tau, as Alzheimer phosphorylated tau, sequesters normal tau.


Behavioural Brain Research | 2018

Alpha-Synuclein transgenic mice, h-α-SynL62, display α-Syn aggregation and a dopaminergic phenotype reminiscent of Parkinson's disease

Silke Frahm; Valeria Melis; David Horsley; Janet Elizabeth Rickard; Gernot Riedel; Paula Fadda; Maria Scherma; Charles R. Harrington; Claude M. Wischik; Franz Theuring; Karima Schwab

HIGHLIGHTSTransgenic mice overexpressing human &agr;‐Syn under the control of the mouse Thy1‐promotor.Three transgenic lines express similar levels of &agr;‐Syn mRNA but a different number of cells expressing human &agr;‐syn.L62 has greatest level of aggregated &agr;‐Syn protein throughout brain and spinal cord and most severe motor phenotype.L62 mice display dopaminergic transmission deficits and altered D1 receptor function.L62 presents a model to study motor changes associated with Parkinsons disease. ABSTRACT Alpha‐Synuclein (&agr;‐Syn) accumulation is considered a major risk factor for the development of synucleinopathies such as Parkinsons disease (PD) and dementia with Lewy bodies. We have generated mice overexpressing full‐length human &agr;‐Syn fused to a membrane‐targeting signal sequence under the control of the mouse Thy1‐promotor. Three separate lines (L56, L58 and L62) with similar gene expression levels, but considerably heightened protein accumulation in L58 and L62, were established. In L62, there was widespread labelling of &agr;‐Syn immunoreactivity in brain including spinal cord, basal forebrain, cortex and striatum. Interestingly, there was no detectable &agr;‐Syn expression in dopaminergic neurones of the substantia nigra, but strong human &agr;‐Syn reactivity in glutamatergic synapses. The human &agr;‐Syn accumulated during aging and formed PK‐resistant, thioflavin‐binding aggregates. Mice displayed early onset bradykinesia and age progressive motor deficits. Functional alterations within the striatum were confirmed: L62 showed normal basal dopamine levels, but impaired dopamine release (upon amphetamine challenge) in the dorsal striatum measured by in vivo brain dialysis at 9 months of age. This impairment was coincident with a reduced response to amphetamine in the activity test. L62 further displayed greater sensitivity to low doses of the dopamine receptor 1 (D1) agonist SKF81297 but reacted normally to the D2 agonist quinpirole in the open field. Since accumulation of &agr;‐Syn aggregates in neurones and synapses and alterations in the dopaminergic tone are characteristics of PD, phenotypes reported for L62 present a good opportunity to further our understanding of motor dysfunction in PD and Lewy body dementia.


Frontiers in Molecular Neuroscience | 2018

A Protein Aggregation Inhibitor, Leuco-Methylthioninium Bis(Hydromethanesulfonate), Decreases α-Synuclein Inclusions in a Transgenic Mouse Model of Synucleinopathy

Karima Schwab; Silke Frahm; David Horsley; Janet Elizabeth Rickard; Valeria Melis; Elizabeth Anne Goatman; Mandy Magbagbeolu; Morag Douglas; Michael G. Leith; Thomas C. Baddeley; John M. D. Storey; Gernot Riedel; Claude M. Wischik; Charles R. Harrington; Franz Theuring

α-Synuclein (α-Syn) aggregation is a pathological feature of synucleinopathies, neurodegenerative disorders that include Parkinson’s disease (PD). We have tested whether N,N,N′,N′-tetramethyl-10H-phenothiazine-3,7-diaminium bis(hydromethanesulfonate) (leuco-methylthioninium bis(hydromethanesulfonate); LMTM), a tau aggregation inhibitor, affects α-Syn aggregation in vitro and in vivo. Both cellular and transgenic models in which the expression of full-length human α-Syn (h-α-Syn) fused with a signal sequence peptide to promote α-Syn aggregation were used. Aggregated α-Syn was observed following differentiation of N1E-115 neuroblastoma cells transfected with h-α-Syn. The appearance of aggregated α-Syn was inhibited by LMTM, with an EC50 of 1.1 μM, with minimal effect on h-α-Syn mRNA levels being observed. Two independent lines of mice (L58 and L62) transgenic for the same fusion protein accumulated neuronal h-α-Syn that, with aging, developed into fibrillary inclusions characterized by both resistance to proteinase K (PK)-cleavage and their ability to bind thiazin red. There was a significant decrease in α-Syn-positive neurons in multiple brain regions following oral treatment of male and female mice with LMTM administered daily for 6 weeks at 5 and 15 mg MT/kg. The early aggregates of α-Syn and the late-stage fibrillar inclusions were both susceptible to inhibition by LMTM, a treatment that also resulted in the rescue of movement and anxiety-related traits in these mice. The results suggest that LMTM may provide a potential disease modification therapy in PD and other synucleinopathies through the inhibition of α-Syn aggregation.


Methods of Molecular Biology | 2017

Assays for the Screening and Characterization of Tau Aggregation Inhibitors

Janet Elizabeth Rickard; David Horsley; Claude M. Wischik; Charles R. Harrington

Aggregation of tau protein is a pathological hallmark of Alzheimers disease and other neurodegenerative tauopathies. Inhibition of tau aggregation could provide a method for the treatment of these disorders. Methods to identify tau aggregation inhibitors (TAIs) in vitro are useful and here we describe assays for TAIs using purified recombinant tau protein fragments in a cell-free immunoassay format and a stably transfected cell model to create a more physiological environment.


Archive | 2002

Materials and methods relating to protein aggregation in neurodegenerative disease

Claude Michel Wischik; Janet Elizabeth Rickard; David Horsley; Charles Robert Harrington; Franz Theuring; Karsten Stamer; Claudia Zabke


Archive | 2002

Napthoquinone derivatives as inhibitors of tau aggregation for the treatment of alzheimer's and related neurodegenerative disorders

Claude M. Wischik; David Horsley; Janet Elizabeth Rickard; Charles R. Harrington

Collaboration


Dive into the David Horsley's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge