Network


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

Hotspot


Dive into the research topics where J. Mark Cooper is active.

Publication


Featured researches published by J. Mark Cooper.


Human Molecular Genetics | 2010

Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy

Matthew E. Gegg; J. Mark Cooper; Kai-Yin Chau; Manuel Rojo; A. H. V. Schapira; Jan-Willem Taanman

Mitochondrial dysfunction and perturbed degradation of proteins have been implicated in Parkinsons disease (PD) pathogenesis. Mutations in the Parkin and PINK1 genes are a cause of familial PD. PINK1 is a putative kinase associated with mitochondria, and loss of PINK1 expression leads to mitochondrial dysfunction, which increases with time. Parkin is suggested to be downstream of PINK1 and also mediates the removal of damaged mitochondria by macroautophagy (mitophagy). We investigated whether mitochondrial dysfunction in dopaminergic SH-SY5Y cells following decreased PINK1 expression by RNAi may in part be due to the inhibition of mitophagy. Reduced flux through the macroautophagy pathway was found to be coincident with the inhibition of ATP synthesis following 12 days of PINK1 silencing. Overexpression of parkin in these cells restored both autophagic flux and ATP synthesis. Overexpression and RNAi studies also indicated that PINK1 and parkin were required for mitophagy following CCCP-induced mitochondrial damage. The ubiquitination of several mitochondrial proteins, including mitofusin 1 and mitofusin 2, were detected within 3 h of CCCP treatment. These post-translational modifications were reduced following the silencing of parkin or PINK1. The ubiquitination of mitochondrial proteins appears to identify mitochondria for degradation and facilitate mitophagy. PINK1 and parkin are thus required for the removal of damaged mitochondria in dopaminergic cells, and inhibition of this pathway may lead to the accumulation of defective mitochondria which may contribute to PD pathogenesis.


Annals of Neurology | 2001

Antioxidant treatment improves in vivo cardiac and skeletal muscle bioenergetics in patients with friedreich's ataxia

Raffaele Lodi; Paul E. Hart; Bheeshma Rajagopalan; Doris J. Taylor; Jenifer G. Crilley; Jane L. Bradley; Andrew M. Blamire; David Neil Manners; Peter Styles; A. H. V. Schapira; J. Mark Cooper

Friedreichs ataxia (FA) is the most common form of autosomal recessive spinocerebellar ataxia and is often associated with a cardiomyopathy. The disease is caused by an expanded intronic GAA repeat, which results in deficiency of a mitochondrial protein called frataxin. In the yeast YFH1 knockout model of the disease there is evidence that frataxin deficiency leads to a severe defect of mitochondrial respiration, intramitochondrial iron accumulation, and associated production of oxygen free radicals. Recently, the analysis of FA cardiac and skeletal muscle samples and in vivo phosphorus magnetic resonance spectroscopy (31P‐MRS) has confirmed the deficits of respiratory chain complexes in these tissues. The role of oxidative stress in FA is further supported by the accumulation of iron and decreased aconitase activities in cardiac muscle. We used 31P‐MRS to evaluate the effect of 6 months of antioxidant treatment (Coenzyme Q10 400 mg/day, vitamin E 2,100 IU/day) on cardiac and calf muscle energy metabolism in 10 FA patients. After only 3 months of treatment, the cardiac phosphocreatine to ATP ratio showed a mean relative increase to 178% (p = 0.03) and the maximum rate of skeletal muscle mitochondrial ATP production increased to 139% (p = 0.01) of their respective baseline values in the FA patients. These improvements, greater in prehypertrophic hearts and in the muscle of patients with longer GAA repeats, were sustained after 6 months of therapy. The neurological and echocardiographic evaluations did not show any consistent benefits of the therapy after 6 months. This study demonstrates partial reversal of a surrogate biochemical marker in FA with antioxidant therapy and supports the evaluation of such therapy as a disease‐modifying strategy in this neurodegenerative disorder.


Brain | 2014

Ambroxol improves lysosomal biochemistry in glucocerebrosidase mutation-linked Parkinson disease cells

Alisdair McNeill; Joana Magalhaes; Chengguo Shen; Kai-Yin Chau; Derralyn Hughes; Atul Mehta; Thomas Foltynie; J. Mark Cooper; Andrey Y. Abramov; Matthew E. Gegg; A. H. V. Schapira

Heterozygous GBA gene mutations are the most frequent Parkinson’s disease risk factor. Using Parkinson’s disease patient derived fibroblasts McNeill et al. show that heterozygous GBA mutations reduce glucosylceramidase activity, and are associated with endoplasmic reticulum and oxidative stress. Ambroxol treatment improved glucosylceramidase activity and reduced oxidative stress in these cells.


American Journal of Human Genetics | 1999

A Missense Mutation of Cytochrome Oxidase Subunit II Causes Defective Assembly and Myopathy

Shamima Rahman; Jan-Willem Taanman; J. Mark Cooper; I. Nelson; Ian Hargreaves; Brigitte Meunier; Michael G. Hanna; José J. Garcı́a; Roderick A. Capaldi; Brian D. Lake; J. V. Leonard; A. H. V. Schapira

We report the first missense mutation in the mtDNA gene for subunit II of cytochrome c oxidase (COX). The mutation was identified in a 14-year-old boy with a proximal myopathy and lactic acidosis. Muscle histochemistry and mitochondrial respiratory-chain enzymology demonstrated a marked reduction in COX activity. Immunohistochemistry and immunoblot analyses with COX subunit-specific monoclonal antibodies showed a pattern suggestive of a primary mtDNA defect, most likely involving CO II, for COX subunit II (COX II). mtDNA-sequence analysis demonstrated a novel heteroplasmic T-->A transversion at nucleotide position 7,671 in CO II. This mutation changes a methionine to a lysine residue in the middle of the first N-terminal membrane-spanning region of COX II. The immunoblot studies demonstrated a severe reduction in cross-reactivity, not only for COX II but also for the mtDNA-encoded subunit COX III and for nuclear-encoded subunits Vb, VIa, VIb, and VIc. Steady-state levels of the mtDNA-encoded subunit COX I showed a mild reduction, but spectrophotometric analysis revealed a dramatic decrease in COX I-associated heme a3 levels. These observations suggest that, in the COX protein, a structural association of COX II with COX I is necessary to stabilize the binding of heme a3 to COX I.


Journal of Neurochemistry | 2004

Pramipexole protects against apoptotic cell death by non-dopaminergic mechanisms

M Gu; M. Irvani; J. Mark Cooper; Diane King; Peter Jenner; A. H. V. Schapira

We have investigated the ability of pramipexole, a dopamine agonist used in the symptomatic treatment of Parkinsons disease (PD), to protect against cell death induced by 1‐methyl‐4‐phenylpyridinium (MPP+) and rotenone in dopaminergic and non‐dopaminergic cells. Pre‐incubation with either the active (–)‐ or inactive (+)‐enantiomer forms of pramipexole (10 μm) decreased cell death in response to MPP+ and rotenone in dopaminergic SHSY‐5Y cells and in non‐dopaminergic JK cells. The protective effect was not prevented by dopamine receptor blockade using sulpiride or clozapine. Protection occurred at concentrations at which pramipexole did not demonstrate antioxidant activity, as shown by the failure to maintain aconitase activity. However, pramipexole reduced caspase‐3 activation, decreased the release of cytochrome c and prevented the fall in the mitochondrial membrane potential induced by MPP+ and rotenone. This suggests that pramipexole has anti‐apoptotic actions. The results extend the evidence for the neuroprotective effects of pramipexole and indicate that this is not dependent on dopamine receptor occupation or antioxidant activity. Further evaluation is required to determine whether the neuroprotective action of pramipexole is translated to a disease‐modifying effect in PD patients.


Neuroscience Research | 2011

Alpha-synuclein release by neurons activates the inflammatory response in a microglial cell line

Lydia Alvarez-Erviti; Yvonne Couch; Jill C. Richardson; J. Mark Cooper; Matthew J.A. Wood

The neurodegenerative process in Parkinsons disease (PD) is accompanied by the presence of a neuroinflammatory response, which has been suggested as one of the principal components involved in PD progression. In this report we assessed the inflammatory potential of alpha-synuclein, a protein central to PD pathogenesis, released by neurons on the mouse microglia cell line BV-2. BV-2 cells were treated with conditioned medium isolated from normal SH-SY5Y cells and clones that over-express WT or mutant A53T alpha-synuclein. Conditioned medium isolated from over-expressing clones induced the transcription and release of pro-inflammatory cytokines. Treatment of SH-SY5Y alpha-synuclein over-expressing cells with MPP+, the active metabolite of the neurotoxin MPTP, increased the inflammatory response in BV-2 cells. In contrast, the direct exposure of BV-2 cells to MPP+ failed to induce an inflammatory response. These results support the hypothesis that WT and A53T alpha-synuclein has an important role in the initiation and maintenance of inflammation in PD, through the activation of a pro-inflammatory response in microglial cells.


Human Molecular Genetics | 2012

G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization

Tatiana D. Papkovskaia; Kai-Yin Chau; Francisco Inesta-Vaquera; Dmitri B. Papkovsky; Daniel G. Healy; Koji Nishio; James Michael Staddon; Michael R. Duchen; John Hardy; A. H. V. Schapira; J. Mark Cooper

The G2019S leucine rich repeat kinase 2 (LRRK2) mutation is the most common genetic cause of Parkinsons disease (PD), clinically and pathologically indistinguishable from idiopathic PD. Mitochondrial abnormalities are a common feature in PD pathogenesis and we have investigated the impact of G2019S mutant LRRK2 expression on mitochondrial bioenergetics. LRRK2 protein expression was detected in fibroblasts and lymphoblasts at levels higher than those observed in the mouse brain. The presence of G2019S LRRK2 mutation did not influence LRRK2 expression in fibroblasts. However, the expression of the G2019S LRRK2 mutation in both fibroblast and neuroblastoma cells was associated with mitochondrial uncoupling. This was characterized by decreased mitochondrial membrane potential and increased oxygen utilization under basal and oligomycin-inhibited conditions. This resulted in a decrease in cellular ATP levels consistent with compromised cellular function. This uncoupling of mitochondrial oxidative phosphorylation was associated with a cell-specific increase in uncoupling protein (UCP) 2 and 4 expression. Restoration of mitochondrial membrane potential by the UCP inhibitor genipin confirmed the role of UCPs in this mechanism. The G2019S LRRK2-induced mitochondrial uncoupling and UCP4 mRNA up-regulation were LRRK2 kinase-dependent, whereas endogenous LRRK2 levels were required for constitutive UCP expression. We propose that normal mitochondrial function was deregulated by the expression of G2019S LRRK2 in a kinase-dependent mechanism that is a modification of the normal LRRK2 function, and this leads to the vulnerability of selected neuronal populations in PD.


Journal of Hepatology | 1998

Liver failure associated with mitochondrial DNA depletion

A. A. M. Morris; Jan-Willem Taanman; Julian Blake; J. Mark Cooper; Brian D. Lake; Marion Malone; Seth Love; Peter Clayton; J. V. Leonard; A. H. V. Schapira

BACKGROUND/AIMS Liver failure in infancy can result from several disorders of the mitochondrial respiratory chain. In some patients, levels of mitochondrial DNA are markedly reduced, a phenomenon referred to as mitochondrial DNA depletion. To facilitate diagnosis of this condition, we have reviewed the clinical and pathological features in five patients with mitochondrial DNA depletion. METHODS Cases were identified by preparing Southern blots of DNA from muscle and liver, hybridising with appropriate probes and quantifying mitochondrial DNA relative to nuclear DNA. RESULTS All our patients with mitochondrial DNA depletion died of liver failure. Other problems included hypotonia, hypoglycaemia, neurological abnormalities (including Leigh syndrome) and cataracts. Liver histology showed geographic areas of fatty change, bile duct proliferation, collapse of liver architecture and fibrosis; some cells showed decreased cytochrome oxidase activity. Muscle from three patients showed mitochondrial proliferation, with loss of cytochrome oxidase activity in some fibres but not in others; in these cases, muscle mitochondrial DNA levels were less than 5% of the median control value. The remaining two patients (from a single pedigree) had normal muscle histology and histochemistry associated with less severe depletion of mitochondrial DNA in muscle. CONCLUSIONS Liver failure is common in patients with mitochondrial DNA depletion. Associated clinical features often include neuromuscular disease. Liver and muscle histology can be helpful in making the diagnosis. Mitochondrial DNA levels should be measured whenever liver failure is thought to have resulted from respiratory chain disease.


Neurochemistry International | 2013

Glucocerebrosidase inhibition causes mitochondrial dysfunction and free radical damage

Michael W.J. Cleeter; Kai-Yin Chau; Caroline Gluck; Atul Mehta; Derralynn Hughes; Michael R. Duchen; Nicholas W. Wood; John Hardy; J. Mark Cooper; A Schapira

Highlights ► Glucocerebrosidase gene mutations are a risk factor for Parkinson’s disease. ► Glucocerebrosidase inhibition causes mitochondrial dysfunction & oxidative stress. ► These changes parallel important pathogenetic of Parkinson’s disease.


Annals of Neurology | 2006

Proteasomal inhibition causes loss of nigral tyrosine hydroxylase neurons

A. H. V. Schapira; Michael W.J. Cleeter; John R. Muddle; Jane M. Workman; J. Mark Cooper; R. H. M. King

Dysfunction of the ubiquitin‐proteasomal system (UPS) has been implicated in the pathogenesis of Parkinsons disease. The systemic administration of UPS inhibitors has been reported to induce nigrostriatal cell death and model Parkinsons disease pathology in rodents. We administered a synthetic, specific UPS inhibitor (PSI) subcutaneously to rats and quantified substantia nigral tyrosine hydroxylase–positive dopaminergic neurons by stereology. PSI caused a 15% decrease in UPS activity at 2 weeks and a 42% reduction in substantia nigra pars compacta tyrosine hydroxylase–positive neurons at 8 weeks. Systemic inhibition of the UPS warrants further evaluation as a means to model Parkinsons disease. Ann Neurol 2006;60:253–255

Collaboration


Dive into the J. Mark Cooper's collaboration.

Top Co-Authors

Avatar

A. H. V. Schapira

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kai-Yin Chau

University College London

View shared research outputs
Top Co-Authors

Avatar

Jane L. Bradley

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul E. Hart

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. V. Leonard

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julian Blake

Norfolk and Norwich University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge