Network


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

Hotspot


Dive into the research topics where Claire O'Leary is active.

Publication


Featured researches published by Claire O'Leary.


The International Journal of Neuropsychopharmacology | 2012

Genetic vs. pharmacological inactivation of COMT influences cannabinoid-induced expression of schizophrenia-related phenotypes

Colm M.P. O'Tuathaigh; Gerard Clarke; Jeremy Walsh; Lieve Desbonnet; Emilie Petit; Claire O'Leary; Orna Tighe; Niamh Clarke; Maria Karayiorgou; Joseph A. Gogos; T.G. Dinan; John F. Cryan; John L. Waddington

Catechol-O-methyltransferase (COMT) is an important enzyme in the metabolism of dopamine and disturbance in dopamine function is proposed to be central to the pathogenesis of schizophrenia. Clinical epidemiological studies have indicated cannabis use to confer a 2-fold increase in risk for subsequent onset of psychosis, with adolescent-onset use conveying even higher risk. There is evidence that a high activity COMT polymorphism moderates the effects of adolescent exposure to cannabis on risk for adult psychosis. In this paper we compared the effect of chronic adolescent exposure to the cannabinoid WIN 55212 on sensorimotor gating, behaviours related to the negative symptoms of schizophrenia, anxiety- and stress-related behaviours, as well as ex-vivo brain dopamine and serotonin levels, in COMT KO vs. wild-type (WT) mice. Additionally, we examined the effect of pretreatment with the COMT inhibitor tolcapone on acute effects of this cannabinoid on sensorimotor gating in C57BL/6 mice. COMT KO mice were shown to be more vulnerable than WT to the disruptive effects of adolescent cannabinoid treatment on prepulse inhibition (PPI). Acute pharmacological inhibition of COMT in C57BL/6 mice also modified acute cannabinoid effects on startle reactivity, as well as PPI, indicating that chronic and acute loss of COMT can produce dissociable effects on the behavioural effects of cannabinoids. COMT KO mice also demonstrated differential effects of adolescent cannabinoid administration on sociability and anxiety-related behaviour, both confirming and extending earlier reports of COMT×cannabinoid effects on the expression of schizophrenia-related endophenotypes.


Neurobiology of Disease | 2014

Resolving pathobiological mechanisms relating to Huntington disease: gait, balance, and involuntary movements in mice with targeted ablation of striatal D1 dopamine receptor cells.

Hyun Ah. Kim; Luning Jiang; Heather B. Madsen; Clare L. Parish; Jim S. Massalas; Arthur Smardencas; Claire O'Leary; Ilse Gantois; Colm M.P. O'Tuathaigh; John L. Waddington; Michelle E. Ehrlich; Andrew J. Lawrence; John Drago

Progressive cell loss is observed in the striatum, cerebral cortex, thalamus, hypothalamus, subthalamic nucleus and hippocampus in Huntington disease. In the striatum, dopamine-responsive medium spiny neurons are preferentially lost. Clinical features include involuntary movements, gait and orofacial impairments in addition to cognitive deficits and psychosis, anxiety and mood disorders. We utilized the Cre-LoxP system to generate mutant mice with selective postnatal ablation of D1 dopamine receptor-expressing striatal neurons to determine which elements of the complex Huntington disease phenotype relate to loss of this neuronal subpopulation. Mutant mice had reduced body weight, locomotor slowing, reduced rearing, ataxia, a short stride length wide-based erratic gait, impairment in orofacial movements and displayed haloperidol-suppressible tic-like movements. The mutation was associated with an anxiolytic profile. Mutant mice had significant striatal-specific atrophy and astrogliosis. D1-expressing cell number was reduced throughout the rostrocaudal extent of the dorsal striatum consistent with partial destruction of the striatonigral pathway. Additional striatal changes included up-regulated D2 and enkephalin mRNA, and an increased density of D2 and preproenkephalin-expressing projection neurons, and striatal neuropeptide Y and cholinergic interneurons. These data suggest that striatal D1-cell-ablation alone may account for the involuntary movements and locomotor, balance and orofacial deficits seen not only in HD but also in HD phenocopy syndromes with striatal atrophy. Therapeutic strategies would therefore need to target striatal D1 cells to ameliorate deficits especially when the clinical presentation is dominated by a bradykinetic/ataxic phenotype with involuntary movements.


Schizophrenia Bulletin | 2017

Epistatic and Independent Effects on Schizophrenia-Related Phenotypes Following Co-disruption of the Risk Factors Neuregulin-1 × DISC1

Colm M.P. O'Tuathaigh; Fabio Fumagalli; Lieve Desbonnet; Francesc Perez-Branguli; Gerard Moloney; Samim Loftus; Claire O'Leary; Emilie Petit; Rachel F. Cox; Orna Tighe; Gerard Clarke; Donna Lai; Richard P. Harvey; John F. Cryan; Kevin J. Mitchell; Timothy G. Dinan; Marco Riva; John L. Waddington

Few studies have addressed likely gene × gene (ie, epistatic) interactions in mediating risk for schizophrenia. Using a preclinical genetic approach, we investigated whether simultaneous disruption of the risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) would produce a disease-relevant phenotypic profile different from that observed following disruption to either gene alone. NRG1 heterozygotes exhibited hyperactivity and disruption to prepulse inhibition, both reversed by antipsychotic treatment, and accompanied by reduced striatal dopamine D2 receptor protein expression, impaired social cognition, and altered glutamatergic synaptic protein expression in selected brain areas. Single gene DISC1 mutants demonstrated a disruption in social cognition and nest-building, altered brain 5-hydroxytryptamine levels and hippocampal ErbB4 expression, and decreased cortical expression of the schizophrenia-associated microRNA miR-29b. Co-disruption of DISC1 and NRG1, indicative of epistasis, evoked an impairment in sociability and enhanced self-grooming, accompanied by changes in hypothalamic oxytocin/vasopressin gene expression. The findings indicate specific behavioral correlates and underlying cellular pathways downstream of main effects of DNA variation in the schizophrenia-associated genes NRG1 and DISC1.


Brain | 2018

Macrophage enzyme and reduced inflammation drive brain correction of mucopolysaccharidosis IIIB by stem cell gene therapy

Rebecca J. Holley; Stuart M. Ellison; Daniel Fil; Claire O'Leary; John McDermott; Nishanthi Senthivel; Alex Langford-Smith; Fiona Wilkinson; Zelpha D'Souza; Helen Parker; Aiyin Liao; Samuel Rowlston; Hélène F.E. Gleitz; Shih hsin Kan; Patricia Dickson; Brian Bigger

Mucopolysaccharidosis IIIB is a paediatric lysosomal storage disease caused by deficiency of the enzyme α-N-acetylglucosaminidase (NAGLU), involved in the degradation of the glycosaminoglycan heparan sulphate. Absence of NAGLU leads to accumulation of partially degraded heparan sulphate within lysosomes and the extracellular matrix, giving rise to severe CNS degeneration with progressive cognitive impairment and behavioural problems. There are no therapies. Haematopoietic stem cell transplant shows great efficacy in the related disease mucopolysaccharidosis I, where donor-derived monocytes can transmigrate into the brain following bone marrow engraftment, secrete the missing enzyme and cross-correct neighbouring cells. However, little neurological correction is achieved in patients with mucopolysaccharidosis IIIB. We have therefore developed an ex vivo haematopoietic stem cell gene therapy approach in a mouse model of mucopolysaccharidosis IIIB, using a high-titre lentiviral vector and the myeloid-specific CD11b promoter, driving the expression of NAGLU (LV.NAGLU). To understand the mechanism of correction we also compared this with a poorly secreted version of NAGLU containing a C-terminal fusion to IGFII (LV.NAGLU-IGFII). Mucopolysaccharidosis IIIB haematopoietic stem cells were transduced with vector, transplanted into myeloablated mucopolysaccharidosis IIIB mice and compared at 8 months of age with mice receiving a wild-type transplant. As the disease is characterized by increased inflammation, we also tested the anti-inflammatory steroidal agent prednisolone alone, or in combination with LV.NAGLU, to understand the importance of inflammation on behaviour. NAGLU enzyme was substantially increased in the brain of LV.NAGLU and LV.NAGLU-IGFII-treated mice, with little expression in wild-type bone marrow transplanted mice. LV.NAGLU treatment led to behavioural correction, normalization of heparan sulphate and sulphation patterning, reduced inflammatory cytokine expression and correction of astrocytosis, microgliosis and lysosomal compartment size throughout the brain. The addition of prednisolone improved inflammatory aspects further. Substantial correction of lysosomal storage in neurons and astrocytes was also achieved in LV.NAGLU-IGFII-treated mice, despite limited enzyme secretion from engrafted macrophages in the brain. Interestingly both wild-type bone marrow transplant and prednisolone treatment alone corrected behaviour, despite having little effect on brain neuropathology. This was attributed to a decrease in peripheral inflammatory cytokines. Here we show significant neurological disease correction is achieved using haematopoietic stem cell gene therapy, suggesting this therapy alone or in combination with anti-inflammatories may improve neurological function in patients.


Neurobiology of Disease | 2015

Motor and behavioral phenotype in conditional mutants with targeted ablation of cortical D1 dopamine receptor-expressing cells

Luning Jiang; Claire O'Leary; Hyun Ah Kim; Clare L. Parish; Jim S. Massalas; John L. Waddington; Michelle E. Ehrlich; Gunter M. Schütz; Ilse Gantois; Andrew J. Lawrence; John Drago

D1-dopamine receptors (Drd1a) are highly expressed in the deep layers of the cerebral cortex and the striatum. A number of human diseases such as Huntington disease and schizophrenia are known to have cortical pathology involving dopamine receptor expressing neurons. To illuminate their functional role, we exploited a Cre/Lox molecular paradigm to generate Emx-1(tox) MUT mice, a transgenic line in which cortical Drd1a-expressing pyramidal neurons were selectively ablated. Emx-1(tox) MUT mice displayed prominent forelimb dystonia, hyperkinesia, ataxia on rotarod testing, heightened anxiety-like behavior, and age-dependent abnormalities in a test of social interaction. The latter occurred in the context of normal working memory on testing in the Y-maze and for novel object recognition. Some motor and behavioral abnormalities in Emx-1(tox) MUT mice overlapped with those in CamKIIα(tox) MUT transgenic mice, a line in which both striatal and cortical Drd1a-expressing cells were ablated. Although Emx-1(tox) MUT mice had normal striatal anatomy, both Emx-1(tox) MUT and CamKIIα(tox) MUT mice displayed selective neuronal loss in cortical layers V and VI. This study shows that loss of cortical Drd1a-expressing cells is sufficient to produce deficits in multiple motor and behavioral domains, independent of striatal mechanisms. Primary cortical changes in the D1 dopamine receptor compartment are therefore likely to model a number of core clinical features in disorders such as Huntington disease and schizophrenia.


PLOS ONE | 2017

Identification of Age-Dependent Motor and Neuropsychological Behavioural Abnormalities in a Mouse Model of Mucopolysaccharidosis Type II

Hélène F.E. Gleitz; Claire O'Leary; Rebecca J. Holley; Brian Bigger

Severe mucopolysaccharidosis type II (MPS II) is a progressive lysosomal storage disease caused by mutations in the IDS gene, leading to a deficiency in the iduronate-2-sulfatase enzyme that is involved in heparan sulphate and dermatan sulphate catabolism. In constitutive form, MPS II is a multi-system disease characterised by progressive neurocognitive decline, severe skeletal abnormalities and hepatosplenomegaly. Although enzyme replacement therapy has been approved for treatment of peripheral organs, no therapy effectively treats the cognitive symptoms of the disease and novel therapies are in development to remediate this. Therapeutic efficacy and subsequent validation can be assessed using a variety of outcome measures that are translatable to clinical practice, such as behavioural measures. We sought to consolidate current knowledge of the cognitive, skeletal and motor abnormalities present in the MPS II mouse model by performing time course behavioural examinations of working memory, anxiety, activity levels, sociability and coordination and balance, up to 8 months of age. Cognitive decline associated with alterations in spatial working memory is detectable at 8 months of age in MPS II mice using spontaneous alternation, together with an altered response to novel environments and anxiolytic behaviour in the open-field. Coordination and balance on the accelerating rotarod were also significantly worse at 8 months, and may be associated with skeletal changes seen in MPS II mice. We demonstrate that the progressive nature of MPS II disease is also seen in the mouse model, and that cognitive and motor differences are detectable at 8 months of age using spontaneous alternation, the accelerating rotarod and the open-field tests. This study establishes neurological, motor and skeletal measures for use in pre-clinical studies to develop therapeutic approaches in MPS II.


BMC Neuroscience | 2013

Phenotyping dividing cells in mouse models of neurodegenerative basal ganglia diseases

Arthur Smardencas; Kerelos Rizkalla; Hyun Ah Kim; Jim S. Massalas; Claire O'Leary; Michelle E. Ehrlich; Günther Schütz; Andrew J. Lawrence; John Drago

BackgroundMice generated by a Cre/LoxP transgenic paradigm were used to model neurodegenerative basal ganglia disease of which Huntington disease (HD) is the prototypical example. In HD, death occurs in striatal projection neurons as well as cortical neurons. Cortical and striatal neurons that express the D1 dopamine receptor (Drd1a) degenerate in HD. The contribution that death of specific neuronal cell populations makes to the HD disease phenotype and the response of the brain to loss of defined cell subtypes is largely unknown.MethodsDrd1a-expressing cells were targeted for cell death and three independent lines generated; a striatal-restricted line, a cortical-restricted line and a global line in which Drd1a cells were deleted from both the striatum and cortex. Two independent experimental approaches were used. In the first, the proliferative marker Ki-67 was used to identify proliferating cells in eighty-week-old mice belonging to a generic global line, a global in which Drd1a cells express green fluorescent protein (GFP-global) and in eighty-week-old mice of a cortical line. In the second experiment, the proliferative response of four-week-old mice belonging to GFP-global and striatal lines was assessed using the thymidine analogue BrdU. The phenotype of proliferating cells was ascertained by double staining for BrdU and Olig2 (an oligodendrocyte marker), Iba1 (a microglial cell marker), S100β (an astroglial cell marker), or NeuN (a neuronal cell marker).ResultsIn the first study, we found that Ki-67-expressing cells were restricted to the striatal side of the lateral ventricles. Control mice had a greater number of Ki-67+ cells than mutant mice. There was no overlap between Ki-67 and GFP staining in control or mutant mice, suggesting that cells did not undergo cell division once they acquired a Drd1a phenotype. In contrast, in the second study we found that BrdU+ cells were identified throughout the cortex, striatum and periventricular region of control and mutant mice. Mutant mice from the GFP-global line showed increased BrdU+ cells in the cortex, striatum and periventricular region relative to control. Striatal line mutant mice had an increased number of BrdU+ cells in the striatum and periventricular region, but not the cortex. The number of microglia, astrocytes, oligodendrocytes and neurons generated from dividing progenitors was increased relative to control mice in most brain regions in mutant mice from the GFP-global line. In contrast, striatal line mutant mice displayed an increase only in the number of dividing microglia in striatal and periventricular regions.ConclusionsGenetically programmed post-natal ablation of Drd1a-expressing neurons is associated with an extensive proliferative response involving multiple cell lineages. The nature of the tissue response has the potential not only to remove cellular debris but also to forge physiologically meaningful brain repair. Age related deficits in proliferation are seen in mutant lines. A blunted endogenous reparative response may underlie the cumulative deficits characteristic of age related neurodegeneration.


Embo Molecular Medicine | 2018

Brain‐targeted stem cell gene therapy corrects mucopolysaccharidosis type II via multiple mechanisms

Hélène F.E. Gleitz; Ai Yin Liao; James R Cook; Samuel Rowlston; Gabriella M.A. Forte; Zelpha D'Souza; Claire O'Leary; Rebecca J. Holley; Brian Bigger

The pediatric lysosomal storage disorder mucopolysaccharidosis type II is caused by mutations in IDS, resulting in accumulation of heparan and dermatan sulfate, causing severe neurodegeneration, skeletal disease, and cardiorespiratory disease. Most patients manifest with cognitive symptoms, which cannot be treated with enzyme replacement therapy, as native IDS does not cross the blood–brain barrier. We tested a brain‐targeted hematopoietic stem cell gene therapy approach using lentiviral IDS fused to ApoEII (IDS.ApoEII) compared to a lentivirus expressing normal IDS or a normal bone marrow transplant. In mucopolysaccharidosis II mice, all treatments corrected peripheral disease, but only IDS.ApoEII mediated complete normalization of brain pathology and behavior, providing significantly enhanced correction compared to IDS. A normal bone marrow transplant achieved no brain correction. Whilst corrected macrophages traffic to the brain, secreting IDS/IDS.ApoEII enzyme for cross‐correction, IDS.ApoEII was additionally more active in plasma and was taken up and transcytosed across brain endothelia significantly better than IDS via both heparan sulfate/ApoE‐dependent receptors and mannose‐6‐phosphate receptors. Brain‐targeted hematopoietic stem cell gene therapy provides a promising therapy for MPS II patients.


Neuropathology and Applied Neurobiology | 2018

A non-myeloablative chimeric mouse model accurately defines microglia and macrophage contribution in glioma

Kenny Yu; Amir Saam Youshani; Fiona Wilkinson; Claire O'Leary; Peter C. Cook; L. Laaniste; Aiyin Liao; D. Mosses; C. Waugh; Hannah Shorrock; Omar Pathmanaban; Andrew S. MacDonald; Ian Kamaly-Asl; Federico Roncaroli; Brian Bigger

Resident and peripherally derived glioma associated microglia/macrophages (GAMM) play a key role in driving tumour progression, angiogenesis, invasion and attenuating host immune responses. Differentiating these cells’ origins is challenging and current preclinical models such as irradiation‐based adoptive transfer, parabiosis and transgenic mice have limitations. We aimed to develop a novel nonmyeloablative transplantation (NMT) mouse model that permits high levels of peripheral chimerism without blood‐brain barrier (BBB) damage or brain infiltration prior to tumour implantation.


The Lancet | 2016

Characterisation of central versus peripheral tumour associated macrophages in glioblastoma multiforme

Amir Saam Youshani; Kenny Yu; Claire O'Leary; Omar Pathmanaban; Ian Kamaly-Asl; Brian Bigger

Abstract Background Glioblastoma multiforme is the most biologically aggressive brain tumour, with only 12 month survival, despite maximum treatment. Tumour-associated macrophages and microglia (TAMM) are myeloid cells populating 30–50% of glioblastoma multiforme tumours and are identified as potential immunotherapy targets. However, characterising functional roles of TAMM subpopulations remains elusive, because of flawed mouse models and inadequate cell-surface markers. In normal rodent brain, antibody markers CD11bhi and CD45hi are proposed to highlight infiltrating monocytes, whereas CD11bhi and CD45lo cells represent microglia and differentiated peripheral macrophages. This marker-set provides conflicting results in the context of the tumour microenvironment. We redressed this issue and aimed to determine whether peripheral or central TAMMs have independent roles in glioblastoma multiforme. Methods With our non-myeloablative bone-marrow transplant model, we can replace a mouse immune system using congenic donor cells marked with green fluorescence protein (GFP) without damaging the blood-brain barrier or inadvertently activating microglia; when applying this technique we subsequently distinguish between central microglial and peripheral macrophage populations. By stereotaxic intracranial implantation of murine glioblastoma multiforme (GL261), we recapitulated the normal development of a glioblastoma multiforme tumour and performed sham intracranial injections as a comparative control. Fluorescence-activated cell sorting was done to separate TAMM subpopulations for RNA sequencing. Findings Our chimeric mouse model consistently produced 70–80% peripheral blood chimerism and overcame the issue surrounding cell-surface expression markers, distinguishing peripherally infiltrating and central tissue-resident brain cells by GFP marker status. Cerebral digestion and analysis of tumour-implanted mice revealed a novel third population (FSChi/SSChi cells) on flow cytometry that stained greater than 90% CD11b/CD45+ suggestive of TAMMs; but this cell population was absent in hemispheres of both non-tumour bearing and sham-injected mice. Interpretation Our preliminary data showed that in chimeric mice implanted with glioblastoma multiforme, peripheral cells (GFP high) were the dominant infiltrating population. With RNA extraction for downstream transcriptional analysis we aim to reliably track trafficking myeloid populations and subsequently address the ambiguity surrounding TAMMs. Thereafter, we aim to develop an immunotherapy against glioblastoma multiforme using the findings from our study. Funding None.

Collaboration


Dive into the Claire O'Leary's collaboration.

Top Co-Authors

Avatar

Brian Bigger

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John L. Waddington

Royal College of Surgeons in Ireland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fiona Wilkinson

Manchester Metropolitan University

View shared research outputs
Top Co-Authors

Avatar

Helen Parker

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

John Drago

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ai Yin Liao

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge