Network


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

Hotspot


Dive into the research topics where Heather L. Melrose is active.

Publication


Featured researches published by Heather L. Melrose.


Nature Genetics | 2000

Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein.

Jada Lewis; Eileen McGowan; Julia Rockwood; Heather L. Melrose; Parimala Nacharaju; Marjon Van Slegtenhorst; Katrina Gwinn-Hardy; Michael P. Murphy; Matt Baker; Xin Yu; Karen Duff; John Hardy; Anthony Corral; Wen Lang Lin; Shu Hui Yen; Dennis W. Dickson; Peter Davies; Mike Hutton

Neurofibrillary tangles (NFT) composed of the microtubule-associated protein tau are prominent in Alzheimer disease (AD), Pick disease, progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). Mutations in the gene (Mtapt) encoding tau protein cause frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), thereby proving that tau dysfunction can directly result in neurodegeneration. Expression of human tau containing the most common FTDP-17 mutation (P301L) results in motor and behavioural deficits in transgenic mice, with age- and gene-dose-dependent development of NFT. This phenotype occurred as early as 6.5 months in hemizygous and 4.5 months in homozygous animals. NFT and Pick-body-like neuronal lesions occurred in the amygdala, septal nuclei, pre-optic nuclei, hypothalamus, midbrain, pons, medulla, deep cerebellar nuclei and spinal cord, with tau-immunoreactive pre-tangles in the cortex, hippocampus and basal ganglia. Areas with the most NFT had reactive gliosis. Spinal cord had axonal spheroids, anterior horn cell loss and axonal degeneration in anterior spinal roots. We also saw peripheral neuropathy and skeletal muscle with neurogenic atrophy. Brain and spinal cord contained insoluble tau that co-migrated with insoluble tau from AD and FTDP-17 brains. The phenotype of mice expressing P301L mutant tau mimics features of human tauopathies and provides a model for investigating the pathogenesis of diseases with NFT.


American Journal of Human Genetics | 2011

VPS35 Mutations in Parkinson Disease

Carles Vilariño-Güell; Christian Wider; Owen A. Ross; Justus C. Dachsel; Jennifer M. Kachergus; Sarah Lincoln; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Greggory J. Wilhoite; Justin A. Bacon; Behrouz Bahareh Behrouz; Heather L. Melrose; Emna Hentati; Andreas Puschmann; Daniel M. Evans; Elizabeth Conibear; Wyeth W. Wasserman; Jan O. Aasly; Pierre Burkhard; Ruth Djaldetti; Joseph Ghika; F. Hentati; Anna Krygowska-Wajs; Timothy Lynch; Eldad Melamed; Alex Rajput; Ali H. Rajput; Alessandra Solida; Ruey-Meei Wu; Ryan J. Uitti

The identification of genetic causes for Mendelian disorders has been based on the collection of multi-incident families, linkage analysis, and sequencing of genes in candidate intervals. This study describes the application of next-generation sequencing technologies to a Swiss kindred presenting with autosomal-dominant, late-onset Parkinson disease (PD). The family has tremor-predominant dopa-responsive parkinsonism with a mean onset of 50.6 ± 7.3 years. Exome analysis suggests that an aspartic-acid-to-asparagine mutation within vacuolar protein sorting 35 (VPS35 c.1858G>A; p.Asp620Asn) is the genetic determinant of disease. VPS35 is a central component of the retromer cargo-recognition complex, is critical for endosome-trans-golgi trafficking and membrane-protein recycling, and is evolutionarily highly conserved. VPS35 c.1858G>A was found in all affected members of the Swiss kindred and in three more families and one patient with sporadic PD, but it was not observed in 3,309 controls. Further sequencing of familial affected probands revealed only one other missense variant, VPS35 c.946C>T; (p.Pro316Ser), in a pedigree with one unaffected and two affected carriers, and thus the pathogenicity of this mutation remains uncertain. Retromer-mediated sorting and transport is best characterized for acid hydrolase receptors. However, the complex has many types of cargo and is involved in a diverse array of biologic pathways from developmental Wnt signaling to lysosome biogenesis. Our study implicates disruption of VPS35 and retromer-mediated trans-membrane protein sorting, rescue, and recycling in the neurodegenerative process leading to PD.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Identification of the α2-δ-1 subunit of voltage-dependent calcium channels as a molecular target for pain mediating the analgesic actions of pregabalin

Mark John Field; Peter Cox; Emma Stott; Heather L. Melrose; James Offord; Ti-zhi Su; Steve Bramwell; Laura Corradini; Steven England; Joanna Winks; Ross A. Kinloch; Jan Hendrich; Annette C. Dolphin; Tony Webb; Dic Williams

Neuropathic pain is a debilitating condition affecting millions of people around the world and is defined as pain that follows a lesion or dysfunction of the nervous system. This type of pain is difficult to treat, but the novel compounds pregabalin (Lyrica) and gabapentin (Neurontin) have proven clinical efficacy. Unlike traditional analgesics such as nonsteroidal antiinflammatory drugs or narcotics, these agents have no frank antiinflammatory actions and no effect on physiological pain. Although extensive preclinical studies have led to a number of suggestions, until recently their mechanism of action has not been clearly defined. Here, we describe studies on the analgesic effects of pregabalin in a mutant mouse containing a single-point mutation within the gene encoding a specific auxiliary subunit protein (α2-δ-1) of voltage-dependent calcium channels. The mice demonstrate normal pain phenotypes and typical responses to other analgesic drugs. We show that the mutation leads to a significant reduction in the binding affinity of pregabalin in the brain and spinal cord and the loss of its analgesic efficacy. These studies show conclusively that the analgesic actions of pregabalin are mediated through the α2-δ-1 subunit of voltage-gated calcium channels and establish this subunit as a therapeutic target for pain control.


American Journal of Human Genetics | 2011

Translation Initiator EIF4G1 Mutations in Familial Parkinson Disease

Marie Christine Chartier-Harlin; Justus C. Dachsel; Carles Vilariño-Güell; Sarah Lincoln; Frédéric Leprêtre; Mary M. Hulihan; Jennifer M. Kachergus; Austen J. Milnerwood; Lucia Tapia; Mee Sook Song; Emilie Le Rhun; Eugénie Mutez; Lydie Larvor; Aurélie Duflot; Christel Vanbesien-Mailliot; Alexandre Kreisler; Owen A. Ross; Kenya Nishioka; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Heather L. Melrose; Bahareh Behrouz; Brett H. Keeling; Justin A. Bacon; Emna Hentati; Williams L; Akiko Yanagiya; Nahum Sonenberg; Paul J. Lockhart; Abba C. Zubair

Genome-wide analysis of a multi-incident family with autosomal-dominant parkinsonism has implicated a locus on chromosomal region 3q26-q28. Linkage and disease segregation is explained by a missense mutation c.3614G>A (p.Arg1205His) in eukaryotic translation initiation factor 4-gamma (EIF4G1). Subsequent sequence and genotype analysis identified EIF4G1 c.1505C>T (p.Ala502Val), c.2056G>T (p.Gly686Cys), c.3490A>C (p.Ser1164Arg), c.3589C>T (p.Arg1197Trp) and c.3614G>A (p.Arg1205His) substitutions in affected subjects with familial parkinsonism and idiopathic Lewy body disease but not in control subjects. Despite different countries of origin, persons with EIF4G1 c.1505C>T (p.Ala502Val) or c.3614G>A (p.Arg1205His) mutations appear to share haplotypes consistent with ancestral founders. eIF4G1 p.Ala502Val and p.Arg1205His disrupt eIF4E or eIF3e binding, although the wild-type protein does not, and render mutant cells more vulnerable to reactive oxidative species. EIF4G1 mutations implicate mRNA translation initiation in familial parkinsonism and highlight a convergent pathway for monogenic, toxin and perhaps virally-induced Parkinson disease.


Neurobiology of Disease | 2010

Impaired dopaminergic neurotransmission and microtubule-associated protein tau alterations in human LRRK2 transgenic mice.

Heather L. Melrose; Justus C. Dachsel; Bahareh Behrouz; Sarah Lincoln; Mei Yue; Kelly M. Hinkle; Caroline Kent; Elena Korvatska; Julie P. Taylor; L. Witten; Y.-Q. Liang; Joel E. Beevers; Mona Boules; Brittany N. Dugger; V.A. Serna; A. Gaukhman; Xin Yu; Monica Castanedes-Casey; Adam Braithwaite; S. Ogholikhan; N. Yu; David I. Bass; G. Tyndall; Gerard D. Schellenberg; Dennis W. Dickson; Christopher Janus; Matthew J. Farrer

Mutations in the Leucine Rich Repeat Kinase 2 (LRRK2) gene, first described in 2004 have now emerged as the most important genetic finding in both autosomal dominant and sporadic Parkinsons disease (PD). While a formidable research effort has ensued since the initial gene discovery, little is known of either the normal or the pathological role of LRRK2. We have created lines of mice that express human wild-type (hWT) or G2019S Lrrk2 via bacterial artificial chromosome (BAC) transgenesis. In vivo analysis of the dopaminergic system revealed abnormal dopamine neurotransmission in both hWT and G2019S transgenic mice evidenced by a decrease in extra-cellular dopamine levels, which was detected without pharmacological manipulation. Immunopathological analysis revealed changes in localization and increased phosphorylation of microtubule binding protein tau in G2019S mice. Quantitative biochemical analysis confirmed the presence of differential phospho-tau species in G2019S mice but surprisingly, upon dephosphorylation the tau isoform banding pattern in G2019S mice remained altered. This suggests that other post-translational modifications of tau occur in G2019S mice. We hypothesize that Lrrk2 may impact on tau processing which subsequently leads to increased phosphorylation. Our models will be useful for further understanding of the mechanistic actions of LRRK2 and future therapeutic screening.


Neurobiology of Disease | 2011

Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice

Beate Winner; Heather L. Melrose; Chunmei Zhao; Kelly M. Hinkle; Mei Yue; Caroline Kent; Adam Braithwaite; S. Ogholikhan; R. Aigner; J. Winkler; Matthew J. Farrer; Fred H. Gage

The generation and maturation of adult neural stem/progenitor cells are impaired in many neurodegenerative diseases, among them is Parkinsons disease (PD). In mammals, including humans, adult neurogenesis is a lifelong feature of cellular brain plasticity in the hippocampal dentate gyrus (DG) and in the subventricular zone (SVZ)/olfactory bulb system. Hyposmia, depression, and anxiety are early non-motor symptoms in PD. There are parallels between brain regions associated with non-motor symptoms in PD and neurogenic regions. In autosomal dominant PD, mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are frequent. LRRK2 homologs in non-vertebrate systems play an important role in chemotaxis, cell polarity, and neurite arborization. We investigated adult neurogenesis and the neurite development of new neurons in the DG and SVZ/olfactory bulb system in bacterial artificial chromosome (BAC) human Lrrk2 G2019S transgenic mice. We report that mutant human Lrrk2 is highly expressed in the hippocampus in the DG and the SVZ of adult Lrrk2 G2019S mice. Proliferation of newly generated cells is significantly decreased and survival of newly generated neurons in the DG and olfactory bulb is also severely impaired. In addition, after stereotactic injection of a GFP retrovirus, newly generated neurons in the DG of Lrrk2 G2019S mice exhibited reduced dendritic arborization and fewer spines. This loss in mature, developed spines might point towards a decrease in synaptic connectivity. Interestingly, physical activity partially reverses the decrease in neuroblasts observed in Lrrk2 G2010S mice. These data further support a role for Lrrk2 in neuronal morphogenesis and provide new insights into the role of Lrrk2 in adult neurogenesis.


Neuroscience | 2006

Anatomical localization of leucine-rich repeat kinase 2 in mouse brain

Heather L. Melrose; Sarah Lincoln; G. Tyndall; Dennis W. Dickson; Matthew J. Farrer

Mutations in leucine-rich repeat kinase 2 (LRRK2) have recently been identified in autosomal dominant late-onset Parkinsons disease. Expression of LRRK2 has previously been reported in brain; however, no precise anatomical information is yet available. We have performed in situ hybridization and quantitative reverse transcription polymerase chain reaction to map LRRK2 mRNA expression in mouse brain. We find LRRK2 is highly expressed in the striatum, cortex and olfactory tubercle; however, little or no expression is found in the substantia nigra, where dopaminergic neurons preferentially degenerate in Parkinsons disease. These findings suggest that LRRK2 mRNA is expressed in dopamine-receptive areas rather than in the dopamine-synthesizing neurons. Consistent with a role LRRK2 in Parkinsons disease, dysfunction of leucine-rich repeat kinase 2 protein in dopamine-innervated areas may to lead to altered dopaminergic neurotransmission and degeneration of the nigro-striatal pathway.


Molecular Neurodegeneration | 2012

LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors

Kelly M. Hinkle; Mei Yue; Bahareh Behrouz; Justus C. Dachsel; Sarah Lincoln; Erin E. Bowles; Joel E. Beevers; Brittany N. Dugger; Beate Winner; Iryna Prots; Caroline Kent; Kenya Nishioka; Wen Lang Lin; Dennis W. Dickson; Christopher Janus; Matthew J. Farrer; Heather L. Melrose

Mutations in the LRRK2 gene are the most common cause of genetic Parkinson’s disease. Although the mechanisms behind the pathogenic effects of LRRK2 mutations are still not clear, data emerging from in vitro and in vivo models suggests roles in regulating neuronal polarity, neurotransmission, membrane and cytoskeletal dynamics and protein degradation.We created mice lacking exon 41 that encodes the activation hinge of the kinase domain of LRRK2. We have performed a comprehensive analysis of these mice up to 20 months of age, including evaluation of dopamine storage, release, uptake and synthesis, behavioral testing, dendritic spine and proliferation/neurogenesis analysis.Our results show that the dopaminergic system was not functionally comprised in LRRK2 knockout mice. However, LRRK2 knockout mice displayed abnormal exploratory activity in the open-field test. Moreover, LRRK2 knockout mice stayed longer than their wild type littermates on the accelerated rod during rotarod testing. Finally, we confirm that loss of LRRK2 caused degeneration in the kidney, accompanied by a progressive enhancement of autophagic activity and accumulation of autofluorescent material, but without evidence of biphasic changes.


Experimental Brain Research | 2006

Parkinson’s disease: a rethink of rodent models

Heather L. Melrose; Sarah Lincoln; Glenn M. Tyndall; Matthew J. Farrer

Parkinson’s disease (PD) is a multifactorial disease with a complex etiology that results from genetic risk factors, environmental exposures and most likely a combination of both. Rodent models of parkinsonism aim to reproduce key pathogenic features of the syndrome including movement disorder induced by the progressive loss of dopaminergic neurons in the substantia nigra, accompanied by the formation of α-synuclein containing Lewy body inclusions. Despite the creation of many excellent models, both chemically induced and genetically engineered, there is none that accurately demonstrates these features. Recent pathological staging studies in man have also emphasized the significant non-CNS component of PD that has yet to be tackled. Herein, we summarize rodent models of PD and what they offer to the field, and suggest future challenges and opportunities.


Human Mutation | 2011

Mutations in LRRK2 increase phosphorylation of peroxiredoxin 3 exacerbating oxidative stress‐induced neuronal death

Dario C. Angeles; Bong Hwa Gan; Luisa Onstead; Yi Zhao; Kah-Leong Lim; Justus C. Dachsel; Heather L. Melrose; Matthew J. Farrer; Zbigniew K. Wszolek; Dennis W. Dickson; Eng-King Tan

Mutations in the leucine rich repeat kinase 2 (LRRK2) gene are responsible for autosomal dominant and sporadic Parkinson disease (PD), possibly exerting their effects via a toxic gain of function. A common p.G2019S mutation (rs34637584:A>G) is responsible for up to 30–40% of PD cases in some ethnic populations. Here, we show that LRRK2 interacts with human peroxiredoxin 3 (PRDX3), a mitochondrial member of the antioxidant family of thioredoxin (Trx) peroxidases. Importantly, mutations in the LRRK2 kinase domain significantly increased phosphorylation of PRDX3 compared to wild‐type. The increase in PRDX3 phosphorylation was associated with decreased peroxidase activity and increased death in LRRK2‐expressing but not in LRRK2‐depleted or vector‐transfected neuronal cells. LRRK2 mutants stimulated mitochondrial factors involved in apoptosis and induced production of reactive oxygen species (ROS) and oxidative modification of macromolecules. Furthermore, immunoblot and immunohistochemical analysis of postmortem human PD patients carrying the p.G2019S mutation showed a marked increase in phosphorylated PRDX3 (p‐PRDX3) relative to normal brain. We showed that LRRK2 mutations increase the inhibition of an endogenous peroxidase by phosphorylation promoting dysregulation of mitochondrial function and oxidative damage. Our findings provide a mechanistic link between the enhanced kinase activity of PD‐linked LRRK2 and neuronal cell death. 32:1390–1397, 2011. ©2011 Wiley Periodicals, Inc.

Collaboration


Dive into the Heather L. Melrose's collaboration.

Top Co-Authors

Avatar

Matthew J. Farrer

University of British Columbia

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