Network


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

Hotspot


Dive into the research topics where Robert P. Mason is active.

Publication


Featured researches published by Robert P. Mason.


Nature Genetics | 2013

Glutathione peroxidase activity is neuroprotective in models of Huntington's disease

Robert P. Mason; Massimiliano Casu; Nicola J. Butler; Carlo Breda; Susanna Campesan; Jannine Clapp; Edward W. Green; Devyani Dhulkhed; Charalambos P. Kyriacou; Flaviano Giorgini

Huntingtons disease is a fatal neurodegenerative disorder caused by a CAG repeat expansion encoding a polyglutamine tract in the huntingtin (Htt) protein. Here we report a genome-wide overexpression suppressor screen in which we identified 317 ORFs that ameliorate the toxicity of a mutant Htt fragment in yeast and that have roles in diverse cellular processes, including mitochondrial import and copper metabolism. Two of these suppressors encode glutathione peroxidases (GPxs), which are conserved antioxidant enzymes that catalyze the reduction of hydrogen peroxide and lipid hydroperoxides. Using genetic and pharmacological approaches in yeast, mammalian cells and Drosophila, we found that GPx activity robustly ameliorates Huntingtons disease–relevant metrics and is more protective than other antioxidant approaches tested here. Notably, we found that GPx activity, unlike many antioxidant treatments, does not inhibit autophagy, which is an important mechanism for clearing mutant Htt. Because previous clinical trials have indicated that GPx mimetics are well tolerated in humans, this study may have important implications for treating Huntingtons disease.


Journal of Biological Chemistry | 2011

Functional Gene Expression Profiling in Yeast Implicates Translational Dysfunction in Mutant Huntingtin Toxicity

Eran Tauber; Leonor Miller-Fleming; Robert P. Mason; Wanda Kwan; Jannine Clapp; Nicola J. Butler; Tiago F. Outeiro; Paul J. Muchowski; Flaviano Giorgini

Huntington disease (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine tract in the huntingtin (htt) protein. To uncover candidate therapeutic targets and networks involved in pathogenesis, we integrated gene expression profiling and functional genetic screening to identify genes critical for mutant htt toxicity in yeast. Using mRNA profiling, we have identified genes differentially expressed in wild-type yeast in response to mutant htt toxicity as well as in three toxicity suppressor strains: bna4Δ, mbf1Δ, and ume1Δ. BNA4 encodes the yeast homolog of kynurenine 3-monooxygenase, a promising drug target for HD. Intriguingly, despite playing diverse cellular roles, these three suppressors share common differentially expressed genes involved in stress response, translation elongation, and mitochondrial transport. We then systematically tested the ability of the differentially expressed genes to suppress mutant htt toxicity when overexpressed and have thereby identified 12 novel suppressors, including genes that play a role in stress response, Golgi to endosome transport, and rRNA processing. Integrating the mRNA profiling data and the genetic screening data, we have generated a robust network that shows enrichment in genes involved in rRNA processing and ribosome biogenesis. Strikingly, these observations implicate dysfunction of translation in the pathology of HD. Recent work has shown that regulation of translation is critical for life span extension in Drosophila and that manipulation of this process is protective in Parkinson disease models. In total, these observations suggest that pharmacological manipulation of translation may have therapeutic value in HD.


Prion | 2011

Modeling Huntington disease in yeast: Perspectives and future directions

Robert P. Mason; Flaviano Giorgini

Yeast have been extensively used to model aspects of protein folding diseases, yielding novel mechanistic insights and identifying promising candidate therapeutic targets. In particular, the neurodegenerative disorder Huntington disease (HD), which is caused by the abnormal expansion of a polyglutamine tract in the huntingtin (htt) protein, has been widely studied in yeast. This work has led to the identification of several promising therapeutic targets and compounds that have been validated in mammalian cells, Drosophila and rodent models of HD. Here we discuss the development of yeast models of mutant htt toxicity and misfolding, as well as the mechanistic insights gleaned from this simple model. The role of yeast prions in the toxicity/misfolding of mutant htt is also highlighted. Furthermore, we provide an overview of the application of HD yeast models in both genetic and chemical screens, and the fruitful results obtained from these approaches. Finally, we discuss the future of yeast in neurodegenerative research, in the context of HD and other diseases.


Biochemical Society Transactions | 2010

Metallothioneins and copper metabolism are candidate therapeutic targets in Huntington's disease

Sarah Hands; Robert P. Mason; M. Umar Sajjad; Flaviano Giorgini; Andreas Wyttenbach

HD (Huntingtons disease) is caused by a polyQ (polyglutamine) expansion in the huntingtin protein, which leads to protein misfolding and aggregation of this protein. Abnormal copper accumulation in the HD brain was first reported more than 15 years ago. Recent findings show that copper-regulatory genes are induced during HD and copper binds to an N-terminal fragment of huntingtin, supporting the involvement of abnormal copper metabolism in HD. We have demonstrated that in vitro copper accelerates the fibrillization of an N-terminal fragment of huntingtin with an expanded polyQ stretch (httExon1). As we found that copper also increases polyQ aggregation and toxicity in mammalian cells expressing httExon1, we investigated further whether overexpression of genes involved in copper metabolism, notably MTs (metallothioneins) known to bind copper, protect against httExon1 toxicity. Using a yeast model of HD, we have shown that overexpression of several genes involved in copper metabolism reduces polyQ-mediated toxicity. Overexpression of MT-3 in mammalian cells significantly reduced polyQ aggregation and toxicity. We propose that copper-binding and/or -chaperoning proteins, especially MTs, are potential therapeutic targets for HD.


Yeast | 2011

Ferric reductase genes involved in high-affinity iron uptake are differentially regulated in yeast and hyphae of Candida albicans

Rose E. Jeeves; Robert P. Mason; Alexandra Woodacre; Annette Cashmore

The pathogenic yeast Candida albicans possesses a reductive iron uptake system which is active in iron‐restricted conditions. The sequestration of iron by this mechanism initially requires the reduction of free iron to the soluble ferrous form, which is catalysed by ferric reductase proteins. Reduced iron is then taken up into the cell by a complex of a multicopper oxidase protein and an iron transport protein. Multicopper oxidase proteins require copper to function and so reductive iron and copper uptake are inextricably linked. It has previously been established that Fre10 is the major cell surface ferric reductase in C. albicans and that transcription of FRE10 is regulated in response to iron levels. We demonstrate here that Fre10 is also a cupric reductase and that Fre7 also makes a significant contribution to cell surface ferric and cupric reductase activity. It is also shown, for the first time, that transcription of FRE10 and FRE7 is lower in hyphae compared to yeast and that this leads to a corresponding decrease in cell surface ferric, but not cupric, reductase activity. This demonstrates that the regulation of two virulence determinants, the reductive iron uptake system and the morphological form of C. albicans, are linked. Copyright


Microbiology | 2008

Copper-dependent transcriptional regulation by Candida albicans Mac1p.

Alexandra Woodacre; Robert P. Mason; Rose E. Jeeves; Annette Cashmore

We have previously shown that copper uptake and regulation in the opportunistic pathogen Candida albicans has some similarities to those in Saccharomyces cerevisiae, including the activation of the copper transporter gene CaCTR1 under low-copper conditions by the transcription factor CaMac1p. However, in this study, further analysis has shown that the actual mechanism of regulation by CaMac1p is different from that of its S. cerevisiae homologue. We demonstrate for the first time, to our knowledge, that the CaMAC1 gene is transcriptionally autoregulated in a copper-dependent manner, in contrast to ScMAC1, which is constitutively transcribed. We also demonstrate that the presence of one copper response element in the promoters of CaCTR1, CaMAC1 and the ferric/cupric reductase gene CaFRE7 is sufficient for normal levels of copper-responsive transcription. In contrast, two promoter elements are essential for normal levels of copper-dependent transcriptional activation by ScMac1p. CaMac1p is also involved in the regulation of the iron-responsive transcriptional repressor gene SFU1 and the alternative oxidase gene AOX2. This work describes a key feature of the copper uptake system in C. albicans that distinguishes it from similar processes in the model yeast S. cerevisiae. The importance of copper uptake in the environment of the human host and the implications for the disease process are discussed.


PLOS ONE | 2010

The Roles of the Saccharomyces cerevisiae RecQ Helicase SGS1 in Meiotic Genome Surveillance

Amit Dipak Amin; Alexandre B. H. Chaix; Robert P. Mason; Richard M. Badge; Rhona H. Borts

Background The Saccharomyces cerevisiae RecQ helicase Sgs1 is essential for mitotic and meiotic genome stability. The stage at which Sgs1 acts during meiosis is subject to debate. Cytological experiments showed that a deletion of SGS1 leads to an increase in synapsis initiation complexes and axial associations leading to the proposal that it has an early role in unwinding surplus strand invasion events. Physical studies of recombination intermediates implicate it in the dissolution of double Holliday junctions between sister chromatids. Methodology/Principal Findings In this work, we observed an increase in meiotic recombination between diverged sequences (homeologous recombination) and an increase in unequal sister chromatid events when SGS1 is deleted. The first of these observations is most consistent with an early role of Sgs1 in unwinding inappropriate strand invasion events while the second is consistent with unwinding or dissolution of recombination intermediates in an Mlh1- and Top3-dependent manner. We also provide data that suggest that Sgs1 is involved in the rejection of ‘second strand capture’ when sequence divergence is present. Finally, we have identified a novel class of tetrads where non-sister spores (pairs of spores where each contains a centromere marker from a different parent) are inviable. We propose a model for this unusual pattern of viability based on the inability of sgs1 mutants to untangle intertwined chromosomes. Our data suggest that this role of Sgs1 is not dependent on its interaction with Top3. We propose that in the absence of SGS1 chromosomes may sometimes remain entangled at the end of pre-meiotic replication. This, combined with reciprocal crossing over, could lead to physical destruction of the recombined and entangled chromosomes. We hypothesise that Sgs1, acting in concert with the topoisomerase Top2, resolves these structures. Conclusions This work provides evidence that Sgs1 interacts with various partner proteins to maintain genome stability throughout meiosis.


Journal of Molecular Medicine | 2016

Connectivity mapping uncovers small molecules that modulate neurodegeneration in Huntington's disease models.

Joshua L. Smalley; Carlo Breda; Robert P. Mason; Gurdeep S. Kooner; Ruth Luthi-Carter; Timothy W. Gant; Flaviano Giorgini

Huntington’s disease (HD) is a genetic disease caused by a CAG trinucleotide repeat expansion encoding a polyglutamine tract in the huntingtin (HTT) protein, ultimately leading to neuronal loss and consequent cognitive decline and death. As no treatments for HD currently exist, several chemical screens have been performed using cell-based models of mutant HTT toxicity. These screens measured single disease-related endpoints, such as cell death, but had low ‘hit rates’ and limited dimensionality for therapeutic detection. Here, we have employed gene expression microarray analysis of HD samples—a snapshot of the expression of 25,000 genes—to define a gene expression signature for HD from publically available data. We used this information to mine a database for chemicals positively and negatively correlated to the HD gene expression signature using the Connectivity Map, a tool for comparing large sets of gene expression patterns. Chemicals with negatively correlated expression profiles were highly enriched for protective characteristics against mutant HTT fragment toxicity in in vitro and in vivo models. This study demonstrates the potential of using gene expression to mine chemical activity, guide chemical screening, and detect potential novel therapeutic compounds.Key messagesSingle-endpoint chemical screens have low therapeutic discovery hit-rates.In the context of HD, we guided a chemical screen using gene expression data.The resulting chemicals were highly enriched for suppressors of mutant HTT fragment toxicity.This study provides a proof of concept for wider usage in all chemical screening.


Movement Disorders (Second Edition)#R##N#Genetics and Models | 2015

Chapter 33 – Modeling Huntington Disease in Yeast and Invertebrates

Robert P. Mason; Carlo Breda; Gurdeep S. Kooner; Giovanna R. Mallucci; Charalambos P. Kyriacou; Flaviano Giorgini

Huntington disease (HD) is a fatal, late-onset neurodegenerative disorder caused by the expansion of a CAG trinucleotide repeat in the huntingtin gene (HTT), which encodes a polyglutamine (polyQ) tract with a high propensity to misfold and aggregate. Mutant HTT is believed to act predominantly via a gain-of-toxic-function mechanism, leading to a myriad of downstream dysfunctions at the cellular level, culminating with generation of disease symptoms. The gain-of-function nature of this mutation has permitted the development of several transgenic models of HD that express all or a portion of human HTT containing the polyQ stretch, including yeast, nematode, fruit fly, and mouse. While murine models of HD have become the mammalian system of choice—and the gold standard for preclinical testing of drug-like compounds—yeast and invertebrate models have been used extensively and have made major contributions to both the understanding of the pathogenesis of HD and the identification of candidate therapeutic targets and compounds for this disorder. Here we provide an overview of these models and discuss how they have contributed to our understanding of HD.


PLOS Currents | 2014

Aggregation-Prone Proteins Modulate Huntingtin Inclusion Body Formation in Yeast

Ralitsa B. Kantcheva; Robert P. Mason; Flaviano Giorgini

Huntingtons disease (HD) is a fatal neurodegenerative disorder caused by a polyglutamine expansion in the huntingtin (HTT) protein. The expression of mutant HTT in the baker’s yeast Saccharomyces cerevisiae recapitulates many of the cellular phenotypes observed in mammalian HD models. Mutant HTT aggregation and toxicity in yeast is influenced by the presence of the Rnq1p and Sup35p prions, as well as other glutamine/asparagine-rich aggregation-prone proteins. Here we investigated the ability of a subset of these proteins to modulate mutant HTT aggregation and to substitute for the prion form of Rnq1p. We find that overexpression of either the putative prion Ybr016wp or the Sup35p prion restores aggregation of mutant HTT in yeast cells lacking the Rnq1p prion. These results indicate that an interchangeable suite of aggregation-prone proteins regulates mutant HTT aggregation dynamics in yeast, which may have implications for mutant HTT aggregation in human cells.

Collaboration


Dive into the Robert P. Mason's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlo Breda

University of Leicester

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

Wanda Kwan

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge