Network


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

Hotspot


Dive into the research topics where Adamantios Mamais is active.

Publication


Featured researches published by Adamantios Mamais.


Biochimica et Biophysica Acta | 2013

Inhibition of LRRK2 kinase activity stimulates macroautophagy.

Claudia Manzoni; Adamantios Mamais; Sybille Dihanich; Rosella Abeti; Marc P.M. Soutar; Helene Plun-Favreau; Paola Giunti; Sharon A. Tooze; Rina Bandopadhyay; Patrick A. Lewis

Leucine Rich Repeat Kinase 2 (LRRK2) is one of the most important genetic contributors to Parkinsons disease. LRRK2 has been implicated in a number of cellular processes, including macroautophagy. To test whether LRRK2 has a role in regulating autophagy, a specific inhibitor of the kinase activity of LRRK2 was applied to human neuroglioma cells and downstream readouts of autophagy examined. The resulting data demonstrate that inhibition of LRRK2 kinase activity stimulates macroautophagy in the absence of any alteration in the translational targets of mTORC1, suggesting that LRRK2 regulates autophagic vesicle formation independent of canonical mTORC1 signaling. This study represents the first pharmacological dissection of the role LRRK2 plays in the autophagy/lysosomal pathway, emphasizing the importance of this pathway as a marker for LRRK2 physiological function. Moreover it highlights the need to dissect autophagy and lysosomal activities in the context of LRRK2 related pathologies with the final aim of understanding their aetiology and identifying specific targets for disease modifying therapies in patients.


The EMBO Journal | 2003

Regulation of V(D)J recombination by nucleosome positioning at recombination signal sequences

Matthias Baumann; Adamantios Mamais; Fraser McBlane; Hua Xiao; Joan Boyes

A key component in the regulation of V(D)J recombination is control of the accessibility of RAG proteins to recombination signal sequences (RSS). Nucleosomes are known to inhibit this accessibility. We show here that the signal sequence itself represses accessibility by causing nucleosome positioning over the RSS. This positioning is mediated, in vitro and in vivo, by the conserved nonamer of the RSS. Consistent with this strong positioning, nucleosomes at RSSs are resistant to remodelling by nucleosome sliding. In vivo we find that consensus RSSs are preferentially protected, whereas those that lack a consensus nonamer, including some cryptic RSSs, fail to position nucleosomes. Decreased protection of these non‐consensus RSSs correlates with their increased use in recombination assays. We therefore suggest that nucleosome positioning by RSSs provides a previously unanticipated level of protection and regulation of V(D)J recombination.


Neurobiology of Aging | 2014

Dysregulation of glucose metabolism is an early event in sporadic Parkinson's disease

Laura Dunn; George F.G. Allen; Adamantios Mamais; Helen Ling; Abi Li; Kate Duberley; Iain Hargreaves; Simon Pope; Janice L. Holton; Andrew J. Lees; Simon Heales; Rina Bandopadhyay

Unlike most other cell types, neurons preferentially metabolize glucose via the pentose phosphate pathway (PPP) to maintain their antioxidant status. Inhibiting the PPP in neuronal cell models causes cell death. In rodents, inhibition of this pathway causes selective dopaminergic cell death leading to motor deficits resembling parkinsonism. Using postmortem human brain tissue, we characterized glucose metabolism via the PPP in sporadic Parkinsons disease (PD), Alzheimers disease (AD), and controls. AD brains showed increased nicotinamide adenine dinucleotide phosphate (NADPH) production in areas affected by disease. In PD however, increased NADPH production was only seen in the affected areas of late-stage cases. Quantifying PPP NADPH-producing enzymes glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase by enzyme-linked immunosorbent assay, showed a reduction in the putamen of early-stage PD and interestingly in the cerebellum of early and late-stage PD. Importantly, there was no decrease in enzyme levels in the cortex, putamen, or cerebellum of AD. Our results suggest that down-regulation of PPP enzymes and a failure to increase antioxidant reserve is an early event in the pathogenesis of sporadic PD.


Biochemical and Biophysical Research Communications | 2013

Pathogenic Parkinson’s disease mutations across the functional domains of LRRK2 alter the autophagic/lysosomal response to starvation

Claudia Manzoni; Adamantios Mamais; Sybille Dihanich; Phillip McGoldrick; Michael J. Devine; Julia Zerle; Eleanna Kara; Jan-Willem Taanman; Daniel G. Healy; Jose Felix Marti-Masso; A. H. V. Schapira; Helene Plun-Favreau; Sharon A. Tooze; John Hardy; Rina Bandopadhyay; Patrick A. Lewis

Highlights • Mutations in the ROC, COR and Kinase domain of LRRK2 alter the autophagic response to starvation.• LC3-I/II ratio following starvation is altered by mutations, as well as p62 and WIPI2 positive puncta.• This occurs independently of any alteration in downstream targets of mTORC1.


Nature Communications | 2014

Phosphorylation of LRRK2 by casein kinase 1α regulates trans -Golgi clustering via differential interaction with ARHGEF7

Ruth Chia; Sara Haddock; Alexandra Beilina; Iakov N. Rudenko; Adamantios Mamais; Alice Kaganovich; Yan-Yan Li; Ravindran Kumaran; Michael A. Nalls; Mark R. Cookson

LRRK2, a gene relevant to Parkinsons disease, encodes a scaffolding protein with both GTPase and kinase activities. LRRK2 protein is itself phosphorylated and therefore subject to regulation by cell signaling but the kinase(s) responsible for this event have not been definitively identified. Here, using an unbiased siRNA kinome screen, we identify and validate casein kinase 1α (CK1α) as being responsible for LRRK2 phosphorylation, including in the adult mouse striatum. We further show that LRRK2 recruitment to TGN46-positive Golgi-derived vesicles is modulated by constitutive LRRK2 phosphorylation by CK1α. These effects are mediated by differential protein interactions of LRRK2 with a guanine nucleotide exchange factor, ARHGEF7. These pathways are therefore likely involved in the physiological maintenance of the Golgi in cells, which may play a role in the pathogenesis of Parkinsons disease.


PLOS ONE | 2012

Phosphorylation of 4E-BP1 in the mammalian brain is not altered by LRRK2 expression or pathogenic mutations.

Alzbeta Trancikova; Adamantios Mamais; Philip J. Webber; Klodjan Stafa; Elpida Tsika; Liliane Glauser; Andrew B. West; Rina Bandopadhyay; Darren J. Moore

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are a common cause of autosomal dominant familial Parkinsons disease (PD). LRRK2 encodes a multi-domain protein containing GTPase and kinase enzymatic domains. Disease-associated mutations in LRRK2 variably influence enzymatic activity with the common G2019S variant leading to enhanced kinase activity. Mutant LRRK2 induces neuronal toxicity through a kinase-dependent mechanism suggesting that kinase activity is important for mediating the pathogenic effects of LRRK2 mutations. A number of LRRK2 kinase substrates have been identified in vitro but whether they represent authentic physiological substrates in mammalian cells or tissues is not yet clear. The eukaryotic initiation factor 4E (eIF4E)-binding protein, 4E-BP1, was recently identified as a potential substrate of LRRK2 kinase activity in vitro and in Drosophila with phosphorylation occurring at Thr37 and Thr46. Here, we explore a potential interaction of LRRK2 and 4E-BP1 in mammalian cells and brain. We find that LRRK2 can weakly phosphorylate 4E-BP1 in vitro but LRRK2 overexpression is not able to alter endogenous 4E-BP1 phosphorylation in mammalian cells. In mammalian neurons LRRK2 and 4E-BP1 display minimal co-localization, whereas the subcellular distribution, protein complex formation and covalent post-translational modification of endogenous 4E-BP1 are not altered in the brains of LRRK2 knockout or mutant LRRK2 transgenic mice. In the brain, the phosphorylation of 4E-BP1 at Thr37 and Thr46 does not change in LRRK2 knockout or mutant LRRK2 transgenic mice, nor is 4E-BP1 phosphorylation altered in idiopathic or G2019S mutant PD brains. Collectively, our results suggest that 4E-BP1 is neither a major nor robust physiological substrate of LRRK2 in mammalian cells or brain.


PLOS ONE | 2013

Fine-Mapping, Gene Expression and Splicing Analysis of the Disease Associated LRRK2 Locus

Daniah Trabzuni; Mina Ryten; Warren Emmett; Adaikalavan Ramasamy; Karl J. Lackner; Tanja Zeller; Robert Walker; Colin Smith; Patrick A. Lewis; Adamantios Mamais; Rohan de Silva; Jana Vandrovcova; Dena Hernandez; Michael A. Nalls; Manu Sharma; Sophie Garnier; Suzanne Lesage; Javier Simón-Sánchez; Thomas Gasser; Peter Heutink; Alexis Brice; Andrew Singleton; Huaibin Cai; Eric E. Schadt; Nicholas W. Wood; Rina Bandopadhyay; Michael E. Weale; John Hardy; Vincent Plagnol

Association studies have identified several signals at the LRRK2 locus for Parkinsons disease (PD), Crohns disease (CD) and leprosy. However, little is known about the molecular mechanisms mediating these effects. To further characterize this locus, we fine-mapped the risk association in 5,802 PD and 5,556 controls using a dense genotyping array (ImmunoChip). Using samples from 134 post-mortem control adult human brains (UK Human Brain Expression Consortium), where up to ten brain regions were available per individual, we studied the regional variation, splicing and regulation of LRRK2. We found convincing evidence for a common variant PD association located outside of the LRRK2 protein coding region (rs117762348, A>G, P = 2.56×10−8, case/control MAF 0.083/0.074, odds ratio 0.86 for the minor allele with 95% confidence interval [0.80–0.91]). We show that mRNA expression levels are highest in cortical regions and lowest in cerebellum. We find an exon quantitative trait locus (QTL) in brain samples that localizes to exons 32–33 and investigate the molecular basis of this eQTL using RNA-Seq data in n = 8 brain samples. The genotype underlying this eQTL is in strong linkage disequilibrium with the CD associated non-synonymous SNP rs3761863 (M2397T). We found two additional QTLs in liver and monocyte samples but none of these explained the common variant PD association at rs117762348. Our results characterize the LRRK2 locus, and highlight the importance and difficulties of fine-mapping and integration of multiple datasets to delineate pathogenic variants and thus develop an understanding of disease mechanisms.


Human Molecular Genetics | 2014

A Parkinson's disease gene regulatory network identifies the signaling protein RGS2 as a modulator of LRRK2 activity and neuronal toxicity

Julien Dusonchet; Hu Li; Maria Guillily; Min Liu; Klodjan Stafa; Claudio Derada Troletti; Joon Y. Boon; Shamol Saha; Liliane Glauser; Adamantios Mamais; Allison Citro; Katherine L. Youmans; Liqun Liu; Bernard L. Schneider; Patrick Aebischer; Zhenyu Yue; Rina Bandopadhyay; Marcie A. Glicksman; Darren J. Moore; James J. Collins; Benjamin Wolozin

Mutations in LRRK2 are one of the primary genetic causes of Parkinsons disease (PD). LRRK2 contains a kinase and a GTPase domain, and familial PD mutations affect both enzymatic activities. However, the signaling mechanisms regulating LRRK2 and the pathogenic effects of familial mutations remain unknown. Identifying the signaling proteins that regulate LRRK2 function and toxicity remains a critical goal for the development of effective therapeutic strategies. In this study, we apply systems biology tools to human PD brain and blood transcriptomes to reverse-engineer a LRRK2-centered gene regulatory network. This network identifies several putative master regulators of LRRK2 function. In particular, the signaling gene RGS2, which encodes for a GTPase-activating protein (GAP), is a key regulatory hub connecting the familial PD-associated genes DJ-1 and PINK1 with LRRK2 in the network. RGS2 expression levels are reduced in the striata of LRRK2 and sporadic PD patients. We identify RGS2 as a novel interacting partner of LRRK2 in vivo. RGS2 regulates both the GTPase and kinase activities of LRRK2. We show in mammalian neurons that RGS2 regulates LRRK2 function in the control of neuronal process length. RGS2 is also protective against neuronal toxicity of the most prevalent mutation in LRRK2, G2019S. We find that RGS2 regulates LRRK2 function and neuronal toxicity through its effects on kinase activity and independently of GTPase activity, which reveals a novel mode of action for GAP proteins. This work identifies RGS2 as a promising target for interfering with neurodegeneration due to LRRK2 mutations in PD patients.


PLOS ONE | 2011

Pathogenic LRRK2 Mutations Do Not Alter Gene Expression in Cell Model Systems or Human Brain Tissue

Michael J. Devine; Alice Kaganovich; Mina Ryten; Adamantios Mamais; Daniah Trabzuni; Claudia Manzoni; Philip McGoldrick; Diane Chan; Allissa Dillman; Julia Zerle; Susannah Horan; Jan-Willem Taanman; John Hardy; Jose-Felix Marti-Masso; Daniel Healey; A. H. V. Schapira; Benjamin Wolozin; Rina Bandopadhyay; Mark R. Cookson; Marcel van der Brug; Patrick A. Lewis

Point mutations in LRRK2 cause autosomal dominant Parkinsons disease. Despite extensive efforts to determine the mechanism of cell death in patients with LRRK2 mutations, the aetiology of LRRK2 PD is not well understood. To examine possible alterations in gene expression linked to the presence of LRRK2 mutations, we carried out a case versus control analysis of global gene expression in three systems: fibroblasts isolated from LRRK2 mutation carriers and healthy, non-mutation carrying controls; brain tissue from G2019S mutation carriers and controls; and HEK293 inducible LRRK2 wild type and mutant cell lines. No significant alteration in gene expression was found in these systems following correction for multiple testing. These data suggest that any alterations in basal gene expression in fibroblasts or cell lines containing mutations in LRRK2 are likely to be quantitatively small. This work suggests that LRRK2 is unlikely to play a direct role in modulation of gene expression, although it remains possible that this protein can influence mRNA expression under pathogenic cicumstances.


Scientific Reports | 2016

mTOR independent regulation of macroautophagy by Leucine Rich Repeat Kinase 2 via Beclin-1.

Claudia Manzoni; Adamantios Mamais; Dorien A. Roosen; Sybille Dihanich; Marc P.M. Soutar; Helene Plun-Favreau; Rina Bandopadhyay; John Hardy; Sharon A. Tooze; Mark R. Cookson; Patrick A. Lewis

Leucine rich repeat kinase 2 is a complex enzyme with both kinase and GTPase activities, closely linked to the pathogenesis of several human disorders including Parkinson’s disease, Crohn’s disease, leprosy and cancer. LRRK2 has been implicated in numerous cellular processes; however its physiological function remains unclear. Recent reports suggest that LRRK2 can act to regulate the cellular catabolic process of macroautophagy, although the precise mechanism whereby this occurs has not been identified. To investigate the signalling events through which LRRK2 acts to influence macroautophagy, the mammalian target of rapamycin (mTOR)/Unc-51-like kinase 1 (ULK1) and Beclin-1/phosphatidylinositol 3-kinase (PI3K) pathways were evaluated in astrocytic cell models in the presence and absence of LRRK2 kinase inhibitors. Chemical inhibition of LRRK2 kinase activity resulted in the stimulation of macroautophagy in a non-canonical fashion, independent of mTOR and ULK1, but dependent upon the activation of Beclin 1-containing class III PI3-kinase.

Collaboration


Dive into the Adamantios Mamais's collaboration.

Top Co-Authors

Avatar

Rina Bandopadhyay

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sybille Dihanich

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Lees

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Janice L. Holton

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Tammaryn Lashley

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

John Hardy

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tamas Revesz

UCL Institute of Neurology

View shared research outputs
Researchain Logo
Decentralizing Knowledge