Network


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

Hotspot


Dive into the research topics where Yugo Tsuchiya is active.

Publication


Featured researches published by Yugo Tsuchiya.


American Journal of Human Genetics | 2014

Exome sequence reveals mutations in CoA synthase as a cause of neurodegeneration with brain iron accumulation

Sabrina Dusi; Lorella Valletta; Tobias B. Haack; Yugo Tsuchiya; Paola Venco; Marco Tigano; Nikita Demchenko; Thomas Wieland; Thomas Schwarzmayr; Tim M. Strom; Federica Invernizzi; Barbara Garavaglia; Allison Gregory; Lynn Sanford; Jeffrey Hamada; Conceição Bettencourt; Henry Houlden; Luisa Chiapparini; Giovanna Zorzi; Manju A. Kurian; Nardo Nardocci; Holger Prokisch; Susan J. Hayflick; Ivan Gout; Valeria Tiranti

Neurodegeneration with brain iron accumulation (NBIA) comprises a clinically and genetically heterogeneous group of disorders with progressive extrapyramidal signs and neurological deterioration, characterized by iron accumulation in the basal ganglia. Exome sequencing revealed the presence of recessive missense mutations in COASY, encoding coenzyme A (CoA) synthase in one NBIA-affected subject. A second unrelated individual carrying mutations in COASY was identified by Sanger sequence analysis. CoA synthase is a bifunctional enzyme catalyzing the final steps of CoA biosynthesis by coupling phosphopantetheine with ATP to form dephospho-CoA and its subsequent phosphorylation to generate CoA. We demonstrate alterations in RNA and protein expression levels of CoA synthase, as well as CoA amount, in fibroblasts derived from the two clinical cases and in yeast. This is the second inborn error of coenzyme A biosynthesis to be implicated in NBIA.


Biochemical Pharmacology | 2015

The phenotype of a knockout mouse identifies flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic ageing

Sandra G. Gonzalez Malagon; Anna N. Melidoni; Diana Hernandez; Bilal A. Omar; Lyndsey Houseman; Sunil Veeravalli; Flora Scott; Dorsa Varshavi; Jeremy R. Everett; Yugo Tsuchiya; John F. Timms; Ian R. Phillips; Elizabeth A. Shephard

Graphical abstract


PLOS ONE | 2014

Changes in Acetyl CoA Levels during the Early Embryonic Development of Xenopus laevis

Yugo Tsuchiya; Uyen Pham; Wanzhou Hu; Shin-ichi Ohnuma; Ivan Gout

Coenzyme A (CoA) is a ubiquitous and fundamental intracellular cofactor. CoA acts as a carrier of metabolically important carboxylic acids in the form of CoA thioesters and is an obligatory component of a multitude of catabolic and anabolic reactions. Acetyl CoA is a CoA thioester derived from catabolism of all major carbon fuels. This metabolite is at a metabolic crossroads, either being further metabolised as an energy source or used as a building block for biosynthesis of lipids and cholesterol. In addition, acetyl CoA serves as the acetyl donor in protein acetylation reactions, linking metabolism to protein post-translational modifications. Recent studies in yeast and cultured mammalian cells have suggested that the intracellular level of acetyl CoA may play a role in the regulation of cell growth, proliferation and apoptosis, by affecting protein acetylation reactions. Yet, how the levels of this metabolite change in vivo during the development of a vertebrate is not known. We measured levels of acetyl CoA, free CoA and total short chain CoA esters during the early embryonic development of Xenopus laevis using HPLC. Acetyl CoA and total short chain CoA esters start to increase around midblastula transition (MBT) and continue to increase through stages of gastrulation, neurulation and early organogenesis. Pre-MBT embryos contain more free CoA relative to acetyl CoA but there is a shift in the ratio of acetyl CoA to CoA after MBT, suggesting a metabolic transition that results in net accumulation of acetyl CoA. At the whole-embryo level, there is an apparent correlation between the levels of acetyl CoA and levels of acetylation of a number of proteins including histones H3 and H2B. This suggests the level of acetyl CoA may be a factor, which determines the degree of acetylation of these proteins, hence may play a role in the regulation of embryogenesis.


Biochemical Society Transactions | 2014

Coenzyme A biosynthetic machinery in mammalian cells.

David Lopez Martinez; Yugo Tsuchiya; Ivan Gout

CoA (coenzyme A) is an essential cofactor in all living organisms. CoA and its thioester derivatives [acetyl-CoA, malonyl-CoA, HMG-CoA (3-hydroxy-3-methylglutaryl-CoA) etc.] participate in diverse anabolic and catabolic pathways, allosteric regulatory interactions and the regulation of gene expression. The biosynthesis of CoA requires pantothenic acid, cysteine and ATP, and involves five enzymatic steps that are highly conserved from prokaryotes to eukaryotes. The intracellular levels of CoA and its derivatives change in response to extracellular stimuli, stresses and metabolites, and in human pathologies, such as cancer, metabolic disorders and neurodegeneration. In the present mini-review, we describe the current understanding of the CoA biosynthetic pathway, provide a detailed overview on expression and subcellular localization of enzymes implicated in CoA biosynthesis, their regulation and the potential to form multi-enzyme complexes for efficient and highly co-ordinated biosynthetic process.


Biochemical Society Transactions | 2014

Signalling functions of coenzyme A and its derivatives in mammalian cells.

Hongorzul Davaapil; Yugo Tsuchiya; Ivan Gout

In all living organisms, CoA (coenzyme A) is synthesized in a highly conserved process that requires pantothenic acid (vitamin B5), cysteine and ATP. CoA is uniquely designed to function as an acyl group carrier and a carbonyl-activating group in diverse biochemical reactions. The role of CoA and its thioester derivatives, including acetyl-CoA, malonyl-CoA and HMG-CoA (3-hydroxy-3-methylglutaryl-CoA), in the regulation of cellular metabolism has been extensively studied and documented. The main purpose of the present review is to summarize current knowledge on extracellular and intracellular signalling functions of CoA/CoA thioesters and to speculate on future developments in this area of research.


Cellular Signalling | 2013

Identification of the general transcription factor Yin Yang 1 as a novel and specific binding partner for S6 Kinase 2

Heba M. Ismail; Olena Myronova; Yugo Tsuchiya; Andrew Niewiarowski; Irina R. Tsaneva; Ivan Gout

S6 kinase is a member of the AGC family of serine/threonine kinases and plays a key role in diverse cellular processes including cell growth and metabolism. Although, the high degree of homology between S6K family members (S6K1 and S6K2) in kinase and kinase-extension domains, the two proteins are highly divergent in the N- and C-terminal regulatory regions, hinting at differential regulation, downstream signalling and cellular function. Deregulated signalling via S6Ks has been linked to various human pathologies, such as diabetes and cancer. Therefore, S6K has emerged as a promising target for drug development. Much of what we know about S6K signalling in health and disease comes from studies of S6K1, as molecular cloning of this isoform was reported a decade earlier than S6K2. In this study, we report for the first time, the identification of the general transcription factor Yin Yang 1 (YY1) as a novel and specific binding partner of S6K2, but not S6K1. The interaction between YY1 and S6K2 was demonstrated by co-immunoprecipitation of transiently overexpressed and endogenous proteins in a number of cell lines, including HEK293, MCF7 and U937. Furthermore, direct association between S6K2 and YY1 was demonstrated by GST pull-down assay using recombinant proteins. A panel of deletion mutants was used to show that the C-terminal regulatory region of S6K2 mediates the interaction with YY1. Interestingly, the complex formation between S6K2 and YY1 can be detected in serum-starved cells, but the interaction is strongly induced in response to mitogenic stimulation. The induction of S6K2/YY1 complex formation in response to serum stimulation is abolished by pre-treatment of cells with the mTOR inhibitor, rapamycin. Furthermore, mTOR is also detected in complex with YY1 and S6K2 in serum-stimulated cells. We utilized size exclusion chromatography along with co-immunoprecipitation analysis to demonstrate the existence of the mTOR/S6K2/YY1 complex in high molecular weight fractions, which might also involve other cellular proteins. The physiological significance of the mTOR/S6K2/YY1 complex, which is induced in response to mitogenic stimulation, remains to be further investigated.


Biochemical Journal | 2017

Protein CoAlation: A Redox-Regulated Protein Modification by Coenzyme A in Mammalian Cells

Yugo Tsuchiya; Sew Yeu Peak-Chew; Clare Newell; Sheritta Miller-Aidoo; Sriyash Mangal; Alexander Zhyvoloup; Jovana Baković; Oksana Malanchuk; Gonçalo C. Pereira; Vassilios N. Kotiadis; Michael R. Duchen; Mark Campbell; Sergio Rodriguez Cuenca; Antonio Vidal-Puig; Andrew M. James; Michael P. Murphy; Valeriy Filonenko; Mark Skehel; Ivan Gout

Coenzyme A (CoA) is an obligatory cofactor in all branches of life. CoA and its derivatives are involved in major metabolic pathways, allosteric interactions and the regulation of gene expression. Abnormal biosynthesis and homeostasis of CoA and its derivatives have been associated with various human pathologies, including cancer, diabetes and neurodegeneration. Using an anti-CoA monoclonal antibody and mass spectrometry, we identified a wide range of cellular proteins which are modified by covalent attachment of CoA to cysteine thiols (CoAlation). We show that protein CoAlation is a reversible post-translational modification that is induced in mammalian cells and tissues by oxidising agents and metabolic stress. Many key cellular enzymes were found to be CoAlated in vitro and in vivo in ways that modified their activities. Our study reveals that protein CoAlation is a widespread post-translational modification which may play an important role in redox regulation under physiological and pathophysiological conditions.


PLOS ONE | 2017

iPSC-derived neuronal models of PANK2-associated neurodegeneration reveal mitochondrial dysfunction contributing to early disease

Charles Arber; Plamena R. Angelova; Sarah Wiethoff; Yugo Tsuchiya; Francesca Mazzacuva; Elisavet Preza; Kailash P. Bhatia; Kevin D. Mills; Ivan Gout; Andrey Y. Abramov; John Hardy; James A. Duce; Henry Houlden; Selina Wray

Mutations in PANK2 lead to neurodegeneration with brain iron accumulation. PANK2 has a role in the biosynthesis of coenzyme A (CoA) from dietary vitamin B5, but the neuropathological mechanism and reasons for iron accumulation remain unknown. In this study, atypical patient-derived fibroblasts were reprogrammed into induced pluripotent stem cells (iPSCs) and subsequently differentiated into cortical neuronal cells for studying disease mechanisms in human neurons. We observed no changes in PANK2 expression between control and patient cells, but a reduction in protein levels was apparent in patient cells. CoA homeostasis and cellular iron handling were normal, mitochondrial function was affected; displaying activated NADH-related and inhibited FADH-related respiration, resulting in increased mitochondrial membrane potential. This led to increased reactive oxygen species generation and lipid peroxidation in patient-derived neurons. These data suggest that mitochondrial deficiency is an early feature of the disease process and can be explained by altered NADH/FADH substrate supply to oxidative phosphorylation. Intriguingly, iron chelation appeared to exacerbate the mitochondrial phenotype in both control and patient neuronal cells. This raises caution for the use iron chelation therapy in general when iron accumulation is absent.


Biochemical Society Transactions | 2014

Methods for measuring CoA and CoA derivatives in biological samples.

Yugo Tsuchiya; Uyen Pham; Ivan Gout

CoA (coenzyme A) is a ubiquitous and essential cofactor that acts as an acyl group carrier in biochemical reactions. Apart from participating in numerous metabolic pathways as substrates and intermediates, CoA and a number of its thioester derivatives, such as acetyl-CoA, can also directly regulate the activity of proteins by allosteric mechanisms and by affecting protein acetylation reactions. Cellular levels of CoA and CoA thioesters change under various physiological and pathological conditions. Defective CoA biosynthesis is implicated in NBIA (neurodegeneration with brain iron accumulation). However, the exact role of CoA in the pathogenesis of NBIA is not well understood. Accurate and reliable assays for measuring CoA species in biological samples are essential for studying the roles of CoA and CoA derivatives in health and disease. The present mini-review discusses methods that are commonly used to measure CoA species in biological samples.


Biochemical Journal | 2018

Protein CoAlation and antioxidant function of coenzyme A in prokaryotic cells

Yugo Tsuchiya; Alexander Zhyvoloup; Jovana Baković; Naam Thomas; Bess Yi Kun Yu; Sayoni Das; Christine Orengo; Clare Newell; John M. Ward; Giorgio Saladino; Federico Comitani; Francesco Luigi Gervasio; Oksana Malanchuk; A. I. Khoruzhenko; Valeriy Filonenko; Sew Yeu Peak-Chew; Mark Skehel; Ivan Gout

In all living organisms, coenzyme A (CoA) is an essential cofactor with a unique design allowing it to function as an acyl group carrier and a carbonyl-activating group in diverse biochemical reactions. It is synthesized in a highly conserved process in prokaryotes and eukaryotes that requires pantothenic acid (vitamin B5), cysteine and ATP. CoA and its thioester derivatives are involved in major metabolic pathways, allosteric interactions and the regulation of gene expression. A novel unconventional function of CoA in redox regulation has been recently discovered in mammalian cells and termed protein CoAlation. Here, we report for the first time that protein CoAlation occurs at a background level in exponentially growing bacteria and is strongly induced in response to oxidizing agents and metabolic stress. Over 12% of Staphylococcus aureus gene products were shown to be CoAlated in response to diamide-induced stress. In vitro CoAlation of S. aureus glyceraldehyde-3-phosphate dehydrogenase was found to inhibit its enzymatic activity and to protect the catalytic cysteine 151 from overoxidation by hydrogen peroxide. These findings suggest that in exponentially growing bacteria, CoA functions to generate metabolically active thioesters, while it also has the potential to act as a low-molecular-weight antioxidant in response to oxidative and metabolic stress.

Collaboration


Dive into the Yugo Tsuchiya's collaboration.

Top Co-Authors

Avatar

Ivan Gout

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clare Newell

University College London

View shared research outputs
Top Co-Authors

Avatar

Henry Houlden

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Jovana Baković

University College London

View shared research outputs
Top Co-Authors

Avatar

Mark Skehel

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Sew Yeu Peak-Chew

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Uyen Pham

University College London

View shared research outputs
Top Co-Authors

Avatar

Oksana Malanchuk

National Academy of Sciences of Ukraine

View shared research outputs
Top Co-Authors

Avatar

Valeriy Filonenko

National Academy of Sciences of Ukraine

View shared research outputs
Researchain Logo
Decentralizing Knowledge