Network


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

Hotspot


Dive into the research topics where Rasheduzzaman Chowdhury is active.

Publication


Featured researches published by Rasheduzzaman Chowdhury.


EMBO Reports | 2011

The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases.

Rasheduzzaman Chowdhury; Kar Kheng Yeoh; Ya-Min Tian; Lars Hillringhaus; Eleanor A. L. Bagg; Nathan R. Rose; Ivanhoe K. H. Leung; Xuan S Li; Esther C. Y. Woon; Ming Yang; Michael A. McDonough; Oliver N. King; Ian J. Clifton; Robert J. Klose; Timothy D. W. Claridge; Peter J. Ratcliffe; Christopher J. Schofield; Akane Kawamura

Mutations in isocitrate dehydrogenases (IDHs) have a gain‐of‐function effect leading to R(−)‐2‐hydroxyglutarate (R‐2HG) accumulation. By using biochemical, structural and cellular assays, we show that either or both R‐ and S‐2HG inhibit 2‐oxoglutarate (2OG)‐dependent oxygenases with varying potencies. Half‐maximal inhibitory concentration (IC50) values for the R‐form of 2HG varied from approximately 25 μM for the histone Nε‐lysine demethylase JMJD2A to more than 5 mM for the hypoxia‐inducible factor (HIF) prolyl hydroxylase. The results indicate that candidate oncogenic pathways in IDH‐associated malignancy should include those that are regulated by other 2OG oxygenases than HIF hydroxylases, in particular those involving the regulation of histone methylation.


Current Opinion in Structural Biology | 2010

Structural studies on human 2-oxoglutarate dependent oxygenases.

Michael A. McDonough; Christoph Loenarz; Rasheduzzaman Chowdhury; Ian J. Clifton; Christopher J. Schofield

2-Oxoglutarate and ferrous iron-dependent oxygenases have emerged as an important family of human enzymes that catalyse hydroxylations and related demethylation reactions. Their substrates in humans include proteins, nucleic acids, lipids and small molecules. They play roles in collagen biosynthesis, hypoxic sensing, regulation of gene expression and lipid biosynthesis/metabolism. Structural analyses, principally employing crystallography, have revealed that all of these oxygenases possess a double-stranded β-helix core fold that supports a highly conserved triad of iron binding residues and a less well conserved 2-oxoglutarate co-substrate binding site. The 2-oxoglutarate binds to the iron in a bidentate manner via its 1-carboxylate and 2-oxo groups. The primary substrate binding elements are more variable and can involve mobile elements.


Structure | 2009

Structural basis for binding of hypoxia-inducible factor to the oxygen-sensing prolyl hydroxylases

Rasheduzzaman Chowdhury; Michael A. McDonough; Jasmin Mecinović; Christoph Loenarz; Emily Flashman; Kirsty S. Hewitson; Carmen Domene; Christopher J. Schofield

The oxygen-dependent hydroxylation of proline residues in the alpha subunit of hypoxia-inducible transcription factor (HIFalpha) is central to the hypoxic response in animals. Prolyl hydroxylation of HIFalpha increases its binding to the von Hippel-Lindau protein (pVHL), so signaling for degradation via the ubiquitin-proteasome system. The HIF prolyl hydroxylases (PHDs, prolyl hydroxylase domain enzymes) are related to the collagen prolyl hydroxylases, but form unusually stable complexes with their Fe(II) cofactor and 2-oxoglutarate cosubstrate. We report crystal structures of the catalytic domain of PHD2, the most important of the human PHDs, in complex with the C-terminal oxygen-dependent degradation domain of HIF-1alpha. Together with biochemical analyses, the results reveal that PHD catalysis involves a mobile region that isolates the hydroxylation site and stabilizes the PHD2.Fe(II).2OG complex. The results will be of use in the design of PHD inhibitors aimed at treating anemia and ischemic disease.


Biochimica et Biophysica Acta | 2014

Targeting histone lysine demethylases - progress, challenges, and the future.

Cyrille C. Thinnes; Katherine S. England; Akane Kawamura; Rasheduzzaman Chowdhury; Christopher J. Schofield; Richard J. Hopkinson

N-Methylation of lysine and arginine residues has emerged as a major mechanism of transcriptional regulation in eukaryotes. In humans, Nε-methyllysine residue demethylation is catalysed by two distinct subfamilies of demethylases (KDMs), the flavin-dependent KDM1 subfamily and the 2-oxoglutarate- (2OG) dependent JmjC subfamily, which both employ oxidative mechanisms. Modulation of histone methylation status is proposed to be important in epigenetic regulation and has substantial medicinal potential for the treatment of diseases including cancer and genetic disorders. This article provides an introduction to the enzymology of the KDMs and the therapeutic possibilities and challenges associated with targeting them, followed by a review of reported KDM inhibitors and their mechanisms of action from kinetic and structural perspectives. This article is part of a Special Issue entitled: Methylation: A Multifaceted Modification — looking at transcription and beyond.


Current Opinion in Structural Biology | 2012

Role of the jelly-roll fold in substrate binding by 2-oxoglutarate oxygenases

WeiShen Aik; Michael A. McDonough; Armin Thalhammer; Rasheduzzaman Chowdhury; Christopher J. Schofield

2-Oxoglutarate (2OG) and ferrous iron dependent oxygenases catalyze two-electron oxidations of a range of small and large molecule substrates, including proteins/peptides/amino acids, nucleic acids/bases, and lipids, as well as natural products including antibiotics and signaling molecules. 2OG oxygenases employ variations of a core double-stranded β-helix (DSBH; a.k.a. jelly-roll, cupin or jumonji C (JmjC)) fold to enable binding of Fe(II) and 2OG in a subfamily conserved manner. The topology of the DSBH limits regions directly involved in substrate binding: commonly the first, second and eighth strands, loops between the second/third and fourth/fifth DSBH strands, and the N-terminal and C-terminal regions are involved in primary substrate, co-substrate and cofactor binding. Insights into substrate recognition by 2OG oxygenases will help to enable selective inhibition and bioengineering studies.


Nature Chemical Biology | 2012

Oxygenase-catalyzed ribosome hydroxylation occurs in prokaryotes and humans

Wei Ge; Alexander Wolf; Tianshu Feng; Chia Hua Ho; Rok Sekirnik; Adam Zayer; Nicolas Granatino; Matthew E. Cockman; Christoph Loenarz; Nikita D. Loik; Adam P. Hardy; Timothy D. W. Claridge; Refaat B. Hamed; Rasheduzzaman Chowdhury; Lingzhi Gong; Carol V. Robinson; David C. Trudgian; Miao Jiang; Mukram Mohamed Mackeen; James S. O. McCullagh; Yuliya Gordiyenko; Armin Thalhammer; Atsushi Yamamoto; Ming Yang; Phebee Liu-Yi; Zhihong Zhang; Marion S. Schmidt-Zachmann; Benedikt M. Kessler; Peter J. Ratcliffe; Gail M. Preston

The finding that oxygenase-catalyzed protein hydroxylation regulates animal transcription raises questions as to whether the translation machinery and prokaryotic proteins are analogously modified. Escherichia coli ycfD is a growth-regulating 2-oxoglutarate oxygenase catalyzing arginyl hydroxylation of the ribosomal protein Rpl16. Human ycfD homologs, Myc-induced nuclear antigen (MINA53) and NO66, are also linked to growth and catalyze histidyl hydroxylation of Rpl27a and Rpl8, respectively. This work reveals new therapeutic possibilities via oxygenase inhibition and by targeting modified over unmodified ribosomes.


Journal of Medicinal Chemistry | 2012

Plant Growth Regulator Daminozide Is a Selective Inhibitor of Human KDM2/7 Histone Demethylases

Nathan R. Rose; Esther C. Y. Woon; Anthony Tumber; Louise J. Walport; Rasheduzzaman Chowdhury; Xuan Shirley Li; Oliver N. King; Clarisse Lejeune; Stanley S. Ng; T. Krojer; Mun Chiang Chan; Anna M. Rydzik; Richard J. Hopkinson; Ka Hing Che; Michelle Daniel; C. Strain-Damerell; C. Gileadi; Grazyna Kochan; Ivanhoe K. H. Leung; J E Dunford; Kar Kheng Yeoh; Peter J. Ratcliffe; N. Burgess-Brown; Frank von Delft; Susanne Müller; Brian D. Marsden; Paul E. Brennan; Michael A. McDonough; U. Oppermann; Robert J. Klose

The JmjC oxygenases catalyze the N-demethylation of N(ε)-methyl lysine residues in histones and are current therapeutic targets. A set of human 2-oxoglutarate analogues were screened using a unified assay platform for JmjC demethylases and related oxygenases. Results led to the finding that daminozide (N-(dimethylamino)succinamic acid, 160 Da), a plant growth regulator, selectively inhibits the KDM2/7 JmjC subfamily. Kinetic and crystallographic studies reveal that daminozide chelates the active site metal via its hydrazide carbonyl and dimethylamino groups.


Angewandte Chemie | 2012

Dynamic Combinatorial Chemistry Employing Boronic Acids/Boronate Esters Leads to Potent Oxygenase Inhibitors

Marina Demetriades; Ivanhoe K. H. Leung; Rasheduzzaman Chowdhury; Mun Chiang Chan; Michael A. McDonough; Kar Kheng Yeoh; Ya-Min Tian; Timothy D. W. Claridge; Peter J. Ratcliffe; Esther C. Y. Woon; Christopher J. Schofield

The application of dynamic reactions is a promising approach for the discovery of small-molecule ligands for proteins. To date, however, this method is limited by the few appropriate reactions and the techniques used for the analysis of protein– ligand complexes. “Dynamic” functional group interconvertions that have been employed include the conversion of thiols to disulfides, the aldol reaction, and the addition of nucleophiles to ketones and aldehydes. The reaction of boronic acids with diols to form boronate esters is attractive for dynamic-library formation, because it is reversible in aqueous solution in a pH-dependent manner. The dynamic boronic acid/boronate ester system has been used to form supramolecular switches, some of which have been used for sugar detection. 5] However, this system has not been used for the identification of protein ligands. Proof of principle work with proteases, which react reversibly with boronic acids, suggests that boronic acid/boronate ester systems might be useful for the identification of enzyme inhibitors. One issue with the application of reversible reactions for ligand identification is the need to analyze labile complexes that are derived from mixtures. High-resolution techniques, such as NMR spectroscopy and X-ray crystallography, are applicable, but these are time-consuming. Our research group and that of Poulsen, have used non-denaturing protein mass spectrometry to identify protein–ligand complexes formed from equilibrating mixtures of thiols/disulfides and aldehydes/hydrazones. The dynamic-combinatorial mass spectrometry (DCMS) technique has the advantages of being efficient and providing information on mass shifts, which can be used for assigning structures to the ligands that bind preferentially. Herein we demonstrate that boronic acid/boronate ester dynamic systems coupled with protein mass spectrometry analysis are useful for the identification of protein inhibitors (Scheme 1). Our target model enzyme was prolyl hydroxylase domain isoform 2 (PHD2), which is a Fe and 2-oxoglutarate (2OG) oxygenase that regulates the human hypoxic response. PHD2 inhibition is of therapeutic interest for the treatment of anemia and ischemia-related diseases. DCMS experiments were carried out using “support ligands” 2 and 3 (Scheme 2), which were designed to participate in Fe chelation in the active site and, through the incorporation of a boronic acid moiety, participate in boronate ester exchange. We selected the 2-(picolinamido)acetic acid scaffold because, based on crystal structures of PHD2, it is predicted to fit into the active site through its chelation with Fe. The low potency of 2-(picolinamido)acetic acid (IC50> 1 mm) enabled the effect of boronate ester substitution to be monitored. Modeling studies suggested that whereas the boronic acid group in support ligand 2 would fit into the active-site subpocket, that of 3 would clash with the active-site wall. Hence, it was envisaged that the reactivity of 3 might serve as a control to investigate possible non-specific binding. The analysis of mixtures of 2 or 3 with PHD2·Fe through the use of non-denaturing ESI-MS led to the observation of a new peak at 27 887 Da (187 2 Da shift), corresponding to a small molecule/protein adduct, in which the OH groups of the boronic acids moiety are cleaved. We have previously observed, through the use of non-denaturing ESI-MS, analogous apparent fragmentation of boronic acids complexed with other enzymes. Notably, the mixture of boronate ester 4 and PHD2·Fe gave the same mass shift (187 2 Da) as that observed with 2 and 3 at a cone voltage of 80 V. However, when a lower cone voltage was used (30 V), the mass shift corresponding to an adduct of 4 with the protein, without fragmentation, was apparent (358 2 Da), demonstrating that boronate ester formation can be observed when sufficiently mild ionization is used. Both 2 and 3 compete with the 2OG analogue N-oxalylglycine (NOG) for the 2OG binding site of PHD2. To ensure that boronate ester formation involving 2 and 3 was favorable under the conditions used (NH4OAc [*] M. Demetriades, I. K. H. Leung, Dr. R. Chowdhury, M. C. Chan, Dr. M. A. McDonough, Dr. K. K. Yeoh, Dr. T. D. W. Claridge, Prof. C. J. Schofield Chemistry Research Laboratory, University of Oxford 12 Mansfield Road, Oxford, OX1 3TA (UK) E-mail: [email protected]


Chemical Science | 2013

5-Carboxy-8-hydroxyquinoline is a Broad Spectrum 2-Oxoglutarate Oxygenase Inhibitor which Causes Iron Translocation.

Richard J. Hopkinson; Anthony Tumber; Clarence Yapp; Rasheduzzaman Chowdhury; WeiShen Aik; Ka Hing Che; Xuan Shirley Li; Jan Kristensen; Oliver N. King; Mun Chiang Chan; Kar Kheng Yeoh; Hwanho Choi; Louise J. Walport; Cyrille C. Thinnes; Jacob T. Bush; Clarisse Lejeune; Anna M. Rydzik; Nathan R. Rose; Eleanor A. L. Bagg; Michael A. McDonough; T. Krojer; W.W. Yue; Stanley S. Ng; Lars Olsen; Paul E. Brennan; U. Oppermann; Susanne Müller-Knapp; Robert J. Klose; Peter J. Ratcliffe; Christopher J. Schofield

2-Oxoglutarate and iron dependent oxygenases are therapeutic targets for human diseases. Using a representative 2OG oxygenase panel, we compare the inhibitory activities of 5-carboxy-8-hydroxyquinoline (IOX1) and 4-carboxy-8-hydroxyquinoline (4C8HQ) with that of two other commonly used 2OG oxygenase inhibitors, N-oxalylglycine (NOG) and 2,4-pyridinedicarboxylic acid (2,4-PDCA). The results reveal that IOX1 has a broad spectrum of activity, as demonstrated by the inhibition of transcription factor hydroxylases, representatives of all 2OG dependent histone demethylase subfamilies, nucleic acid demethylases and γ-butyrobetaine hydroxylase. Cellular assays show that, unlike NOG and 2,4-PDCA, IOX1 is active against both cytosolic and nuclear 2OG oxygenases without ester derivatisation. Unexpectedly, crystallographic studies on these oxygenases demonstrate that IOX1, but not 4C8HQ, can cause translocation of the active site metal, revealing a rare example of protein ligand-induced metal movement.


Journal of Biological Chemistry | 2008

Kinetic Rationale for Selectivity toward N- and C-terminal Oxygen-dependent Degradation Domain Substrates Mediated by a Loop Region of Hypoxia-Inducible Factor Prolyl Hydroxylases

Emily Flashman; Eleanor A. L. Bagg; Rasheduzzaman Chowdhury; Jasmin Mecinović; Christoph Loenarz; Michael A. McDonough; Kirsty S. Hewitson; Christopher J. Schofield

Hydroxylation of two conserved prolyl residues in the N- and C-terminal oxygen-dependent degradation domains (NODD and CODD) of the α-subunit of hypoxia-inducible factor (HIF) signals for its degradation via the ubiquitin-proteasome pathway. In human cells, three prolyl hydroxylases (PHDs 1–3) belonging to the Fe(II) and 2-oxoglutarate (2OG)-dependent oxygenase family catalyze prolyl hydroxylation with differing selectivity for CODD and NODD. Sequence analysis of the catalytic domains of the PHDs in the light of crystal structures for PHD2, and results for other 2OG oxygenases, suggested that either the C-terminal region or a loop linking two β-strands (β2 and β3 in human PHD2) are important in determining substrate selectivity. Mutation analyses on PHD2 revealed that the β2β3 loop is a major determinant in conferring selectivity for CODD over NODD peptides. A chimeric PHD in which the β2β3 loop of PHD2 was replaced with that of PHD3 displayed an almost complete selectivity for CODD (in competition experiments), as observed for wild-type PHD3. CODD was observed to bind much more tightly to this chimeric protein than the wild type PHD2 catalytic domain.

Collaboration


Dive into the Rasheduzzaman Chowdhury's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge