Network


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

Hotspot


Dive into the research topics where Nham T. Nguyen is active.

Publication


Featured researches published by Nham T. Nguyen.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Structural basis of collagen fiber degradation by cathepsin K

Adeleke H. Aguda; Preety Panwar; Xin Du; Nham T. Nguyen; Gary D. Brayer; Dieter Brömme

Significance Fibrillar collagens constitute 90% of the organic bone matrix and are subjected either to physiological remodeling or excessive degradation during diseases such as osteoporosis. Cathepsin K is the critical collagenase in bone and represents a major antiresorptive drug target. Despite its critical role in bone remodeling, its mechanism of collagen degradation remained elusive. Here, we demonstrate that the degradation of fibrillar collagen requires the presence of a cathepsin K dimer bound at the surface of collagen fibers via glycosaminoglycans. Structural modifications of the protease dimerization site or the removal of collagen fiber-associated glycosaminoglycans specifically block fibrillar collagen degradation. The provided structure allows the development of a strategy to inhibit this highly relevant drug target in a substrate-specific manner. Cathepsin K is the major collagenolytic protease in bone that facilitates physiological as well as pathological bone degradation. Despite its key role in bone remodeling and for being a highly sought-after drug target for the treatment of osteoporosis, the mechanism of collagen fiber degradation by cathepsin K remained elusive. Here, we report the structure of a collagenolytically active cathepsin K protein dimer. Cathepsin K is organized into elongated C-shaped protease dimers that reveal a putative collagen-binding interface aided by glycosaminoglycans. Molecular modeling of collagen binding to the dimer indicates the participation of nonactive site amino acid residues, Q21 and Q92, in collagen unfolding. Mutations at these sites as well as perturbation of the dimer protein–protein interface completely inhibit cathepsin-K–mediated fiber degradation without affecting the hydrolysis of gelatin or synthetic peptide. Using scanning electron microscopy, we demonstrate the specific binding of cathepsin K at the edge of the fibrillar gap region of collagen fibers, which suggest initial cleavage events at the N- and C-terminal ends of tropocollagen molecules. Edman degradation analysis of collagen fiber degradation products revealed those initial cleavage sites. We propose that one cathepsin K molecule binds to collagen-bound glycosaminoglycans at the gap region and recruits a second protease molecule that provides an unfolding and cleavage mechanism for triple helical collagen. Removal of collagen-associated glycosaminoglycans prevents cathepsin K binding and subsequently fiber hydrolysis. Cathepsin K dimer and glycosaminoglycan binding sites represent novel targeting sites for the development of nonactive site-directed second-generation inhibitors of this important drug target.


Biochemical Journal | 1998

Structural and spectroscopic studies of azide complexes of horse heart myoglobin and the His-64-->Thr variant.

Robert Maurus; R Bogumil; Nham T. Nguyen; and A. Grant Mauk; Gary D. Brayer

The high-resolution X-ray crystallographic structures of horse heart azidometmyoglobin complexes of the wild-type protein and the His-64-->Thr variant have been determined to 2.0 and 1.8 A respectively. Azide binds to wild-type metmyoglobin in a bent configuration with an Fe-N-1-N-3 angle of 119 degrees and is oriented into the distal crevice in the direction of Ile-107. The proximity of the His-64 NE2 atom to the N-1 atom of the bound azide indicates stabilization of the ligand by the His-64 side chain through hydrogen bonding. In addition, structural characterization of wild-type horse heart azidometmyoglobin establishes that the only structural change induced by ligand binding is a small movement of the Leu-29 side chain away from the azide ligand. EPR and Fourier transform infrared spectroscopy were used to characterize the myoglobin azide complexes further. EPR spectroscopy revealed that, in contrast with wild-type azidometmyoglobin, two slightly different low-spin species are formed by azide bound to the His-64-->Thr variant both in solution and in a polycrystalline sample. One of these low-spin species has a greater relative intensity, with g values very similar to those of the azide complex of the wild-type protein. These EPR results together with structural information on this variant indicate the presence of two distinct conformations of bound azide, with one form predominating. The major conformation is comparable to that formed by wild-type myoglobin in which azide is oriented into the distal crevice. In the minor conformation the azide is oriented towards the exterior of the protein.


Nature Chemical Biology | 2015

The amylase inhibitor montbretin A reveals a new glycosidase inhibition motif

Leslie K. Williams; Xiaohua Zhang; Sami Caner; Christina Tysoe; Nham T. Nguyen; Jacqueline Wicki; David E. Williams; John Coleman; John H. McNeill; Violet G. Yuen; Raymond J. Andersen; Stephen G. Withers; Gary D. Brayer

The complex plant flavonol glycoside montbretin A is a potent (Ki = 8 nM) and specific inhibitor of human pancreatic α-amylase with potential as a therapeutic for diabetes and obesity. Controlled degradation studies on montbretin A, coupled with inhibition analyses, identified an essential high-affinity core structure comprising the myricetin and caffeic acid moieties linked via a disaccharide. X-ray structural analyses of the montbretin A-human α-amylase complex confirmed the importance of this core structure and revealed a novel mode of glycosidase inhibition wherein internal π-stacking interactions between the myricetin and caffeic acid organize their ring hydroxyls for optimal hydrogen bonding to the α-amylase catalytic residues D197 and E233. This novel inhibitory motif can be reproduced in a greatly simplified analog, offering potential for new strategies for glycosidase inhibition and therapeutic development.


Journal of Biological Chemistry | 2009

Structure, Binding, and Activity of Syd, a SecY-interacting Protein

Kush Dalal; Nham T. Nguyen; Meriem Alami; Jennifer Tan; Trevor F. Moraes; Woo Cheol Lee; Robert Maurus; Stephen S. Sligar; Gary D. Brayer; Franck Duong

The Syd protein has been implicated in the Sec-dependent transport of polypeptides across the bacterial inner membrane. Using Nanodiscs, we here provide direct evidence that Syd binds the SecY complex, and we demonstrate that interaction involves the two electropositive and cytosolic loops of the SecY subunit. We solve the crystal structure of Syd and together with cysteine cross-link analysis, we show that a conserved concave and electronegative groove constitutes the SecY-binding site. At the membrane, Syd decreases the activity of the translocon containing loosely associated SecY-SecE subunits, whereas in detergent solution Syd disrupts the SecYEG heterotrimeric associations. These results support the role of Syd in proofreading the SecY complex biogenesis and point to the electrostatic nature of the Sec channel interaction with its cytosolic partners.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Introduction and characterization of a functionally linked metal ion binding site at the exposed heme edge of myoglobin

C.L Hunter; Robert Maurus; M.R Mauk; H Lee; Emma Lloyd Raven; H Tong; Nham T. Nguyen; M. Smith; Gary D. Brayer; and A. Grant Mauk

A binding site for metal ions has been created on the surface of horse heart myoglobin (Mb) near the heme 6-propionate group by replacing K45 and K63 with glutamyl residues. One-dimensional 1H NMR spectroscopy indicates that Mn2+ binds in the vicinity of the heme 6-propionate as anticipated, and potentiometric titrations establish that the affinity of the new site for Mn2+ is 1.28(4) × 104 M−1 (pH 6.96, ionic strength I = 17.2 μM, 25°C). In addition, these substitutions lower the reduction potential of the protein and increase the pKa for the water molecule coordinated to the heme iron of metmyoglobin. The peroxidase [2,2′-azinobis(3-ethylbenzthiazoline-6-sulfonic acid), ABTS, as substrate] and the Mn2+-peroxidase activity of the variant are both increased ≈3-fold. In contrast to wild-type Mb, both the affinity for azide and the midpoint potential of the variant are significantly influenced by the addition of Mn2+. The structure of the variant has been determined by x-ray crystallography to define the coordination environment of bound Mn2+ and Cd2+. Although slight differences are observed between the geometry of the binding of the two metal ions, both are hexacoordinate, and neither involves coordination by E63.


Glycobiology | 2013

The structure of the Mycobacterium smegmatis trehalose synthase reveals an unusual active site configuration and acarbose-binding mode

Sami Caner; Nham T. Nguyen; Adeleke H. Aguda; Ran Zhang; Yuan T. Pan; Stephen G. Withers; Gary D. Brayer

Trehalose synthase (TreS) catalyzes the reversible conversion of maltose into trehalose in mycobacteria as one of three biosynthetic pathways to this nonreducing disaccharide. Given the importance of trehalose to survival of mycobacteria, there has been considerable interest in understanding the enzymes involved in its production; indeed the structures of the key enzymes in the other two pathways have already been determined. Herein, we present the first structure of TreS from Mycobacterium smegmatis, thereby providing insights into the catalytic machinery involved in this intriguing intramolecular reaction. This structure, which is of interest both mechanistically and as a potential pharmaceutical target, reveals a narrow and enclosed active site pocket within which intramolecular substrate rearrangements can occur. We also present the structure of a complex of TreS with acarbose, revealing a hitherto unsuspected oligosaccharide-binding site within the C-terminal domain. This may well provide an anchor point for the association of TreS with glycogen, thereby enhancing its role in glycogen biosynthesis and degradation.


ACS central science | 2016

Potent human α-amylase inhibition by the β-defensin-like protein helianthamide

Christina Tysoe; Leslie K. Williams; Robert A. Keyzers; Nham T. Nguyen; Chris A. Tarling; Jacqueline Wicki; Ethan D. Goddard-Borger; Adeleke H. Aguda; Suzanne Perry; Leonard J. Foster; Raymond J. Andersen; Gary D. Brayer; Stephen G. Withers

Selective inhibitors of human pancreatic α-amylase (HPA) are an effective means of controlling blood sugar levels in the management of diabetes. A high-throughput screen of marine natural product extracts led to the identification of a potent (Ki = 10 pM) peptidic HPA inhibitor, helianthamide, from the Caribbean sea anemone Stichodactyla helianthus. Active helianthamide was produced in Escherichia coli via secretion as a barnase fusion protein. X-ray crystallographic analysis of the complex of helianthamide with porcine pancreatic α-amylase revealed that helianthamide adopts a β-defensin fold and binds into and across the amylase active site, utilizing a contiguous YIYH inhibitory motif. Helianthamide represents the first of a novel class of glycosidase inhibitors and provides an unusual example of functional malleability of the β-defensin fold, which is rarely seen outside of its traditional role in antimicrobial peptides.


Biochemical Journal | 2017

Identification of mouse cathepsin K structural elements that regulate the potency of odanacatib.

Simon Law; Pierre-Marie Andrault; Adeleke H. Aguda; Nham T. Nguyen; Natasha Kruglyak; Gary D. Brayer; Dieter Brömme

Cathepsin K (CatK) is the predominant mammalian bone-degrading protease and thus an ideal target for antiosteoporotic drug development. Rodent models of osteoporosis are preferred due to their close reflection of the human disease and their ease of handling, genetic manipulation and economic affordability. However, large differences in the potency of CatK inhibitors for the mouse/rat vs. the human protease orthologs have made it impossible to use rodent models. This is even more of a problem considering that the most advanced CatK inhibitors, including odanacatib (ODN) and balicatib, failed in human clinical trials due to side effects and rodent models are not available to investigate the mechanism of these failures. Here, we elucidated the structural elements of the potency differences between mouse and human CatK (hCatK) using ODN. We determined and compared the structures of inhibitor-free mouse CatK (mCatK), hCatK and ODN bound to hCatK. Two structural differences were identified and investigated by mutational analysis. Humanizing subsite 2 in mCatK led to a 5-fold improvement of ODN binding, whereas the replacement of Tyr61 in mCatK with Asp resulted in an hCatK with comparable ODN potency. Combining both sites further improved the inhibition of the mCatK variant. Similar results were obtained for balicatib. These findings will allow the generation of transgenic CatK mice that will facilitate the evaluation of CatK inhibitor adverse effects and to explore routes to avoid them.


Journal of Natural Products | 2016

Affinity Crystallography: A New Approach to Extracting High-Affinity Enzyme Inhibitors from Natural Extracts.

Adeleke H. Aguda; Vincent Lavallee; Ping Cheng; Tina M. Bott; Labros G. Meimetis; Simon Law; Nham T. Nguyen; David E. Williams; Jadwiga Kaleta; Ivan Villanueva; Julian Davies; Raymond J. Andersen; Gary D. Brayer; Dieter Brömme

Natural products are an important source of novel drug scaffolds. The highly variable and unpredictable timelines associated with isolating novel compounds and elucidating their structures have led to the demise of exploring natural product extract libraries in drug discovery programs. Here we introduce affinity crystallography as a new methodology that significantly shortens the time of the hit to active structure cycle in bioactive natural product discovery research. This affinity crystallography approach is illustrated by using semipure fractions of an actinomycetes culture extract to isolate and identify a cathepsin K inhibitor and to compare the outcome with the traditional assay-guided purification/structural analysis approach. The traditional approach resulted in the identification of the known inhibitor antipain (1) and its new but lower potency dehydration product 2, while the affinity crystallography approach led to the identification of a new high-affinity inhibitor named lichostatinal (3). The structure and potency of lichostatinal (3) was verified by total synthesis and kinetic characterization. To the best of our knowledge, this is the first example of isolating and characterizing a potent enzyme inhibitor from a partially purified crude natural product extract using a protein crystallographic approach.


Biochimica et Biophysica Acta | 2013

Enzyme-substrate complexes of allosteric citrate synthase: Evidence for a novel intermediate in substrate binding.

Harry W. Duckworth; Nham T. Nguyen; Yin Gao; Lynda J. Donald; Robert Maurus; Ayeda Ayed; Brigitte Bruneau; Gary D. Brayer

The citrate synthase (CS) of Escherichia coli is an allosteric hexameric enzyme specifically inhibited by NADH. The crystal structure of wild type (WT) E. coli CS, determined by us previously, has no substrates bound, and part of the active site is in a highly mobile region that is shifted from the position needed for catalysis. The CS of Acetobacter aceti has a similar structure, but has been successfully crystallized with bound substrates: both oxaloacetic acid (OAA) and an analog of acetyl coenzyme A (AcCoA). We engineered a variant of E. coli CS wherein five amino acids in the mobile region have been replaced by those in the A. aceti sequence. The purified enzyme shows unusual kinetics with a low affinity for both substrates. Although the crystal structure without ligands is very similar to that of the WT enzyme (except in the mutated region), complexes are formed with both substrates and the allosteric inhibitor NADH. The complex with OAA in the active site identifies a novel OAA-binding residue, Arg306, which has no functional counterpart in other known CS-OAA complexes. This structure may represent an intermediate in a multi-step substrate binding process where Arg306 changes roles from OAA binding to AcCoA binding. The second complex has the substrate analog, S-carboxymethyl-coenzyme A, in the allosteric NADH-binding site and the AcCoA site is not formed. Additional CS variants unable to bind adenylates at the allosteric site show that this second complex is not a factor in positive allosteric activation of AcCoA binding.

Collaboration


Dive into the Nham T. Nguyen's collaboration.

Top Co-Authors

Avatar

Gary D. Brayer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert Maurus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Adeleke H. Aguda

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Stephen G. Withers

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sami Caner

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayeda Ayed

University of Manitoba

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dieter Brömme

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Raymond J. Andersen

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge