Network


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

Hotspot


Dive into the research topics where Syed Shahzad-ul-Hussan is active.

Publication


Featured researches published by Syed Shahzad-ul-Hussan.


Nature | 2011

Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9

Jason S. McLellan; Marie Pancera; Chris Carrico; Jason Gorman; Jean-Philippe Julien; Reza Khayat; Robert K. Louder; Robert Pejchal; Mallika Sastry; Kaifan Dai; Sijy O’Dell; Nikita Patel; Syed Shahzad-ul-Hussan; Yongping Yang; Baoshan Zhang; Tongqing Zhou; Jiang Zhu; Jeffrey C. Boyington; Gwo-Yu Chuang; Devan Diwanji; Ivelin S. Georgiev; Young Do Kwon; Doyung Lee; Mark K. Louder; Stephanie Moquin; Stephen D. Schmidt; Zhi-Yong Yang; Mattia Bonsignori; John A. Crump; Saidi Kapiga

Variable regions 1 and 2 (V1/V2) of human immunodeficiency virus-1 (HIV-1) gp120 envelope glycoprotein are critical for viral evasion of antibody neutralization, and are themselves protected by extraordinary sequence diversity and N-linked glycosylation. Human antibodies such as PG9 nonetheless engage V1/V2 and neutralize 80% of HIV-1 isolates. Here we report the structure of V1/V2 in complex with PG9. V1/V2 forms a four-stranded β-sheet domain, in which sequence diversity and glycosylation are largely segregated to strand-connecting loops. PG9 recognition involves electrostatic, sequence-independent and glycan interactions: the latter account for over half the interactive surface but are of sufficiently weak affinity to avoid autoreactivity. The structures of V1/V2-directed antibodies CH04 and PGT145 indicate that they share a common mode of glycan penetration by extended anionic loops. In addition to structurally defining V1/V2, the results thus identify a paradigm of antibody recognition for highly glycosylated antigens, which—with PG9—involves a site of vulnerability comprising just two glycans and a strand.


Nature Structural & Molecular Biology | 2013

Structural basis for diverse N-glycan recognition by HIV-1–neutralizing V1–V2–directed antibody PG16

Marie Pancera; Syed Shahzad-ul-Hussan; Nicole A. Doria-Rose; Jason S. McLellan; Robert T. Bailer; Kaifan Dai; Sandra Loesgen; Mark K. Louder; Ryan P. Staupe; Yongping Yang; Baoshan Zhang; Robert Parks; Joshua Eudailey; Krissey E. Lloyd; Julie Blinn; S. Munir Alam; Barton F. Haynes; Mohammed N. Amin; Lai-Xi Wang; Dennis R. Burton; Wayne C. Koff; Gary J. Nabel; John R. Mascola; Carole A. Bewley; Peter D. Kwong

HIV-1 uses a diverse N-linked-glycan shield to evade recognition by antibody. Select human antibodies, such as the clonally related PG9 and PG16, recognize glycopeptide epitopes in the HIV-1 V1–V2 region and penetrate this shield, but their ability to accommodate diverse glycans is unclear. Here we report the structure of antibody PG16 bound to a scaffolded V1–V2, showing an epitope comprising both high mannose–type and complex-type N-linked glycans. We combined structure, NMR and mutagenesis analyses to characterize glycan recognition by PG9 and PG16. Three PG16-specific residues, arginine, serine and histidine (RSH), were critical for binding sialic acid on complex-type glycans, and introduction of these residues into PG9 produced a chimeric antibody with enhanced HIV-1 neutralization. Although HIV-1–glycan diversity facilitates evasion, antibody somatic diversity can overcome this and can provide clues to guide the design of modified antibodies with enhanced neutralization.


Journal of Virology | 2012

Structural Basis for Norovirus Inhibition and Fucose Mimicry by Citrate

Grant S. Hansman; Syed Shahzad-ul-Hussan; Jason S. McLellan; Gwo-Yu Chuang; Ivelin S. Georgiev; Takashi Shimoike; Kazuhiko Katayama; Carole A. Bewley; Peter D. Kwong

ABSTRACT Human noroviruses bind with their capsid-protruding domains to histo-blood-group antigens (HBGAs), an interaction thought to direct their entry into cells. Although human noroviruses are the major cause of gastroenteritis outbreaks, development of antivirals has been lacking, mainly because human noroviruses cannot be cultivated. Here we use X-ray crystallography and saturation transfer difference nuclear magnetic resonance (STD NMR) to analyze the interaction of citrate with genogroup II (GII) noroviruses. Crystals of citrate in complex with the protruding domain from norovirus GII.10 Vietnam026 diffracted to 1.4 Å and showed a single citrate bound at the site of HBGA interaction. The citrate interaction was coordinated with a set of capsid interactions almost identical to that involved in recognizing the terminal HBGA fucose, the saccharide which forms the primary conserved interaction between HBGAs and GII noroviruses. Citrate and a water molecule formed a ring-like structure that mimicked the pyranoside ring of fucose. STD NMR showed the protruding domain to have weak affinity for citrate (460 μM). This affinity, however, was similar to the affinities of the protruding domain for fucose (460 μM) and H type 2 trisaccharide (390 μM), an HBGA shown previously to be specifically recognized by human noroviruses. Importantly, competition STD NMR showed that citrate could compete with HBGA for norovirus binding. Together, the results suggest that citrate and other glycomimetics have the potential to block human noroviruses from binding to HBGAs.


Journal of Biological Chemistry | 2011

Solution Structure of the Monovalent Lectin Microvirin in Complex with Manα(1–2)Man Provides a Basis for Anti-HIV Activity with Low Toxicity

Syed Shahzad-ul-Hussan; Elena Gustchina; Rodolfo Ghirlando; G. Marius Clore; Carole A. Bewley

Lectins that bind surface envelope glycoprotein gp120 of HIV with high avidity can potently inhibit viral entry. Yet properties such as multivalency that facilitate strong interactions can also cause nonspecific binding and toxicity. The cyanobacterial lectin microvirin (MVN) is unusual as it potently inhibits HIV-1 with negligible toxicity compared with cyanovirin-N (CVN), its well studied antiviral homolog. To understand the structural and mechanistic basis for these differences, we solved the solution structure of MVN free and in complex with its ligand Manα(1–2)Man, and we compared specificity and time windows of inhibition with CVN and Manα(1–2)Man-specific mAb 2G12. We show by NMR and analytical ultracentrifugation that MVN is monomeric in solution, and we demonstrate by NMR that Manα(1–2)Man-terminating carbohydrates interact with a single carbohydrate-binding site. Synchronized infectivity assays show that 2G12, MVN, and CVN inhibit entry with distinct kinetics. Despite shared specificity for Manα(1–2)Man termini, combinations of the inhibitors are synergistic suggesting they recognize discrete glycans and/or dynamic glycan conformations on gp120. Entry assays employing amphotropic viruses show that MVN is inactive, whereas CVN potently inhibits both. In addition to demonstrating that HIV-1 can be inhibited through monovalent interactions, given the similarity of the carbohydrate-binding site common to MVN and CVN, these data suggest that gp120 behaves as a clustered glycan epitope and that multivalent-protein interactions achievable with CVN but not MVN are required for inhibition of some viruses.


Journal of Biological Chemistry | 2012

Peptides from Second Extracellular Loop of C-C Chemokine Receptor Type 5 (CCR5) Inhibit Diverse Strains of HIV-1

Cajetan Dogo-Isonagie; Son N. Lam; Elena Gustchina; Priyamvada Acharya; Yongping Yang; Syed Shahzad-ul-Hussan; G. Marius Clore; Peter D. Kwong; Carole A. Bewley

Background: Extracellular regions ECL2 and the N terminus of HIV coreceptor CCR5 mediate HIV-1 entry. Results: A C-terminal CCR5 ECL2 peptide inhibits HIV-1 entry and binds to gp120 of CCR5- and CXCR4-using strains. Conclusion: The binding site for CCR5 ECL2 is conserved in CCR5- and CXCR4-using viruses. Significance: Our data provide new insights into HIV-1 gp120-CCR5 interactions that can be used for inhibitor design. To initiate HIV entry, the HIV envelope protein gp120 must engage its primary receptor CD4 and a coreceptor CCR5 or CXCR4. In the absence of a high resolution structure of a gp120-coreceptor complex, biochemical studies of CCR5 have revealed the importance of its N terminus and second extracellular loop (ECL2) in binding gp120 and mediating viral entry. Using a panel of synthetic CCR5 ECL2-derived peptides, we show that the C-terminal portion of ECL2 (2C, comprising amino acids Cys-178 to Lys-191) inhibit HIV-1 entry of both CCR5- and CXCR4-using isolates at low micromolar concentrations. In functional viral assays, these peptides inhibited HIV-1 entry in a CD4-independent manner. Neutralization assays designed to measure the effects of CCR5 ECL2 peptides when combined with either with the small molecule CD4 mimetic NBD-556, soluble CD4, or the CCR5 N terminus showed additive inhibition for each, indicating that ECL2 binds gp120 at a site distinct from that of N terminus and acts independently of CD4. Using saturation transfer difference NMR, we determined the region of CCR5 ECL2 used for binding gp120, showed that it can bind to gp120 from both R5 and X4 isolates, and demonstrated that the peptide interacts with a CD4-gp120 complex in a similar manner as to gp120 alone. As the CCR5 N terminus-gp120 interactions are dependent on CD4 activation, our data suggest that gp120 has separate binding sites for the CCR5 N terminus and ECL2, the ECL2 binding site is present prior to CD4 engagement, and it is conserved across CCR5- and CXCR4-using strains. These peptides may serve as a starting point for the design of inhibitors with broad spectrum anti-HIV activity.


Molecular Pharmaceutics | 2013

Inhibition of Hepatitis C Virus by the Cyanobacterial Protein Microcystis viridis Lectin: Mechanistic Differences between the High-Mannose Specific Lectins MVL, CV-N, and GNA

Alla Kachko; Sandra Loesgen; Syed Shahzad-ul-Hussan; Wendy Tan; Iryna Zubkova; Kazuyo Takeda; Frances Wells; Steven Rubin; Carole A. Bewley; Marian E. Major

Plant or microbial lectins are known to exhibit potent antiviral activities against viruses with glycosylated surface proteins, yet the mechanism(s) by which these carbohydrate-binding proteins exert their antiviral activities is not fully understood. Hepatitis C virus (HCV) is known to possess glycosylated envelope proteins (gpE1E2) and to be potently inhibited by lectins. Here, we tested in detail the antiviral properties of the newly discovered Microcystis viridis lectin (MVL) along with cyanovirin-N (CV-N) and Galanthus nivalis agglutinin (GNA) against cell culture HCV, as well as their binding properties toward viral particles, target cells, and recombinant HCV glycoproteins. Using infectivity assays, CV-N, MVL, and GNA inhibited HCV with IC50 values of 0.6 nM, 30.4 nM, and 11.1 nM, respectively. Biolayer interferometry analysis demonstrated a higher affinity of GNA to immobilized recombinant HCV glycoproteins compared to CV-N and MVL. Complementary studies, including fluorescence-activated cell sorting (FACS) analysis, confocal microscopy, and pre- and post-virus binding assays, showed a complex mechanism of inhibition for CV-N and MVL that includes both viral and cell association, while GNA functions by binding directly to the viral particle. Combinations of GNA with CV-N or MVL in HCV infection studies revealed synergistic inhibitory effects, which can be explained by different glycan recognition profiles of the mainly high-mannoside specific lectins, and supports the hypothesis that these lectins inhibit through different and complex modes of action. Our findings provide important insights into the mechanisms by which lectins inhibit HCV infection. Overall, the data suggest MVL and CV-N have the potential for toxicity due to interactions with cellular proteins while GNA may be a better therapeutic agent due to specificity for the HCV gpE1E2.


Biopolymers | 2013

Characterizing carbohydrate-protein interactions by nuclear magnetic resonance spectroscopy.

Carole A. Bewley; Syed Shahzad-ul-Hussan

Interactions between proteins and soluble carbohydrates and/or surface displayed glycans are central to countless recognition, attachment and signaling events in biology. The physical chemical features associated with these binding events vary considerably, depending on the biological system of interest. For example, carbohydrate-protein interactions can be stoichiometric or multivalent, the protein receptors can be monomeric or oligomeric, and the specificity of recognition can be highly stringent or rather promiscuous. Equilibrium dissociation constants for carbohydrate binding are known to vary from micromolar to millimolar, with weak interactions being far more prevalent; and individual carbohydrate-binding sites can be truly symmetrical or merely homologous, and hence, the affinities of individual sites within a single protein can vary, as can the order of binding. Several factors, including the weak affinities with which glycans bind their protein receptors, the dynamic nature of the glycans themselves, and the nonequivalent interactions among oligomeric carbohydrate receptors, have made nuclear magnetic resonance (NMR) an especially powerful tool for studying and defining carbohydrate-protein interactions. Here, we describe those NMR approaches that have proven to be the most robust in characterizing these systems, and explain what type of information can (or cannot) be obtained from each. Our goal is to provide the reader the information necessary for selecting the correct experiment or sets of experiments to characterize their carbohydrate-protein interaction of interest.


Journal of the American Chemical Society | 2012

Characterization and Carbohydrate Specificity of Pradimicin S

Syed Shahzad-ul-Hussan; Rodolfo Ghirlando; Cajetan Dogo-Isonagie; Yasuhiro Igarashi; Jan Balzarini; Carole A. Bewley

The pradimicin family of antibiotics is attracting attention due to its anti-infective properties and as a model for understanding the requirements for carbohydrate recognition by small molecules. Members of the pradimicin family are unique among natural products in their ability to bind sugars in a Ca(2+)-dependent manner, but the oligomerization to insoluble aggregates that occurs upon Ca(2+) binding has prevented detailed characterization of their carbohydrate specificity and biologically relevant form. Here we take advantage of the water solubility of pradimicin S (PRM-S), a sulfated glucose-containing analogue of pradimicin A (PRM-A), to show by NMR spectroscopy and analytical ultracentrifugation that at biologically relevant concentrations, PRM-S binds Ca(2+) to form a tetrameric species that selectively binds and engulfs the trisaccharide Manα1-3(Manα1-6)Man over mannose or mannobiose. In functional HIV-1 entry assays, IC(50) values of 2-4 μM for PRM-S corrrelate with the concentrations at which oligomerization occurs as well as the affinities with which PRM-S binds the HIV surface envelope glycoprotein gp120. Together these data reveal the biologically active form of PRM-S, provide an explanation for previous speculations that PRM-A may contain a second mannose binding site, and expand our understanding of the characteristics that can engender a small molecule with the ability to function as a carbohydrate receptor.


Immunity | 2018

A Neutralizing Antibody Recognizing Primarily N-Linked Glycan Targets the Silent Face of the HIV Envelope

Tongqing Zhou; Anqi Zheng; Ulrich Baxa; Gwo-Yu Chuang; Ivelin S. Georgiev; Rui Kong; Sijy O’Dell; Syed Shahzad-ul-Hussan; Chen-Hsiang Shen; Yaroslav Tsybovsky; Robert T. Bailer; Syna K. Gift; Mark K. Louder; Krisha McKee; Reda Rawi; Catherine H. Stevenson; Guillaume Stewart-Jones; Justin D. Taft; Eric Waltari; Yongping Yang; Baoshan Zhang; Sachin S. Shivatare; Vidya S. Shivatare; Chang-Chun D. Lee; Chung-Yi Wu; Betty Benjamin; Robert W. Blakesley; Gerry Bouffard; Shelise Brooks; Holly Coleman

&NA; Virtually the entire surface of the HIV‐1‐envelope trimer is recognized by neutralizing antibodies, except for a highly glycosylated region at the center of the “silent face” on the gp120 subunit. From an HIV‐1‐infected donor, #74, we identified antibody VRC‐PG05, which neutralized 27% of HIV‐1 strains. The crystal structure of the antigen‐binding fragment of VRC‐PG05 in complex with gp120 revealed an epitope comprised primarily of N‐linked glycans from N262, N295, and N448 at the silent face center. Somatic hypermutation occurred preferentially at antibody residues that interacted with these glycans, suggesting somatic development of glycan recognition. Resistance to VRC‐PG05 in donor #74 involved shifting of glycan‐N448 to N446 or mutation of glycan‐proximal residue E293. HIV‐1 neutralization can thus be achieved at the silent face center by glycan‐recognizing antibody; along with other known epitopes, the VRC‐PG05 epitope completes coverage by neutralizing antibody of all major exposed regions of the prefusion closed trimer. Graphical Abstract Figure. No caption available. HighlightsIdentified and defined crystal structure of antibody VRC‐PG05 in complex with gp120VRC‐PG05 epitope is at the center of the glycosylated silent face of HIV‐1 gp120VRC‐PG05 utilizes both glycopeptide and glycan‐cluster mechanisms of recognitionVRC‐PG05 completes neutralizing antibody coverage of the prefusion‐closed Env trimer &NA; The center of the “silent face” on the HIV‐1 envelope is shielded by glycans and has been devoid of antibody recognition. Zhou et al. identify the antibody VRC‐PG05, which binds a glycan‐dominated epitope at the silent face center and completes antibody recognition of all major exposed regions of the envelope trimer.


Journal of the American Chemical Society | 2009

Unprecedented glycosidase activity at a lectin carbohydrate-binding site exemplified by the cyanobacterial lectin MVL.

Syed Shahzad-ul-Hussan; Mengli Cai; Carole A. Bewley

Carbohydrate binding proteins, or lectins, are engendered with the ability to bind specific carbohydrate structures, thereby mediating cell-cell and cell-pathogen interactions. Lectins are distinct from carbohydrate modifying enzymes and antibodies, respectively, as they do not carry out glycosidase or glycosyl transferase reactions, and they are of nonimmune origin. Cyanobacterial and algal lectins have become prominent in recent years due to their unique biophysical traits, such as exhibiting novel protein folds and unusually high carbohydrate affinity, and ability to potently inhibit HIV-1 entry through high affinity carbohydrate-mediated interactions with the HIV envelope glycoprotein gp120. The antiviral cyanobacterial lectin Microcystis viridis lectin (MVL), which contains two high affinity oligomannose binding sites, is one such example. Here we used glycan microarray profiling, NMR spectroscopy, and mutagenesis to show that one of the two oligomannose binding sites of MVL can catalyze the cleavage of chitin fragments (such as chitotriose) to GlcNAc, to determine the mode of MVL binding to and cleavage of chitotriose, to identify Asp75 as the primary catalytic residue involved in this cleavage, and to solve the solution structure of an inactive mutant of MVL in complex with this unexpected substrate. These studies represent the first demonstration of dual catalytic activity and carbohydrate recognition for discrete oligosaccharides at the same carbohydrate-binding site in a lectin. Sequence comparisons between the N- and C-domains of MVL, together with the sequences of new MVL homologues identified through bioinformatics, provide insight into the evolving roles of carbohydrate recognition.

Collaboration


Dive into the Syed Shahzad-ul-Hussan's collaboration.

Top Co-Authors

Avatar

Carole A. Bewley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Baoshan Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter D. Kwong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yongping Yang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gwo-Yu Chuang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ivelin S. Georgiev

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marie Pancera

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mark K. Louder

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sandra Loesgen

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge