Network


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

Hotspot


Dive into the research topics where Kavitha Baruah is active.

Publication


Featured researches published by Kavitha Baruah.


Journal of Clinical Investigation | 2013

Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity

Margaret E. Ackerman; Matthew Crispin; Xiaojie Yu; Kavitha Baruah; Austin W. Boesch; David J. Harvey; Anne Sophie Dugast; Erin L. Heizen; Altan Ercan; Ickwon Choi; Hendrik Streeck; Peter Nigrovic; Chris Bailey-Kellogg; Chris Scanlan; Galit Alter

While the induction of a neutralizing antibody response against HIV remains a daunting goal, data from both natural infection and vaccine-induced immune responses suggest that it may be possible to induce antibodies with enhanced Fc effector activity and improved antiviral control via vaccination. However, the specific features of naturally induced HIV-specific antibodies that allow for the potent recruitment of antiviral activity and the means by which these functions are regulated are poorly defined. Because antibody effector functions are critically dependent on antibody Fc domain glycosylation, we aimed to define the natural glycoforms associated with robust Fc-mediated antiviral activity. We demonstrate that spontaneous control of HIV and improved antiviral activity are associated with a dramatic shift in the global antibody-glycosylation profile toward agalactosylated glycoforms. HIV-specific antibodies exhibited an even greater frequency of agalactosylated, afucosylated, and asialylated glycans. These glycoforms were associated with enhanced Fc-mediated reduction of viral replication and enhanced Fc receptor binding and were consistent with transcriptional profiling of glycosyltransferases in peripheral B cells. These data suggest that B cell programs tune antibody glycosylation actively in an antigen-specific manner, potentially contributing to antiviral control during HIV infection.


Journal of the American Chemical Society | 2012

An endoglycosidase with alternative glycan specificity allows broadened glycoprotein remodelling.

Jonathan J. Goodfellow; Kavitha Baruah; Keisuke Yamamoto; Camille Bonomelli; Benjamin Krishna; David J. Harvey; Matthew Crispin; Christopher N. Scanlan; Benjamin G. Davis

Protein endoglycosidases are useful for biocatalytic alteration of glycans on protein surfaces, but the currently limited selectivity of endoglycosidases has prevented effective manipulation of certain N-linked glycans widely found in nature. Here we reveal that a bacterial endoglycosidase from Streptococcus pyogenes , EndoS, is complementary to other known endoglycosidases (EndoA, EndoH) used for current protein remodeling. It allows processing of complex-type N-linked glycans +/- core fucosylation but does not process oligomannose- or hybrid-type glycans. This biocatalytic activity now addresses previously refractory antibody glycoforms.


Journal of the American Chemical Society | 2013

Engineering hydrophobic protein-carbohydrate interactions to fine-tune monoclonal antibodies.

Xiaojie Yu; Kavitha Baruah; David J. Harvey; Snezana Vasiljevic; Dominic S. Alonzi; Byeong-Doo Song; Matthew K. Higgins; Thomas A. Bowden; Christopher N. Scanlan; Max Crispin

Biologically active conformations of the IgG1 Fc homodimer are maintained by multiple hydrophobic interactions between the protein surface and the N-glycan. The Fc glycan modulates biological effector functions, including antibody-dependent cellular cytotoxicity (ADCC) which is mediated in part through the activatory Fc receptor, FcγRIIIA. Consistent with previous reports, we found that site-directed mutations disrupting the protein–carbohydrate interface (F241A, F243A, V262E, and V264E) increased galactosylation and sialylation of the Fc and, concomitantly, reduced the affinity for FcγRIIIA. We rationalized this effect by crystallographic analysis of the IgG1 Fc F241A mutant, determined here to a resolution of 1.9 Å, which revealed localized destabilization of this glycan–protein interface. Given that sialylation of Fc glycans decreases ADCC, one explanation for the effect of these mutants on FcγRIIIA binding is their increased sialylation. However, a glycan-engineered IgG1 with hypergalactosylated and hypersialylated glycans exhibited unchanged binding affinity to FcγRIIIA. Moreover, when we expressed these mutants as a chemically uniform (Man5GlcNAc2) glycoform, the individual effect of each mutation on FcγRIIIA affinity was preserved. This effect was broadly recapitulated for other Fc receptors (FcγRI, FcγRIIA, FcγRIIB, and FcγRIIIB). These data indicate that destabilization of the glycan–protein interactions, rather than increased galactosylation and sialylation, modifies the Fc conformation(s) relevant for FcγR binding. Engineering of the protein–carbohydrate interface thus provides an independent parameter in the engineering of Fc effector functions and a route to the synthesis of new classes of Fc domain with novel combinations of affinities for activatory and inhibitory Fc receptors.


Journal of the American Chemical Society | 2012

Chemical and Structural Analysis of an Antibody Folding Intermediate Trapped during Glycan Biosynthesis

Thomas A. Bowden; Kavitha Baruah; Charlotte H. Coles; David J. Harvey; Xiaojie Yu; Byeong Doo Song; David I. Stuart; A. Radu Aricescu; Christopher N. Scanlan; E. Yvonne Jones; Matthew Crispin

Human IgG Fc glycosylation modulates immunological effector functions such as antibody-dependent cellular cytotoxicity and phagocytosis. Engineering of Fc glycans therefore enables fine-tuning of the therapeutic properties of monoclonal antibodies. The N-linked glycans of Fc are typically complex-type, forming a network of noncovalent interactions along the protein surface of the Cγ2 domain. Here, we manipulate the mammalian glycan-processing pathway to trap IgG1 Fc at sequential stages of maturation, from oligomannose- to hybrid- to complex-type glycans, and show that the Fc is structurally stabilized following the transition of glycans from their hybrid- to complex-type state. X-ray crystallographic analysis of this hybrid-type intermediate reveals that N-linked glycans undergo conformational changes upon maturation, including a flip within the trimannosyl core. Our crystal structure of this intermediate reveals a molecular basis for antibody biogenesis and provides a template for the structure-guided engineering of the protein–glycan interface of therapeutic antibodies.


Journal of Molecular Biology | 2012

Selective deactivation of serum IgG: a general strategy for the enhancement of monoclonal antibody receptor interactions

Kavitha Baruah; Thomas A. Bowden; Benjamin Krishna; Raymond A. Dwek; Matthew Crispin; Christopher N. Scanlan

Serum IgG is a potent inhibitor of monoclonal antibody (mAb) binding to the cell-surface Fcγ receptors (FcγRs), which mediate cytotoxic and phagocytic effector functions. Here, we show that this competition can be eliminated, selectively, by the introduction to serum of (i) an enzyme that displaces Fc from FcγRs and (ii) a modification present in the therapeutic mAb that renders it resistant to that enzyme. Specifically, we show that (i) EndoS (endoglycosidase S) cleaves only complex-type glycans of the type found on IgG but (ii) is inactive against an engineered IgG Fc with oligomannose-type glycans. EndoS thus reduces FcγR binding of serum IgG, but not that of engineered mAb. Introduction of both the engineered mAb and endoglycosidase in serum leads to a dramatic increase in FcγR binding compared to the introduction of mAb in serum alone. Antibody receptor refocusing is a general technique for boosting the effector signal of therapeutic antibodies.


Electrophoresis | 2013

Travelling wave ion mobility and negative ion fragmentation for the structural determination of N-linked glycans.

David J. Harvey; Charlotte A. Scarff; Matthew Edgeworth; Max Crispin; Christopher N. Scanlan; Frank Sobott; Sarah Allman; Kavitha Baruah; Laura K. Pritchard; James H. Scrivens

Travelling wave ion mobility was investigated for its ability to separate N‐glycans from other compounds and for resolution of isomers. Charged glycans, exemplified by sialylated complex N‐glycans released from bovine fetuin and ionised by electrospray, could be separated from residual glycopeptides allowing the minor, more highly sialylated compounds to be detected where their ions were obscured by ions from other compounds in different charge states. This technique was also found to be excellent for extracting the N‐glycan profiles from contaminated samples. Structural identification of the glycans was performed by negative ion CID fragmentation, a method that provides a wealth of structurally diagnostic ions. However, fragment ions can also appear in the glycan profiles where they can be mistaken for glycan molecular ions. Fragments and molecular ions were frequently shown to have different drift time profiles, allowing them to be differentiated. Some separation of isomers was found but only for the smallest compounds. Differentiation from conformers was achieved by plotting drift time profiles of the fragments; these profiles matched those of the precursor ions where conformers were present. The techniques were applied to investigations of N‐glycans released from the fungus Piptoporus betulinus where the technique was used to separate different carbohydrate types present in biological extracts.


Journal of Immunological Methods | 2015

A method for high-throughput, sensitive analysis of IgG Fc and Fab glycosylation by capillary electrophoresis

Alison E. Mahan; Jacquelynne Tedesco; Kendall Dionne; Kavitha Baruah; Hao D. Cheng; Philip L. De Jager; Dan H. Barouch; Todd J. Suscovich; Margaret E. Ackerman; Max Crispin; Galit Alter

The N-glycan of the IgG constant region (Fc) plays a central role in tuning and directing multiple antibody functions in vivo, including antibody-dependent cellular cytotoxicity, complement deposition, and the regulation of inflammation, among others. However, traditional methods of N-glycan analysis, including HPLC and mass spectrometry, are technically challenging and ill suited to handle the large numbers of low concentration samples analyzed in clinical or animal studies of the N-glycosylation of polyclonal IgG. Here we describe a capillary electrophoresis-based technique to analyze plasma-derived polyclonal IgG-glycosylation quickly and accurately in a cost-effective, sensitive manner that is well suited for high-throughput analyses. Additionally, because a significant fraction of polyclonal IgG is glycosylated on both Fc and Fab domains, we developed an approach to separate and analyze domain-specific glycosylation in polyclonal human, rhesus and mouse IgGs. Overall, this protocol allows for the rapid, accurate, and sensitive analysis of Fc-specific IgG glycosylation, which is critical for population-level studies of how antibody glycosylation may vary in response to vaccination or infection, and across disease states ranging from autoimmunity to cancer in both clinical and animal studies.


Journal of Biological Chemistry | 2014

Fragments of Bacterial Endoglycosidase S and Immunoglobulin G Reveal Subdomains of Each That Contribute to Deglycosylation

Ev Dixon; Jolyon K. Claridge; Dj Harvey; Kavitha Baruah; Xiaojie Yu; S Vesiljevic; S Mattick; Laura K. Pritchard; Benjamin Krishna; Christopher N. Scanlan; Schnell; Matthew K. Higgins; Nicole Zitzmann; Max Crispin

Background: Endoglycosidase S (EndoS) is an immune evasion factor from Streptococcus pyogenes that impedes IgG effector functions by deglycosylation. Results: Analysis of fragments of enzyme and substrate identify components of each that contribute to catalysis. Conclusion: Regions outside of the catalytic domain of EndoS contribute to IgG deglycosylation, and catalysis does not require all antibody subunits. Significance: Engineering of EndoS specificity requires consideration of both catalytic and non-catalytic regions. Endoglycosidase S (EndoS) is a glycoside-hydrolase secreted by the bacterium Streptococcus pyogenes. EndoS preferentially hydrolyzes the N-linked glycans from the Fc region of IgG during infection. This hydrolysis impedes Fc functionality and contributes to the immune evasion strategy of S. pyogenes. Here, we investigate the mechanism of human serum IgG deactivation by EndoS. We expressed fragments of IgG1 and demonstrated that EndoS was catalytically active against all of them including the isolated CH2 domain of the Fc domain. Similarly, we sought to investigate which domains within EndoS could contribute to activity. Bioinformatics analysis of the domain organization of EndoS confirmed the previous predictions of a chitinase domain and leucine-rich repeat but also revealed a putative carbohydrate binding module (CBM) followed by a C-terminal region. Using expressed fragments of EndoS, circular dichroism of the isolated CBM, and a CBM-C-terminal region fusion revealed folded domains dominated by β sheet and α helical structure, respectively. Nuclear magnetic resonance analysis of the CBM with monosaccharides was suggestive of carbohydrate binding functionality. Functional analysis of truncations of EndoS revealed that, whereas the C-terminal of EndoS is dispensable for activity, its deletion impedes the hydrolysis of IgG glycans.


Protein Science | 2015

Eliminating antibody polyreactivity through addition of N-linked glycosylation

Gwo-Yu Chuang; Baoshan Zhang; Krisha McKee; Sijy O'Dell; Young Do Kwon; Tongqing Zhou; Julie Blinn; Krissey E. Lloyd; Robert Parks; Tarra Von Holle; Sung-Youl Ko; Wing-Pui Kong; Amarendra Pegu; Kavitha Baruah; Max Crispin; John R. Mascola; M. Anthony Moody; Barton F. Haynes; Ivelin S. Georgiev; Peter D. Kwong

Antibody polyreactivity can be an obstacle to translating a candidate antibody into a clinical product. Standard tests such as antibody binding to cardiolipin, HEp‐2 cells, or nuclear antigens provide measures of polyreactivity, but its causes and the means to resolve are often unclear. Here we present a method for eliminating antibody polyreactivity through the computational design and genetic addition of N‐linked glycosylation near known sites of polyreactivity. We used the HIV‐1‐neutralizing antibody, VRC07, as a test case, since efforts to increase VRC07 potency at three spatially distinct sites resulted in enhanced polyreactivity. The addition of N‐linked glycans proximal to the polyreactivity‐enhancing mutations at each of the spatially distinct sites resulted in reduced antibody polyreactivity as measured by (i) anti‐cardiolipin ELISA, (ii) Luminex AtheNA Multi‐Lyte ANA binding, and (iii) HEp‐2 cell staining. The reduced polyreactivity trended with increased antibody concentration over time in mice, but not with improved overall protein stability as measured by differential scanning calorimetry. Moreover, glycan proximity to the site of polyreactivity appeared to be a critical factor. The results provide evidence that antibody polyreactivity can result from local, rather than global, features of an antibody and that addition of N‐linked glycosylation can be an effective approach to reducing antibody polyreactivity.


Antibody Fc#R##N#Linking Adaptive and Innate Immunity | 2014

Chapter 10 – Antibody Glycosylation

Xiaojie Yu; Kavitha Baruah; Christopher N. Scanlan; Max Crispin

The antibody Fc region is posttranslationally modified by N-linked glycosylation. In immunoglobulin G (IgG), the processing of the glycans is restricted by the presence of extensive interaction with the protein surface. The resulting set of antibody glycoforms exhibit a range of effector functions. In this chapter, we outline the impact of glycosylation on the immune function of antibodies and discuss the implications for monoclonal antibody and intravenous immunoglobulin therapies.

Collaboration


Dive into the Kavitha Baruah's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Max Crispin

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

Xiaojie Yu

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas A. Bowden

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge