1
|
Gschwandtner M, Derler R, Talker E, Trojacher C, Gubensäk N, Becker W, Gerlza T, Klaus Z, Stocki P, Walsh FS, Rutkowski JL, Kungl A. A Single-Domain VNAR Nanobody Binds with High-Affinity and Selectivity to the Heparin Pentasaccharide Fondaparinux. Int J Mol Sci 2025; 26:4045. [PMID: 40362285 PMCID: PMC12071740 DOI: 10.3390/ijms26094045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Glycosaminoglycans (GAGs) are key ligands for proteins involved in physiological and pathological processes. Specific GAG-binding patterns are rarely identified, with the heparin pentasaccharide as an Antithrombin-III ligand being the best characterized. Generating glycan-specific antibodies is difficult due to their size, pattern dispersion, and flexibility. Single-domain variable new antigen receptors (VNAR nanobodies) from nurse sharks are highly soluble, stable, and versatile. Their unique properties suggest advantages over conventional antibodies, particularly for challenging biotherapeutic targets. Here we have used VNAR semi-synthetic phage libraries to select high-affinity fondaparinux-binding VNARs that did not show cross-reactivity with other GAG species. Competition ELISA and surface plasmon resonance identified a single fondaparinux-selective VNAR clone. This VNAR exhibited an extraordinarily stable protein fold: the beta-strands are stabilized by a robust hydrophobic network, as revealed by heteronuclear NMR. Docking fondaparinux to the VNAR structure revealed a large contact surface area between the CDR3 loop of the antibody and the glycan. Fusing the VNAR with a human Fc domain resulted in a stable product with a high affinity for fondaparinux (Kd = 9.3 × 10-8 M) that could efficiently discriminate between fondaparinux and other glycosaminoglycans. This novel glycan-targeting screening technology represents a promising therapeutic strategy for addressing GAG-related diseases.
Collapse
Affiliation(s)
- Martha Gschwandtner
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
| | - Rupert Derler
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
| | - Elisa Talker
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
| | - Christina Trojacher
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
| | - Nina Gubensäk
- Institute of Chemistry, Karl-Franzens-University Graz, Heinrichstraße 28, A-8010 Graz, Austria; (N.G.); (W.B.); (Z.K.)
| | - Walter Becker
- Institute of Chemistry, Karl-Franzens-University Graz, Heinrichstraße 28, A-8010 Graz, Austria; (N.G.); (W.B.); (Z.K.)
| | - Tanja Gerlza
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
| | - Zangger Klaus
- Institute of Chemistry, Karl-Franzens-University Graz, Heinrichstraße 28, A-8010 Graz, Austria; (N.G.); (W.B.); (Z.K.)
| | - Pawel Stocki
- Ossianix, Inc., Stevenage Bioscience Catalyst, Gunnels Wood Rd, Stevenage, Herts SG1 2FX, UK; (P.S.); (F.S.W.); (J.L.R.)
| | - Frank S. Walsh
- Ossianix, Inc., Stevenage Bioscience Catalyst, Gunnels Wood Rd, Stevenage, Herts SG1 2FX, UK; (P.S.); (F.S.W.); (J.L.R.)
| | - Julia Lynn Rutkowski
- Ossianix, Inc., Stevenage Bioscience Catalyst, Gunnels Wood Rd, Stevenage, Herts SG1 2FX, UK; (P.S.); (F.S.W.); (J.L.R.)
| | - Andreas Kungl
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, A-8010 Graz, Austria (E.T.); (C.T.); (T.G.)
- Antagonis Biotherapeutics GmbH, Strasserhofweg 77a, A-8045 Graz, Austria
| |
Collapse
|
2
|
Novak J, King RG, Yother J, Renfrow MB, Green TJ. O-glycosylation of IgA1 and the pathogenesis of an autoimmune disease IgA nephropathy. Glycobiology 2024; 34:cwae060. [PMID: 39095059 PMCID: PMC11442006 DOI: 10.1093/glycob/cwae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/21/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024] Open
Abstract
IgA nephropathy is a kidney disease characterized by deposition of immune complexes containing abnormally O-glycosylated IgA1 in the glomeruli. Specifically, some O-glycans are missing galactose that is normally β1,3-linked to N-acetylgalactosamine of the core 1 glycans. These galactose-deficient IgA1 glycoforms are produced by IgA1-secreting cells due to a dysregulated expression and activity of several glycosyltransferases. Galactose-deficient IgA1 in the circulation of patients with IgA nephropathy is bound by IgG autoantibodies and the resultant immune complexes can contain additional proteins, such as complement C3. These complexes, if not removed from the circulation, can enter the glomerular mesangium, activate the resident mesangial cells, and induce glomerular injury. In this review, we briefly summarize clinical and pathological features of IgA nephropathy, review normal and aberrant IgA1 O-glycosylation pathways, and discuss the origins and potential significance of natural anti-glycan antibodies, namely those recognizing N-acetylgalactosamine. We also discuss the features of autoantibodies specific for galactose-deficient IgA1 and the characteristics of pathogenic immune complexes containing IgA1 and IgG. In IgA nephropathy, kidneys are injured by IgA1-containing immune complexes as innocent bystanders. Most patients with IgA nephropathy progress to kidney failure and require dialysis or transplantation. Moreover, most patients after transplantation experience a recurrent disease. Thus, a better understanding of the pathogenetic mechanisms is needed to develop new disease-specific treatments.
Collapse
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, United States
| | - R Glenn King
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, United States
| | - Janet Yother
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, United States
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 720 20th Street South, Birmingham, AL 35294, United States
| | - Todd J Green
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, United States
| |
Collapse
|
3
|
Thomas M, Nguyen TH, Drnevich J, D’Souza AM, de Alarcon PA, Gnanamony M. Hu14.18K.322A Causes Direct Cell Cytotoxicity and Synergizes with Induction Chemotherapy in High-Risk Neuroblastoma. Cancers (Basel) 2024; 16:2064. [PMID: 38893185 PMCID: PMC11171330 DOI: 10.3390/cancers16112064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
The disialoganglioside, GD2, is a promising therapeutic target due to its overexpression in certain tumors, particularly neuroblastoma (NB), with limited expression in normal tissues. Despite progress, the intricate mechanisms of action and the full spectrum of the direct cellular responses to anti-GD2 antibodies remain incompletely understood. In this study, we examined the direct cytotoxic effects of the humanized anti-GD2 antibody hu14.18K322A (hu14) on NB cell lines, by exploring the associated cell-death pathways. Additionally, we assessed the synergy between hu14 and conventional induction chemotherapy drugs. Our results revealed that hu14 treatment induced direct cytotoxic effects in CHLA15 and SK-N-BE1 cell lines, with a pronounced impact on proliferation and colony formation. Apoptosis emerged as the predominant cell-death pathway triggered by hu14. Furthermore, we saw a reduction in GD2 surface expression in response to hu14 treatment. Hu14 demonstrated synergy with induction chemotherapy drugs with alterations in GD2 expression. Our comprehensive investigation provides valuable insights into the multifaceted effects of hu14 on NB cells, shedding light on its direct cytotoxicity, cell-death pathways, and interactions with induction chemotherapy drugs. This study contributes to the evolving understanding of anti-GD2 antibody therapy and its potential synergies with conventional treatments in the context of NB.
Collapse
Affiliation(s)
- Maria Thomas
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Thu Hien Nguyen
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, The University of Illinois at Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL 61801, USA;
| | - Amber M. D’Souza
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Pedro A. de Alarcon
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Manu Gnanamony
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| |
Collapse
|
4
|
Moeller T, Shah SB, Lai K, Lopez-Barbosa N, Desai P, Wang W, Zhong Z, Redmond D, Singh A, DeLisa MP. Profiling Germinal Center-like B Cell Responses to Conjugate Vaccines Using Synthetic Immune Organoids. ACS CENTRAL SCIENCE 2023; 9:787-804. [PMID: 37122450 PMCID: PMC10141597 DOI: 10.1021/acscentsci.2c01473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Indexed: 05/03/2023]
Abstract
Glycoengineered bacteria have emerged as a cost-effective platform for rapid and controllable biosynthesis of designer conjugate vaccines. However, little is known about the engagement of such conjugates with naïve B cells to induce the formation of germinal centers (GC), a subanatomical microenvironment that converts naïve B cells into antibody-secreting plasma cells. Using a three-dimensional biomaterials-based B-cell follicular organoid system, we demonstrate that conjugates triggered robust expression of hallmark GC markers, B cell receptor clustering, intracellular signaling, and somatic hypermutation. These responses depended on the relative immunogenicity of the conjugate and correlated with the humoral response in vivo. The occurrence of these mechanisms was exploited for the discovery of high-affinity antibodies against components of the conjugate on a time scale that was significantly shorter than for typical animal immunization-based workflows. Collectively, these findings highlight the potential of synthetic organoids for rapidly predicting conjugate vaccine efficacy as well as expediting antigen-specific antibody discovery.
Collapse
Affiliation(s)
- Tyler
D. Moeller
- Robert
F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Shivem B. Shah
- Nancy
E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kristine Lai
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Natalia Lopez-Barbosa
- Robert
F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Primit Desai
- Biochemistry,
Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, United States
| | - Weiyao Wang
- Robert
F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Zhe Zhong
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - David Redmond
- Institute
for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, New York 10021, United States
- Department
of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, New York 10021, United States
| | - Ankur Singh
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Matthew P. DeLisa
- Robert
F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Nancy
E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
- Biochemistry,
Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, United States
- Cornell
Institute of Biotechnology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
5
|
Zhang Y, Li Q, Luo L, Duan C, Shen J, Wang Z. Application of germline antibody features to vaccine development, antibody discovery, antibody optimization and disease diagnosis. Biotechnol Adv 2023; 65:108143. [PMID: 37023966 DOI: 10.1016/j.biotechadv.2023.108143] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Although the efficacy and commercial success of vaccines and therapeutic antibodies have been tremendous, designing and discovering new drug candidates remains a labor-, time- and cost-intensive endeavor with high risks. The main challenges of vaccine development are inducing a strong immune response in broad populations and providing effective prevention against a group of highly variable pathogens. Meanwhile, antibody discovery faces several great obstacles, especially the blindness in antibody screening and the unpredictability of the developability and druggability of antibody drugs. These challenges are largely due to poorly understanding of germline antibodies and the antibody responses to pathogen invasions. Thanks to the recent developments in high-throughput sequencing and structural biology, we have gained insight into the germline immunoglobulin (Ig) genes and germline antibodies and then the germline antibody features associated with antigens and disease manifestation. In this review, we firstly outline the broad associations between germline antibodies and antigens. Moreover, we comprehensively review the recent applications of antigen-specific germline antibody features, physicochemical properties-associated germline antibody features, and disease manifestation-associated germline antibody features on vaccine development, antibody discovery, antibody optimization, and disease diagnosis. Lastly, we discuss the bottlenecks and perspectives of current and potential applications of germline antibody features in the biotechnology field.
Collapse
Affiliation(s)
- Yingjie Zhang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Changfei Duan
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China.
| |
Collapse
|
6
|
Minibody-Based and scFv-Based Antibody Fragment-Drug Conjugates Selectively Eliminate GD2-Positive Tumor Cells. Int J Mol Sci 2023; 24:ijms24021239. [PMID: 36674755 PMCID: PMC9860947 DOI: 10.3390/ijms24021239] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Ganglioside GD2 is a well-established target expressed on multiple solid tumors, many of which are characterized by low treatment efficiency. Antibody-drug conjugates (ADCs) have demonstrated marked success in a number of solid tumors, and GD2-directed drug conjugates may also hold strong therapeutic potential. In a recent study, we showed that ADCs based on the approved antibody dinutuximab and the drugs monomethyl auristatin E (MMAE) or F (MMAF) manifested potent and selective cytotoxicity in a panel of tumor cell lines and strongly inhibited solid tumor growth in GD2-positive mouse cancer models. Here, we employed two different GD2-binding moieties-minibodies and scFv fragments that carry variable antibody domains identical to those of dinutuximab, and site-directly conjugated them to MMAE or MMAF by thiol-maleimide chemistry with drug-to-antibody ratios (DAR) of 2 and 1, respectively. Specific binding of the antibody fragment-drug conjugates (FDCs) to GD2 was confirmed in direct ELISA, flow cytometry, and confocal microscopy. Selective cytotoxic and cytostatic effects of the conjugates were observed in GD2-positive but not GD2-negative neuroblastoma and melanoma cell lines. Minibody-based FDCs demonstrated more pronounced cytotoxic effects and stronger antigen binding compared to scFv-based FDCs. The developed molecules may offer considerable practical benefit, since antibody fragment-drug conjugates are capable of enhancing therapeutic efficacy of ADCs by improving their pharmacokinetic characteristics and reducing side effects.
Collapse
|
7
|
Pashov A, Murali R, Makhoul I, Karbassi B, Kieber-Emmons T. Harnessing Antibody Polyspecificity for Cancer Immunotherapy. Monoclon Antib Immunodiagn Immunother 2022; 41:290-300. [PMID: 36306515 DOI: 10.1089/mab.2022.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Targeting the diverse glycan repertoire expressed on tumor cells is considered a viable therapeutic strategy to deal with tumor cell heterogeneity. Inherently polyspecific, natural, glycan-reactive antibodies are purported to be protective in thwarting infections and in cancer immunotherapy. Tumor-associated carbohydrate antigens (TACAs) are related to pathogen glycans, to which nascent or natural antibodies exist and IgM responses are elicited. To capture the polyspecific nature of anticarbohydrate responses, we have focused on the rational design of carbohydrate mimetic peptides (CMPs) cross-reactive with TACA reactive antibodies. In particular, we have focused on the development of CMPs that display reactivity to GD2 and Lewis Y (LeY) reactive monoclonal antibodies. They would serve as templates for pan-immunogens inducing biosimilar polyreactive antibodies. In the design, we relied on structural analyses of CMP's enhanced binding to the templates using molecular modeling. Glycan reactivity patterns of affinity CMP-purified human antibodies further refined specificity profiles in comparison with the immune response to the CMP in clinical trials. In this study, we further define the molecular characteristics for this mimicry by considering the polyspecificity of LeY and GD2 reactive antibodies binding to the lacto-ceramide core Galβ(1,4)Glcβ(1-1')Cer. Binding to this minimum building block can be capitalized on for cancer therapy and diagnostics and illustrates a new approach in designing cancer vaccines taking advantage of the latent polyspecificity of antibodies and the relevance of natural antibodies in antigen discovery and design.
Collapse
Affiliation(s)
- Anastas Pashov
- Department of Immunology, Stephan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ramachandran Murali
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Issam Makhoul
- Department of Medicine and Pathology, Winthrop P. Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Behjatolah Karbassi
- Department of Medicine and Pathology, Winthrop P. Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Thomas Kieber-Emmons
- Department of Medicine and Pathology, Winthrop P. Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
8
|
DeLaitsch AT, Pridgen JR, Tytla A, Peach ML, Hu R, Farnsworth DW, McMillan AK, Flanagan N, Temme JS, Nicklaus MC, Gildersleeve JC. Selective Recognition of Carbohydrate Antigens by Germline Antibodies Isolated from AID Knockout Mice. J Am Chem Soc 2022; 144:4925-4941. [PMID: 35282679 PMCID: PMC10506689 DOI: 10.1021/jacs.1c12745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Germline antibodies, the initial set of antibodies produced by the immune system, are critical for host defense, and information about their binding properties can be useful for designing vaccines, understanding the origins of autoantibodies, and developing monoclonal antibodies. Numerous studies have found that germline antibodies are polyreactive with malleable, flexible binding pockets. While insightful, it remains unclear how broadly this model applies, as there are many families of antibodies that have not yet been studied. In addition, the methods used to obtain germline antibodies typically rely on assumptions and do not work well for many antibodies. Herein, we present a distinct approach for isolating germline antibodies that involves immunizing activation-induced cytidine deaminase (AID) knockout mice. This strategy amplifies antigen-specific B cells, but somatic hypermutation does not occur because AID is absent. Using synthetic haptens, glycoproteins, and whole cells, we obtained germline antibodies to an assortment of clinically important tumor-associated carbohydrate antigens, including Lewis Y, the Tn antigen, sialyl Lewis C, and Lewis X (CD15/SSEA-1). Through glycan microarray profiling and cell binding, we demonstrate that all but one of these germline antibodies had high selectivity for their glycan targets. Using molecular dynamics simulations, we provide insights into the structural basis of glycan recognition. The results have important implications for designing carbohydrate-based vaccines, developing anti-glycan monoclonal antibodies, and understanding antibody evolution within the immune system.
Collapse
Affiliation(s)
- Andrew T DeLaitsch
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Jacey R Pridgen
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Avery Tytla
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Megan L Peach
- Basic Science Program, Chemical Biology Laboratory, Leidos Biomedical Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Rayleen Hu
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - David W Farnsworth
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Aislinn K McMillan
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Natalie Flanagan
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - J Sebastian Temme
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Marc C Nicklaus
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Jeffrey C Gildersleeve
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
9
|
Ishida E, Corrigan DT, Malonis RJ, Hofmann D, Chen T, Amin AG, Chatterjee D, Joe M, Lowary TL, Lai JR, Achkar JM. Monoclonal antibodies from humans with Mycobacterium tuberculosis exposure or latent infection recognize distinct arabinomannan epitopes. Commun Biol 2021; 4:1181. [PMID: 34642445 PMCID: PMC8511196 DOI: 10.1038/s42003-021-02714-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022] Open
Abstract
The surface polysacharide arabinomannan (AM) and related glycolipid lipoarabinomannan (LAM) play critical roles in tuberculosis pathogenesis. Human antibody responses to AM/LAM are heterogenous and knowledge of reactivity to specific glycan epitopes at the monoclonal level is limited, especially in individuals who can control M. tuberculosis infection. We generated human IgG mAbs to AM/LAM from B cells of two asymptomatic individuals exposed to or latently infected with M. tuberculosis. Here, we show that two of these mAbs have high affinity to AM/LAM, are non-competing, and recognize different glycan epitopes distinct from other anti-AM/LAM mAbs reported. Both mAbs recognize virulent M. tuberculosis and nontuberculous mycobacteria with marked differences, can be used for the detection of urinary LAM, and can detect M. tuberculosis and LAM in infected lungs. These mAbs enhance our understanding of the spectrum of antibodies to AM/LAM epitopes in humans and are valuable for tuberculosis diagnostic and research applications. Elise Ishida et al. generate human monoclonal antibodies that can selectively recognize specific oligosaccharide epitopes of the polysaccharides arabinomannan and lipoarabinomannan, which are critical for M. tuberculosis pathogenesis. The authors demonstrate the utility of these antibodies in both diagnostic and laboratory settings, making them important tools for M. tuberculosis research.
Collapse
Affiliation(s)
- Elise Ishida
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Devin T Corrigan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ryan J Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel Hofmann
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anita G Amin
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Delphi Chatterjee
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maju Joe
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.,Institute of Biological Chemistry, Academia Sinica, Nangang, Taipei, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jacqueline M Achkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
Exploring the Impact of Ketodeoxynonulosonic Acid in Host-Pathogen Interactions Using Uptake and Surface Display by Nontypeable Haemophilus influenzae. mBio 2021; 12:mBio.03226-20. [PMID: 33468699 PMCID: PMC7845648 DOI: 10.1128/mbio.03226-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
All cells in vertebrates are coated with a dense array of glycans often capped with sugars called sialic acids. Sialic acids have many functions, including serving as a signal for recognition of “self” cells by the immune system, thereby guiding an appropriate immune response against foreign “nonself” and/or damaged cells. Surface expression of the common vertebrate sialic acid (Sia) N-acetylneuraminic acid (Neu5Ac) by commensal and pathogenic microbes appears structurally to represent “molecular mimicry” of host sialoglycans, facilitating multiple mechanisms of host immune evasion. In contrast, ketodeoxynonulosonic acid (Kdn) is a more ancestral Sia also present in prokaryotic glycoconjugates that are structurally quite distinct from vertebrate sialoglycans. We detected human antibodies against Kdn-terminated glycans, and sialoglycan microarray studies found these anti-Kdn antibodies to be directed against Kdn-sialoglycans structurally similar to those on human cell surface Neu5Ac-sialoglycans. Anti-Kdn-glycan antibodies appear during infancy in a pattern similar to those generated following incorporation of the nonhuman Sia N-glycolylneuraminic acid (Neu5Gc) onto the surface of nontypeable Haemophilus influenzae (NTHi), a human commensal and opportunistic pathogen. NTHi grown in the presence of free Kdn took up and incorporated the Sia into its lipooligosaccharide (LOS). Surface display of the Kdn within NTHi LOS blunted several virulence attributes of the pathogen, including Neu5Ac-mediated resistance to complement and whole blood killing, complement C3 deposition, IgM binding, and engagement of Siglec-9. Upper airway administration of Kdn reduced NTHi infection in human-like Cmah null (Neu5Gc-deficient) mice that express a Neu5Ac-rich sialome. We propose a mechanism for the induction of anti-Kdn antibodies in humans, suggesting that Kdn could be a natural and/or therapeutic “Trojan horse” that impairs colonization and virulence phenotypes of free Neu5Ac-assimilating human pathogens.
Collapse
|
11
|
V Kholodenko I, V Kalinovsky D, V Svirshchevskaya E, I Doronin I, V Konovalova M, V Kibardin A, V Shamanskaya T, S Larin S, M Deyev S, V Kholodenko R. Multimerization through Pegylation Improves Pharmacokinetic Properties of scFv Fragments of GD2-Specific Antibodies. Molecules 2019; 24:molecules24213835. [PMID: 31653037 PMCID: PMC6864547 DOI: 10.3390/molecules24213835] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Antigen-binding fragments of antibodies specific to the tumor-associated ganglioside GD2 are well poised to play a substantial role in modern GD2-targeted cancer therapies, however, rapid elimination from the body and reduced affinity compared to full-length antibodies limit their therapeutic potential. In this study, scFv fragments of GD2-specific antibodies 14.18 were produced in a mammalian expression system that specifically bind to ganglioside GD2, followed by site-directed pegylation to generate mono-, di-, and tetra-scFv fragments. Fractionated pegylated dimers and tetramers of scFv fragments showed significant increase of the binding to GD2 which was not accompanied by cross-reactivity with other gangliosides. Pegylated multimeric di-scFvs and tetra-scFvs exhibited cytotoxic effects in GD2-positive tumor cells, while their circulation time in blood significantly increased compared with monomeric antibody fragments. We also demonstrated a more efficient tumor uptake of the multimers in a syngeneic GD2-positive mouse cancer model. The findings of this study provide the rationale for improving therapeutic characteristics of GD2-specific antibody fragments by multimerization and propose a strategy to generate such molecules. On the basis of multimeric antibody fragments, bispecific antibodies and conjugates with cytotoxic drugs or radioactive isotopes may be developed that will possess improved pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Irina V Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow 119121, Russia.
| | - Daniel V Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Elena V Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Igor I Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia.
| | - Maria V Konovalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Alexey V Kibardin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Tatyana V Shamanskaya
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Sergey S Larin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia.
| | - Roman V Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia.
| |
Collapse
|
12
|
Feng YL, Chen H, Chen DQ, Vaziri ND, Su W, Ma SX, Shang YQ, Mao JR, Yu XY, Zhang L, Guo Y, Zhao YY. Activated NF-κB/Nrf2 and Wnt/β-catenin pathways are associated with lipid metabolism in CKD patients with microalbuminuria and macroalbuminuria. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2317-2332. [PMID: 31102786 DOI: 10.1016/j.bbadis.2019.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/17/2023]
Abstract
Early diagnosis of CKD patients at risk for microalbuminuria or macroalbuminuria could facilitate clinical outcomes and long-term survival. Considering the few and limited efficacy of current biomarkers in early detection, we aim to discover plasma lipids that effectively predict the development of CKD paitents with microalbuminuria or macroalbuminuria. A total of 380 healthy controls and 1156 patients with CKD stages 3 to 5 were stratified by urine albumin-creatinine ratio as microalbuminuria (30-300 mg/g) and macroalbuminuria (>300 mg/g). Fasting plasma samples were determined by UPLC-HDMS based on lipidomics. Quantitative real-time polymerase chain reaction, Western blot and immunohistochemical analyses were used to validate the lipid metabolism-associated pathways. Pathway analysis demonstrated that these lipids were closely associated with PPARγ, inflammatory mediator regulation of TRP channels and RAS signaling, which were intimately involved in activated NF-κB and Nrf2 pathways. We further carried out pathway validation and demonstrated that NF-κB pathway was activated in patients with macroalbuminuria compared with CKD patients with microalbuminuria, while Nrf2-associated protein expression was downregulated, which was accompanied by the up-regulation of Wnt/β-catenin signaling pathway. Four lipids including DTA, 5,8-TDA, GGD3 and DHA that showed great potential in the discrimination of CKD patients with microalbuminuria and healthy controls were selected by logistic regression analysis. Additionally, six lipid species including CDCA, glucosylceramide, GGD2, TTA, DHA and EDA that contributed to the discrimination of CKD patients with microalbuminuria and macroalbuminuria were selected by logistic LASSO regression Gangliosides were first identified and might be promising therapeutic targets for CKD patients with the different degree of albuminuria. Collectively, this study first demonstrates the association of plasma inflammation, oxidative stress, Wnt/β-catenin and lipid metabolism in CKD patients with microalbuminuria and macroalbuminuria.
Collapse
Affiliation(s)
- Ya-Long Feng
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Hua Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Dan-Qian Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92897, USA
| | - Wei Su
- Department of Nephrology, Baoji Central Hospital, No. 8 Jiangtan Road, Baoji, Shaanxi 721008, China
| | - Shi-Xing Ma
- Department of Nephrology, Baoji Central Hospital, No. 8 Jiangtan Road, Baoji, Shaanxi 721008, China
| | - You-Quan Shang
- Department of Nephrology, Baoji Central Hospital, No. 8 Jiangtan Road, Baoji, Shaanxi 721008, China
| | - Jia-Rong Mao
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, No. 2 Xihuamen, Xi'an, Shaanxi 710003, China
| | - Xiao-Yong Yu
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, No. 2 Xihuamen, Xi'an, Shaanxi 710003, China
| | - Li Zhang
- Department of Nephrology, Xi'an No. 4 Hospital, No. 21 Jiefang Road, Xi'an 710004, China
| | - Yan Guo
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China; Department of Internal Medicine, University of New Mexico, Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Ying-Yong Zhao
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
13
|
Schepens B, De Vlieger D, Saelens X. Vaccine options for influenza: thinking small. Curr Opin Immunol 2018; 53:22-29. [PMID: 29631195 DOI: 10.1016/j.coi.2018.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 11/16/2022]
|
14
|
Jeliazkov JR, Sljoka A, Kuroda D, Tsuchimura N, Katoh N, Tsumoto K, Gray JJ. Repertoire Analysis of Antibody CDR-H3 Loops Suggests Affinity Maturation Does Not Typically Result in Rigidification. Front Immunol 2018; 9:413. [PMID: 29545810 PMCID: PMC5840193 DOI: 10.3389/fimmu.2018.00413] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/14/2018] [Indexed: 12/18/2022] Open
Abstract
Antibodies can rapidly evolve in specific response to antigens. Affinity maturation drives this evolution through cycles of mutation and selection leading to enhanced antibody specificity and affinity. Elucidating the biophysical mechanisms that underlie affinity maturation is fundamental to understanding B-cell immunity. An emergent hypothesis is that affinity maturation reduces the conformational flexibility of the antibody's antigen-binding paratope to minimize entropic losses incurred upon binding. In recent years, computational and experimental approaches have tested this hypothesis on a small number of antibodies, often observing a decrease in the flexibility of the complementarity determining region (CDR) loops that typically comprise the paratope and in particular the CDR-H3 loop, which contributes a plurality of antigen contacts. However, there were a few exceptions and previous studies were limited to a small handful of cases. Here, we determined the structural flexibility of the CDR-H3 loop for thousands of recent homology models of the human peripheral blood cell antibody repertoire using rigidity theory. We found no clear delineation in the flexibility of naïve and antigen-experienced antibodies. To account for possible sources of error, we additionally analyzed hundreds of human and mouse antibodies in the Protein Data Bank through both rigidity theory and B-factor analysis. By both metrics, we observed only a slight decrease in the CDR-H3 loop flexibility when comparing affinity matured antibodies to naïve antibodies, and the decrease was not as drastic as previously reported. Further analysis, incorporating molecular dynamics simulations, revealed a spectrum of changes in flexibility. Our results suggest that rigidification may be just one of many biophysical mechanisms for increasing affinity.
Collapse
Affiliation(s)
- Jeliazko R Jeliazkov
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, United States
| | - Adnan Sljoka
- Department of Informatics, School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.,Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Nobuyuki Tsuchimura
- Department of Informatics, School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Naoki Katoh
- Department of Informatics, School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.,Laboratory of Medical Proteomics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jeffrey J Gray
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, United States.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
15
|
Mishra AK, Mariuzza RA. Insights into the Structural Basis of Antibody Affinity Maturation from Next-Generation Sequencing. Front Immunol 2018; 9:117. [PMID: 29449843 PMCID: PMC5799246 DOI: 10.3389/fimmu.2018.00117] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/15/2018] [Indexed: 12/26/2022] Open
Abstract
Affinity maturation is the process whereby the immune system generates antibodies of higher affinities during a response to antigen. It is unique in being the only evolutionary mechanism known to operate on a molecule in an organism's own body. Deciphering the structural mechanisms through which somatic mutations in antibody genes increase affinity is critical to understanding the evolution of immune repertoires. Next-generation sequencing (NGS) has allowed the reconstruction of antibody clonal lineages in response to viral pathogens, such as HIV-1, which was not possible in earlier studies of affinity maturation. Crystal structures of antibodies from these lineages bound to their target antigens have revealed, at the atomic level, how antibodies evolve to penetrate the glycan shield of envelope glycoproteins, and how viruses in turn evolve to escape neutralization. Collectively, structural studies of affinity maturation have shown that increased antibody affinity can arise from any one or any combination of multiple diverse mechanisms, including improved shape complementarity at the interface with antigen, increased buried surface area upon complex formation, additional interfacial polar or hydrophobic interactions, and preorganization or rigidification of the antigen-binding site.
Collapse
Affiliation(s)
- Arjun K Mishra
- W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Rockville, MD, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, United States
| | - Roy A Mariuzza
- W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Rockville, MD, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, United States
| |
Collapse
|