Bio


Dr. Taia Wang is an Assistant Professor of Medicine and of Microbiology and Immunology with research aimed at defining mechanisms in human immunity and disease. The Wang laboratory uses in vivo and in vitro systems to study how antibodies can modulate viral infections or anti-viral vaccine responses through signaling interactions with Fc gamma receptors (FcγRs). The overarching goal of projects in the Wang lab is to elucidate FcγR pathways that can be harnessed towards the development of enhanced vaccines and therapeutics.

Dr. Wang obtained M.D. and Ph.D. training at Mount Sinai School of Medicine. Her Ph.D. training focused on mechanisms in immunity against influenza viruses (P. Palese). Following this training, she completed postdoctoral research at the Rockefeller University where she studied human IgG and FcγR biology in the Laboratory of Molecular Genetics and Immunology (J. Ravetch). During this time she also undertook formal training in clinical research, earning a Masters of Science in Clinical Investigation at The Rockefeller University.

The Wang laboratory's work on viral immunity and disease pathogenesis has been recognized with several awards including the Searle Scholar’s Award and the Burroughs Wellcome Fund Investigator Award for Investigators in the Pathogenesis of Infectious Disease. Dr. Wang is also the recipient of a Bravo Family Junior Faculty Endowment.

SARS-CoV-2, dengue viruses, influenza viruses, disease pathogenesis, influenza virus vaccines

Academic Appointments


Administrative Appointments


  • Associate Director, Stanford Medical Scientist Training Program (2020 - Present)

Honors & Awards


  • Investigator in the Pathogenesis of Infectious Disease Award, Burroughs Wellcome Fund (2022)
  • Searle Scholars Award, The Searle Scholars Program (2018)
  • Investigator Award, Chan Zuckerberg Biohub (2017)
  • Young Physician-Scientist Award, The American Society for Clinical Investigation (2017)
  • Leona M. and Harry B. Helmsley Scholar, Helmsley Charitable Trust (2015)
  • Niarchos Scholar, Stavros Niarchos Foundation (2014)
  • Iris and Junming Le Scholar, The Iris and Junming Le Foundation (2013)

Boards, Advisory Committees, Professional Organizations


  • Member and Abstract Programming Chair, The American Association of Immunologists (2021 - Present)
  • Member, Infectious Diseases Society of America (2019 - Present)
  • Member, Henry Kunkel Society (2018 - Present)
  • Member, American Society for Microbiology (2016 - Present)
  • Associate Scientific Advisor, Science Translational Medicine (2015 - 2016)

Professional Education


  • Postdoctoral training, Rockefeller University, Fc receptor biology and human immunology (2016)
  • MSCI, Rockefeller University, Masters of Science in Clinical Investigation (2015)
  • MD, Mount Sinai School of Medicine, Medicine (2012)
  • PhD, Mount Sinai School of Medicine, Virology (2010)

Current Research and Scholarly Interests


Laboratory of Mechanisms in Human Immunity and Disease Pathogenesis

Antibodies are a critical component of host defense. While the importance of humoral immunity has been recognized for decades, substantial gaps in knowledge remain around how antibodies function, and how their function is regulated, in vivo. Our laboratory performs studies designed to fill in these gaps, with the goal of enabling new vaccine and therapeutic strategies to prevent human disease. My interest in this area culminated from training in medicine, RNA virus biology (PhD), and molecular antibody biology (postdoctoral training). The intersection of these topics, viral immunity and disease pathogenesis, is the focus of our work. The essential question driving our research is why a small subset of people develop severe or fatal disease during viral infection while most infections result in a subclinical or mild outcome, even in at-risk populations. Our hypothesis is that the antibody signaling pathways that are engaged during viral infection through Fc gamma receptors (FcγRs) are a key driver of these distinct outcomes. We are focused on several major unknowns to address this hypothesis: How are antibody effector functions regulated in vivo and does this change in disease? How do distinct signaling pathways engaged by IgG immune complex-FcγR interactions impact host cell genetic regulation and the ultimate inflammatory/immune response? What are the tissue-specific functions that antibodies engage? How does the heterogeneity in post-translational modifications (PTMs) of human antibodies contribute to heterogeneity in viral immunity?



Current clinical studies:
Recruiting:

An Open Label Study of IgG Fc Glycan Composition in Human Immunity
Principal Investigator: Taia T. Wang, MD, PhD
ClinicalTrials.gov Identifier:
NCT01967238

Projects


  • Regulation of the IgG Fc domain repertoire - NIH NIAID R01, Stanford University

    Major goal: To define how IgG Fc glycoforms are regulated in vivo.

    Location

    Stanford, CA

  • Immune determinants of dengue disease severity - Searle Scholars Award, Stanford University

    Major goal: To define the role of FcγRIIIa in antibody-dependent enhancement of dengue infections.

    Location

    Stanford, CA

  • Defining the role of IgG Fc domains and their receptors in antiviral immunity - Chan Zuckerberg Initiative

    Major goal: To define Fc-Fc receptor interactions that are essential for immunity against influenza and dengue viruses.

    Location

    Stanford, CA

  • Enhancing IgG transfer to prevent perinatal infections - Bill & Melinda Gates Foundation, Rockefeller University

    Major goal: This work will define mechanisms for enhancing the durability of passive immunization of neonates

    Location

    Stanford, CA

    Collaborators

    • Jeffrey Ravetch, Professor, Rockefeller University
  • Impact of Initial Influenza Exposure on Immunity in Infants - NIH NIAID U19, St. Jude Children’s Research Hospital

    Impact of Initial Influenza Exposure on Immunity in Infants

    Location

    Stanford, CA

    Collaborators

    • Paul Thomas, Professor, St. Jude Children’s Research Hospital
  • Center for Influenza Vaccine Immunology and Development - NIH NIAID U19, Icahn School of Medicine at Mount Sinai

    Major Goals: To define the role of antibodies in broad protection against influenza viruses

    Location

    Stanford, CA

    Collaborators

    • Florian Krammer, Professor, Icahn School of Medicine at Mount Sinai
    • Rafi Ahmed, Professor, Emory University
  • Rockefeller University Cooperative Centers of Human Immunology - NIH NIAID U19

    Major Goals: PROJECT 3: To determine whether specific forms of IgG antibodies predispose to severe dengue disease during infection.

    Location

    Stanford, CA

    Collaborators

    • Jeffrey Ravetch, Professor, Rockefeller University
    • Charles Rice, Professor, Rockefeller University
    • Michel Nussenzweig, Professor, Rockefeller University
  • Stanford Cooperative Centers of Human Immunology - NIH NIAID U19, Stanford University

    Major Goals: Defining adaptive and innate immunity, memory and repertoire in vaccination and infection

    Location

    Stanford, CA

  • Mechanisms and Duration of Immunity to SARS-CoV-2 - NIH NIAID U19, Stanford University

    Location

    Stanford, CA

    Collaborators

    • Scott Boyd, Professor, Stanford University
    • Mark Davis, Director, Stanford Institute for Immunity, Transplantation and Infection and the Burt and Marion Avery Family Professor, Stanford University
  • Antibody responses in symptomatic and asymptomatic SARS-CoV-2 infections - NIH NIAID, Rockefeller University

    Supplement

    Location

    Stanford, CA

  • Immunity against COVID-19 - Fast Grants, Stanford University

    Location

    Stanford, CA

  • SARS-CoV-2 vaccine designed to enhance immunogenicity of the receptor binding domain - Bill and Melinda Gates Foundation, Stanford University

    Location

    Stanford, CA

  • Enhancing anti-CSP antibodies for in vivo potency against malaria infection - Bill & Melinda Gates Foundation

    Location

    Stanford, USA

  • Exploring new therapeutic concepts to mitigate emergent respiratory viral pandemics - HHMI Emerging Pathogens Initiative

    Location

    Stanford, USA

  • Biology of afucosylated antibodies in infectious disease - Burroughs Wellcome Fund PATH

    Location

    Stanford, USA

2023-24 Courses


Stanford Advisees


All Publications


  • Multi-omics analysis of mucosal and systemic immunity to SARS-CoV-2 after birth. Cell Wimmers, F., Burrell, A. R., Feng, Y., Zheng, H., Arunachalam, P. S., Hu, M., Spranger, S., Nyhoff, L. E., Joshi, D., Trisal, M., Awasthi, M., Bellusci, L., Ashraf, U., Kowli, S., Konvinse, K. C., Yang, E., Blanco, M., Pellegrini, K., Tharp, G., Hagan, T., Chinthrajah, R. S., Nguyen, T. T., Grifoni, A., Sette, A., Nadeau, K. C., Haslam, D. B., Bosinger, S. E., Wrammert, J., Maecker, H. T., Utz, P. J., Wang, T. T., Khurana, S., Khatri, P., Staat, M. A., Pulendran, B. 2023

    Abstract

    The dynamics of immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infants and young children by analyzing blood samples and weekly nasal swabs collected before, during, and after infection with Omicron and non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, showed no sign of decay for up to 300 days. Infants mounted a robust mucosal immune response characterized by inflammatory cytokines, interferon (IFN) α, and T helper (Th) 17 and neutrophil markers (interleukin [IL]-17, IL-8, and CXCL1). The immune response in blood was characterized by upregulation of activation markers on innate cells, no inflammatory cytokines, but several chemokines and IFNα. The latter correlated with viral load and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell multi-omics. Together, these data provide a snapshot of immunity to infection during the initial weeks and months of life.

    View details for DOI 10.1016/j.cell.2023.08.044

    View details for PubMedID 37776858

  • Spheromers reveal robust T cell responses to the Pfizer/BioNTech vaccine and attenuated peripheral CD8+ T cell responses post SARS-CoV-2 infection. Immunity Gao, F., Mallajoysula, V., Arunachalam, P. S., van der Ploeg, K., Manohar, M., Röltgen, K., Yang, F., Wirz, O., Hoh, R., Haraguchi, E., Lee, J. Y., Willis, R., Ramachandiran, V., Li, J., Kathuria, K. R., Li, C., Lee, A. S., Shah, M. M., Sindher, S. B., Gonzalez, J., Altman, J. D., Wang, T. T., Boyd, S. D., Pulendran, B., Jagannathan, P., Nadeau, K. C., Davis, M. M. 2023

    Abstract

    T cells are a critical component of the response to SARS-CoV-2, but their kinetics after infection and vaccination are insufficiently understood. Using "spheromer" peptide-MHC multimer reagents, we analyzed healthy subjects receiving two doses of the Pfizer/BioNTech BNT162b2 vaccine. Vaccination resulted in robust spike-specific T cell responses for the dominant CD4+ (HLA-DRB1∗15:01/S191) and CD8+ (HLA-A∗02/S691) T cell epitopes. Antigen-specific CD4+ and CD8+ T cell responses were asynchronous, with the peak CD4+ T cell responses occurring 1 week post the second vaccination (boost), whereas CD8+ T cells peaked 2 weeks later. These peripheral T cell responses were elevated compared with COVID-19 patients. We also found that previous SARS-CoV-2 infection resulted in decreased CD8+ T cell activation and expansion, suggesting that previous infection can influence the T cell response to vaccination.

    View details for DOI 10.1016/j.immuni.2023.03.005

    View details for PubMedID 36996809

    View details for PubMedCentralID PMC10017386

  • Autoantibodies are highly prevalent in non-SARS-CoV-2 respiratory infections and critical illness. JCI insight Feng, A., Yang, E. Y., Moore, A. R., Dhingra, S., Chang, S. E., Yin, X., Pi, R., Mack, E. K., Völkel, S., Geßner, R., Gündisch, M., Neubauer, A., Renz, H., Tsiodras, S., Fragkou, P. C., Asuni, A. A., Levitt, J. E., Wilson, J. G., Leong, M., Lumb, J. H., Mao, R., Pinedo, K., Roque, J., Richards, C. M., Stabile, M., Swaminathan, G., Salagianni, M. L., Triantafyllia, V., Bertrams, W., Blish, C. A., Carette, J. E., Frankovich, J., Meffre, E., Nadeau, K. C., Singh, U., Wang, T. T., Luning Prak, E. T., Herold, S., Andreakos, E., Schmeck, B., Skevaki, C., Rogers, A. J., Utz, P. J. 2023; 8 (3)

    Abstract

    The widespread presence of autoantibodies in acute infection with SARS-CoV-2 is increasingly recognized, but the prevalence of autoantibodies in non-SARS-CoV-2 infections and critical illness has not yet been reported. We profiled IgG autoantibodies in 267 patients from 5 independent cohorts with non-SARS-CoV-2 viral, bacterial, and noninfectious critical illness. Serum samples were screened using Luminex arrays that included 58 cytokines and 55 autoantigens, many of which are associated with connective tissue diseases (CTDs). Samples positive for anti-cytokine antibodies were tested for receptor blocking activity using cell-based functional assays. Anti-cytokine antibodies were identified in > 50% of patients across all 5 acutely ill cohorts. In critically ill patients, anti-cytokine antibodies were far more common in infected versus uninfected patients. In cell-based functional assays, 11 of 39 samples positive for select anti-cytokine antibodies displayed receptor blocking activity against surface receptors for Type I IFN, GM-CSF, and IL-6. Autoantibodies against CTD-associated autoantigens were also commonly observed, including newly detected antibodies that emerged in longitudinal samples. These findings demonstrate that anti-cytokine and autoantibodies are common across different viral and nonviral infections and range in severity of illness.

    View details for DOI 10.1172/jci.insight.163150

    View details for PubMedID 36752204

  • Early immune markers of clinical, virological, and immunological outcomes in patients with COVID-19: a multi-omics study. eLife Hu, Z., van der Ploeg, K., Chakraborty, S., Arunachalam, P. S., Mori, D. A., Jacobson, K. B., Bonilla, H., Parsonnet, J., Andrews, J. R., Holubar, M., Subramanian, A., Khosla, C., Maldonado, Y., Hedlin, H., de la Parte, L., Press, K., Ty, M., Tan, G. S., Blish, C., Takahashi, S., Rodriguez-Barraquer, I., Greenhouse, B., Butte, A. J., Singh, U., Pulendran, B., Wang, T. T., Jagannathan, P. 2022; 11

    Abstract

    The great majority of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections are mild and uncomplicated, but some individuals with initially mild COVID-19 progressively develop more severe symptoms. Furthermore, there is substantial heterogeneity in SARS-CoV-2-specific memory immune responses following infection. There remains a critical need to identify host immune biomarkers predictive of clinical and immunological outcomes in SARS-CoV-2-infected patients.Leveraging longitudinal samples and data from a clinical trial (N=108) in SARS-CoV-2-infected outpatients, we used host proteomics and transcriptomics to characterize the trajectory of the immune response in COVID-19 patients. We characterized the association between early immune markers and subsequent disease progression, control of viral shedding, and SARS-CoV-2-specific T cell and antibody responses measured up to 7 months after enrollment. We further compared associations between early immune markers and subsequent T cell and antibody responses following natural infection with those following mRNA vaccination. We developed machine-learning models to predict patient outcomes and validated the predictive model using data from 54 individuals enrolled in an independent clinical trial.We identify early immune signatures, including plasma RIG-I levels, early IFN signaling, and related cytokines (CXCL10, MCP1, MCP-2, and MCP-3) associated with subsequent disease progression, control of viral shedding, and the SARS-CoV-2-specific T cell and antibody response measured up to 7 months after enrollment. We found that several biomarkers for immunological outcomes are shared between individuals receiving BNT162b2 (Pfizer-BioNTech) vaccine and COVID-19 patients. Finally, we demonstrate that machine-learning models using 2-7 plasma protein markers measured early within the course of infection are able to accurately predict disease progression, T cell memory, and the antibody response post-infection in a second, independent dataset.Early immune signatures following infection can accurately predict clinical and immunological outcomes in outpatients with COVID-19 using validated machine-learning models.Support for the study was provided from National Institute of Health/National Institute of Allergy and Infectious Diseases (NIH/NIAID) (U01 AI150741-01S1 and T32-AI052073), the Stanford's Innovative Medicines Accelerator, National Institutes of Health/National Institute on Drug Abuse (NIH/NIDA) DP1DA046089, and anonymous donors to Stanford University. Peginterferon lambda provided by Eiger BioPharmaceuticals.

    View details for DOI 10.7554/eLife.77943

    View details for PubMedID 36239699

  • Durable protection against the SARS-CoV-2 Omicron variant is induced by an adjuvanted subunit vaccine. Science translational medicine Arunachalam, P. S., Feng, Y., Ashraf, U., Hu, M., Walls, A. C., Edara, V. V., Zarnitsyna, V. I., Aye, P. P., Golden, N., Miranda, M. C., Green, K. W., Threeton, B. M., Maness, N. J., Beddingfield, B. J., Bohm, R. P., Scheuermann, S. E., Goff, K., Dufour, J., Russell-Lodrigue, K., Kepl, E., Fiala, B., Wrenn, S., Ravichandran, R., Ellis, D., Carter, L., Rogers, K., Shirreff, L. M., Ferrell, D. E., Deb Adhikary, N. R., Fontenot, J., Hammond, H. L., Frieman, M., Grifoni, A., Sette, A., O'Hagan, D. T., Van Der Most, R., Rappuoli, R., Villinger, F., Kleanthous, H., Rappaport, J., Suthar, M. S., Veesler, D., Wang, T. T., King, N. P., Pulendran, B. 2022; 14 (658): eabq4130

    Abstract

    Despite the remarkable efficacy of COVID-19 vaccines, waning immunity and the emergence of SARS-CoV-2 variants such as Omicron represents a global health challenge. Here, we present data from a study in nonhuman primates demonstrating durable protection against the Omicron BA.1 variant induced by a subunit SARS-CoV-2 vaccine comprising the receptor binding domain of the ancestral strain (RBD-Wu) on the I53-50 nanoparticle adjuvanted with AS03, which was recently authorized for use in individuals 18 years or older. Vaccination induced neutralizing antibody (nAb) titers that were maintained at high concentrations for at least 1 year after two doses, with a pseudovirus nAb geometric mean titer (GMT) of 1978 and a live virus nAb GMT of 1331 against the ancestral strain but not against the Omicron BA.1 variant. However, a booster dose at 6 to 12 months with RBD-Wu or RBD-beta (RBD from the Beta variant) displayed on I53-50 elicited high neutralizing titers against the ancestral and Omicron variants. In addition, we observed persistent neutralization titers against a panel of sarbecoviruses, including SARS-CoV. Furthermore, there were substantial and persistent memory T and B cell responses reactive to Beta and Omicron variants. Vaccination resulted in protection against Omicron infection in the lung and suppression of viral burden in the nares at 6 weeks after the final booster immunization. Even at 6 months after vaccination, we observed protection in the lung and rapid control of virus in the nares. These results highlight the durable and cross-protective immunity elicited by the AS03-adjuvanted RBD-I53-50 nanoparticle vaccine.

    View details for DOI 10.1126/scitranslmed.abq4130

    View details for PubMedID 35976993

  • Anti-nucleocapsid antibody levels and pulmonary comorbid conditions are linked to post-COVID-19 syndrome. JCI insight Jia, X., Cao, S., Lee, A. S., Manohar, M., Sindher, S. B., Ahuja, N., Artandi, M., Blish, C. A., Blomkalns, A. L., Chang, I., Collins, W. J., Desai, M., Din, H. N., Do, E., Fernandes, A., Geng, L. N., Rosenberg-Hasson, Y., Mahoney, M. R., Glascock, A. L., Chan, L. Y., Fong, S. Y., Phelps, M., Raeber, O., Purington, N., Röltgen, K., Rogers, A. J., Snow, T., Wang, T. T., Solis, D., Vaughan, L., Verghese, M., Maecker, H., Wittman, R., Puri, R., Kistler, A., Yang, S., Boyd, S. D., Pinsky, B. A., Chinthrajah, S., Nadeau, K. C. 2022; 7 (13)

    Abstract

    BACKGROUNDProlonged symptoms after SARS-CoV-2 infection are well documented. However, which factors influence development of long-term symptoms, how symptoms vary across ethnic groups, and whether long-term symptoms correlate with biomarkers are points that remain elusive.METHODSAdult SARS-CoV-2 reverse transcription PCR-positive (RT-PCR-positive) patients were recruited at Stanford from March 2020 to February 2021. Study participants were seen for in-person visits at diagnosis and every 1-3 months for up to 1 year after diagnosis; they completed symptom surveys and underwent blood draws and nasal swab collections at each visit.RESULTSOur cohort (n = 617) ranged from asymptomatic to critical COVID-19 infections. In total, 40% of participants reported at least 1 symptom associated with COVID-19 six months after diagnosis. Median time from diagnosis to first resolution of all symptoms was 44 days; median time from diagnosis to sustained symptom resolution with no recurring symptoms for 1 month or longer was 214 days. Anti-nucleocapsid IgG level in the first week after positive RT-PCR test and history of lung disease were associated with time to sustained symptom resolution. COVID-19 disease severity, ethnicity, age, sex, and remdesivir use did not affect time to sustained symptom resolution.CONCLUSIONWe found that all disease severities had a similar risk of developing post-COVID-19 syndrome in an ethnically diverse population. Comorbid lung disease and lower levels of initial IgG response to SARS-CoV-2 nucleocapsid antigen were associated with longer symptom duration.TRIAL REGISTRATIONClinicalTrials.gov, NCT04373148.FUNDINGNIH UL1TR003142 CTSA grant, NIH U54CA260517 grant, NIEHS R21 ES03304901, Sean N Parker Center for Allergy and Asthma Research at Stanford University, Chan Zuckerberg Biohub, Chan Zuckerberg Initiative, Sunshine Foundation, Crown Foundation, and Parker Foundation.

    View details for DOI 10.1172/jci.insight.156713

    View details for PubMedID 35801588

  • Harnessing IgG Fc glycosylation for clinical benefit. Current opinion in immunology Archer, E. J., Gonzalez, J. C., Ghosh, D., Mellins, E. D., Wang, T. T. 2022; 77: 102231

    Abstract

    The effector activity of IgG antibodies is regulated at several levels, including IgG subclass, modifications of the Fc glycan, and the distribution of Type I and II Fcgamma receptors (FcgammaR) on effector cells. Here, we explore how Fc glycosylation, particularly sialylation and fucosylation, tunes cellular responses to immune complexes. We review the current understanding of the pathways and mechanisms underlying this biology, address FcgammaR in antigen presentation, and discuss aspects of the clinical understanding of Fc glycans in therapies and disease.

    View details for DOI 10.1016/j.coi.2022.102231

    View details for PubMedID 35797920

  • Heterogeneity in IgG-CD16 signaling in infectious disease outcomes IMMUNOLOGICAL REVIEWS Gonzalez, J. C., Chakraborty, S., Thulin, N. K., Wang, T. T. 2022

    Abstract

    In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).

    View details for DOI 10.1111/imr.13109

    View details for Web of Science ID 000820199400001

    View details for PubMedID 35781671

  • TNF-alpha+ CD4+ Tcells dominate the SARS-CoV-2 specific T cell response in COVID-19 outpatients and are associated with durable antibodies. Cell reports. Medicine van der Ploeg, K., Kirosingh, A. S., Mori, D. A., Chakraborty, S., Hu, Z., Sievers, B. L., Jacobson, K. B., Bonilla, H., Parsonnet, J., Andrews, J. R., Press, K. D., Ty, M. C., Ruiz-Betancourt, D. R., de la Parte, L., Tan, G. S., Blish, C. A., Takahashi, S., Rodriguez-Barraquer, I., Greenhouse, B., Singh, U., Wang, T. T., Jagannathan, P. 2022: 100640

    Abstract

    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific CD4+ Tcells are likely important in immunity against coronavirus 2019 (COVID-19), but our understanding of CD4+ longitudinal dynamics following infection and of specific features that correlate with the maintenance of neutralizing antibodies remains limited. Here, we characterize SARS-CoV-2-specific CD4+ Tcells in a longitudinal cohort of 109 COVID-19 outpatients enrolled during acute infection. The quality of the SARS-CoV-2-specific CD4+ response shifts from cells producing interferon gamma (IFNgamma) to tumor necrosis factor alpha (TNF-alpha) from 5days to 4months post-enrollment, with IFNgamma-IL-21-TNF-alpha+ CD4+ Tcells the predominant population detected at later time points. Greater percentages of IFNgamma-IL-21-TNF-alpha+ CD4+ Tcells on day 28 correlate with SARS-CoV-2-neutralizing antibodies measured 7months post-infection (⍴= 0.4, p= 0.01). mRNA vaccination following SARS-CoV-2 infection boosts both IFNgamma- and TNF-alpha-producing, spike-protein-specific CD4+ Tcells. These data suggest that SARS-CoV-2-specific, TNF-alpha-producing CD4+ Tcells may play an important role in antibody maintenance following COVID-19.

    View details for DOI 10.1016/j.xcrm.2022.100640

    View details for PubMedID 35588734

  • Differential Peripheral Blood Glycoprotein Profiles in Symptomatic and Asymptomatic COVID-19. Viruses Pickering, C., Zhou, B., Xu, G., Rice, R., Ramachandran, P., Huang, H., Pham, T. D., Schapiro, J. M., Cong, X., Chakraborty, S., Edwards, K., Reddy, S. T., Guirgis, F., Wang, T. T., Serie, D., Lindpaintner, K. 2022; 14 (3)

    Abstract

    Glycosylation is the most common form of post-translational modification of proteins, critically affecting their structure and function. Using liquid chromatography and mass spectrometry for high-resolution site-specific quantification of glycopeptides coupled with high-throughput artificial intelligence-powered data processing, we analyzed differential protein glycoisoform distributions of 597 abundant serum glycopeptides and nonglycosylated peptides in 50 individuals who had been seriously ill with COVID-19 and in 22 individuals who had recovered after an asymptomatic course of COVID-19. As additional comparison reference phenotypes, we included 12 individuals with a history of infection with a common cold coronavirus, 16 patients with bacterial sepsis, and 15 healthy subjects without history of coronavirus exposure. We found statistically significant differences, at FDR < 0.05, for normalized abundances of 374 of the 597 peptides and glycopeptides interrogated between symptomatic and asymptomatic COVID-19 patients. Similar statistically significant differences were seen when comparing symptomatic COVID-19 patients to healthy controls (350 differentially abundant peptides and glycopeptides) and common cold coronavirus seropositive subjects (353 differentially abundant peptides and glycopeptides). Among healthy controls and sepsis patients, 326 peptides and glycopeptides were found to be differentially abundant, of which 277 overlapped with biomarkers that showed differential expression between symptomatic COVID-19 cases and healthy controls. Among symptomatic COVID-19 cases and sepsis patients, 101 glycopeptide and peptide biomarkers were found to be statistically significantly abundant. Using both supervised and unsupervised machine learning techniques, we found specific glycoprotein profiles to be strongly predictive of symptomatic COVID-19 infection. LASSO-regularized multivariable logistic regression and K-means clustering yielded accuracies of 100% in an independent test set and of 96% overall, respectively. Our findings are consistent with the interpretation that a majority of glycoprotein modifications observed which are shared among symptomatic COVID-19 and sepsis patients likely represent a generic consequence of a severe systemic immune and inflammatory state. However, there are glycoisoform changes that are specific and particular to severe COVID-19 infection. These may be representative of either COVID-19-specific consequences or susceptibility to or predisposition for a severe course of the disease. Our findings support the potential value of glycoproteomic biomarkers in the biomedical understanding and, potentially, the clinical management of serious acute infectious conditions.

    View details for DOI 10.3390/v14030553

    View details for PubMedID 35336960

  • Early non-neutralizing, afucosylated antibody responses are associated with COVID-19 severity. Science translational medicine Chakraborty, S., Gonzalez, J. C., Sievers, B. L., Mallajosyula, V., Chakraborty, S., Dubey, M., Ashraf, U., Cheng, B. Y., Kathale, N., Tran, K. Q., Scallan, C., Sinnott, A., Cassidy, A., Chen, S. T., Gelbart, T., Gao, F., Golan, Y., Ji, X., Kim-Schulze, S., Prahl, M., Gaw, S. L., Gnjatic, S., Marron, T. U., Merad, M., Arunachalam, P. S., Boyd, S. D., Davis, M. M., Holubar, M., Khosla, C., Maecker, H. T., Maldonado, Y., Mellins, E. D., Nadeau, K. C., Pulendran, B., Singh, U., Subramanian, A., Utz, P. J., Sherwood, R., Zhang, S., Jagannathan, P., Tan, G. S., Wang, T. T. 1800: eabm7853

    Abstract

    A damaging inflammatory response is implicated in the pathogenesis of severe coronavirus disease 2019 (COVID-19), but mechanisms contributing to this response are unclear. In two prospective cohorts, early non-neutralizing, afucosylated IgG antibodies specific to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were associated with progression from mild to more severe COVID-19. In contrast to the antibody structures that were associated with disease progression, antibodies that were elicited by mRNA SARS-CoV-2 vaccines were instead highly fucosylated and enriched in sialylation, both modifications that reduce the inflammatory potential of IgG. To study the biology afucosylated IgG immune complexes, we developed an in vivo model that revealed that human IgG-Fc gamma receptor (FcgammaR) interactions could regulate inflammation in the lung. Afucosylated IgG immune complexes isolated from COVID-19 patients induced inflammatory cytokine production and robust infiltration of the lung by immune cells. By contrast, vaccine-elicited IgG did not promote an inflammatory lung response. Together, these results show that IgG-FcgammaR interactions are able to regulate inflammation in the lung and may define distinct lung activities associated with the IgG that are associated with severe COVID-19 and protection against infection with SARS-CoV-2.

    View details for DOI 10.1126/scitranslmed.abm7853

    View details for PubMedID 35040666

  • Antibodies elicited by SARS-CoV-2 infection or mRNA vaccines have reduced neutralizing activity against Beta and Omicron pseudoviruses. Science translational medicine Sievers, B. L., Chakraborty, S., Xue, Y., Gelbart, T., Gonzalez, J. C., Cassidy, A. G., Golan, Y., Prahl, M., Gaw, S. L., Arunachalam, P. S., Blish, C. A., Boyd, S. D., Davis, M. M., Jagannathan, P., Nadeau, K. C., Pulendran, B., Singh, U., Scheuermann, R. H., Frieman, M. B., Vashee, S., Wang, T. T., Tan, G. S. 1800: eabn7842

    Abstract

    Multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that possess mutations associated with increased transmission and antibody escape have arisen over the course of the current pandemic. Although the current vaccines have largely been effective against past variants, the number of mutations found on the Omicron (B.1.1.529) spike protein appear to diminish the protection conferred by pre-existing immunity. Using vesicular stomatitis virus (VSV) pseudoparticles expressing the spike protein of several SARS-CoV-2 variants, we evaluated the magnitude and breadth of the neutralizing antibody response over time in individuals after infection and in mRNA-vaccinated individuals. We observed that boosting increases the magnitude of the antibody response to wildtype (D614), Beta, Delta, and Omicron variants; however, the Omicron variant was the most resistant to neutralization. We further observed that vaccinated healthy adults had robust and broad antibody responses whereas responses may have been reduced in vaccinated pregnant women, underscoring the importance of learning how to maximize mRNA vaccine responses in pregnant populations. Findings from this study show substantial heterogeneity in the magnitude and breadth of responses after infection and mRNA vaccination and may support the addition of more conserved viral antigens to existing SARS-CoV-2 vaccines.

    View details for DOI 10.1126/scitranslmed.abn7842

    View details for PubMedID 35025672

  • Current and novel biomarkers of thrombotic risk in COVID-19: a Consensus Statement from the International COVID-19 Thrombosis Biomarkers Colloquium. Nature reviews. Cardiology Gorog, D. A., Storey, R. F., Gurbel, P. A., Tantry, U. S., Berger, J. S., Chan, M. Y., Duerschmied, D., Smyth, S. S., Parker, W. A., Ajjan, R. A., Vilahur, G., Badimon, L., Berg, J. M., Cate, H. T., Peyvandi, F., Wang, T. T., Becker, R. C. 1800

    Abstract

    Coronavirus disease 2019 (COVID-19) predisposes patients to thrombotic and thromboembolic events, owing to excessive inflammation, endothelial cell activation and injury, platelet activation and hypercoagulability. Patients with COVID-19 have a prothrombotic or thrombophilic state, with elevations in the levels of several biomarkers of thrombosis, which are associated with disease severity and prognosis. Although some biomarkers of COVID-19-associated coagulopathy, including high levels of fibrinogen and D-dimer, were recognized early during the pandemic, many new biomarkers of thrombotic risk in COVID-19 have emerged. In this Consensus Statement, we delineate the thrombotic signature of COVID-19 and present the latest biomarkers and platforms to assess the risk of thrombosis in these patients, including markers of platelet activation, platelet aggregation, endothelial cell activation or injury, coagulation and fibrinolysis as well as biomarkers of the newly recognized post-vaccine thrombosis with thrombocytopenia syndrome. We then make consensus recommendations for the clinical use of these biomarkers to inform prognosis, assess disease acuity, and predict thrombotic risk and in-hospital mortality. A thorough understanding of these biomarkers might aid risk stratification and prognostication, guide interventions and provide a platform for future research.

    View details for DOI 10.1038/s41569-021-00665-7

    View details for PubMedID 35027697

  • CELLULAR CORRELATES FOR PROTECTION AGAINST MALARIA ACQUIRED ACROSS MULTIPLE PREGNANCIES Kirosingh, A., De La Parte, L., Ty, M., Kakuru, A., Muhindo, M. K., Thulin, N., Kamya, M., Feeney, M., Dorsey, G., Wang, T., Jagannathan, P. AMER SOC TROP MED & HYGIENE. 2021: 219
  • New-onset IgG autoantibodies in hospitalized patients with COVID-19. Nature communications Chang, S. E., Feng, A., Meng, W., Apostolidis, S. A., Mack, E., Artandi, M., Barman, L., Bennett, K., Chakraborty, S., Chang, I., Cheung, P., Chinthrajah, S., Dhingra, S., Do, E., Finck, A., Gaano, A., GeSSner, R., Giannini, H. M., Gonzalez, J., Greib, S., Gundisch, M., Hsu, A. R., Kuo, A., Manohar, M., Mao, R., Neeli, I., Neubauer, A., Oniyide, O., Powell, A. E., Puri, R., Renz, H., Schapiro, J., Weidenbacher, P. A., Wittman, R., Ahuja, N., Chung, H., Jagannathan, P., James, J. A., Kim, P. S., Meyer, N. J., Nadeau, K. C., Radic, M., Robinson, W. H., Singh, U., Wang, T. T., Wherry, E. J., Skevaki, C., Luning Prak, E. T., Utz, P. J. 2021; 12 (1): 5417

    Abstract

    COVID-19 is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. Here we develop three protein arrays to measure IgG autoantibodies associated with connective tissue diseases, anti-cytokine antibodies, and anti-viral antibody responses in serum from 147 hospitalized COVID-19 patients. Autoantibodies are identified in approximately 50% of patients but in less than 15% of healthy controls. When present, autoantibodies largely target autoantigens associated with rare disorders such as myositis, systemic sclerosis and overlap syndromes. A subset of autoantibodies targeting traditional autoantigens or cytokines develop de novo following SARS-CoV-2 infection. Autoantibodies track with longitudinal development of IgG antibodies recognizing SARS-CoV-2 structural proteins and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.

    View details for DOI 10.1038/s41467-021-25509-3

    View details for PubMedID 34521836

  • New-Onset IgG Autoantibodies in Hospitalized Patients with COVID-19 Chang, S., Feng, A., Meng, W., Apostolidis, S., Mack, E., Artandi, M., Barman, L., Bennett, K., Chakraborty, S., Chang, I., Cheung, P., Chinthrajah, S., Dhingra, S., Do, E., Finck, A., Gaano, A., Gessner, R., Giannini, H., Gonzalez, J., Greib, S., Gundisch, M., Hsu, A., Kuo, A., Manohar, M., Mao, R., Neeli, I., Neubauer, A., Oniyide, O., Powell, A., Puri, R., Renz, H., Schapiro, J., Weidenbacher, P., Wittman, R., Ahuja, N., Chung, H., Jagannathan, P., James, J., Kim, P., Meyer, N., Nadeau, K., Radic, M., Robinson, W., Singh, U., Wang, T., Wherry, J., Skevaki, C., Prak, E., Utz, P. WILEY. 2021: 3202-3205
  • An aberrant inflammatory response in severe COVID-19. Cell host & microbe Merad, M., Subramanian, A., Wang, T. T. 2021; 29 (7): 1043-1047

    Abstract

    Severe COVID-19 arises from the convergence of inadequate pre-existing immunity and a host response that damages, rather than repairs, tissues. We outline clinical presentations of COVID-19 that are likely driven by dysregulated host immunity, discuss potential mechanisms underlying pathological responses, and highlight important areas for basic research on this topic.

    View details for DOI 10.1016/j.chom.2021.06.018

    View details for PubMedID 34265243

  • Immunity after SARS-CoV-2 infections. Nature immunology Jagannathan, P., Wang, T. T. 2021

    View details for DOI 10.1038/s41590-021-00923-3

    View details for PubMedID 33875881

  • Engineering luminescent biosensors for point-of-care SARS-CoV-2 antibody detection. Nature biotechnology Elledge, S. K., Zhou, X. X., Byrnes, J. R., Martinko, A. J., Lui, I., Pance, K., Lim, S. A., Glasgow, J. E., Glasgow, A. A., Turcios, K., Iyer, N. S., Torres, L., Peluso, M. J., Henrich, T. J., Wang, T. T., Tato, C. M., Leung, K. K., Greenhouse, B., Wells, J. A. 2021

    Abstract

    Current serology tests for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies mainly take the form of enzyme-linked immunosorbent assays, chemiluminescent microparticle immunoassays or lateral flow assays, which are either laborious, expensive or lacking sufficient sensitivity and scalability. Here we present the development and validation of a rapid, low-cost, solution-based assay to detect antibodies in serum, plasma, whole blood and to a lesser extent saliva, using rationally designed split luciferase antibody biosensors. This new assay, which generates quantitative results in 30min, substantially reduces the complexity and improves the scalability of coronavirus disease 2019 (COVID-19) antibody tests. This assay is well-suited for point-of-care, broad population testing, and applications in low-resource settings, for monitoring host humoral responses to vaccination or viral infection.

    View details for DOI 10.1038/s41587-021-00878-8

    View details for PubMedID 33767397

  • Peginterferon Lambda-1a for treatment of outpatients with uncomplicated COVID-19: a randomized placebo-controlled trial. Nature communications Jagannathan, P. n., Andrews, J. R., Bonilla, H. n., Hedlin, H. n., Jacobson, K. B., Balasubramanian, V. n., Purington, N. n., Kamble, S. n., de Vries, C. R., Quintero, O. n., Feng, K. n., Ley, C. n., Winslow, D. n., Newberry, J. n., Edwards, K. n., Hislop, C. n., Choong, I. n., Maldonado, Y. n., Glenn, J. n., Bhatt, A. n., Blish, C. n., Wang, T. n., Khosla, C. n., Pinsky, B. A., Desai, M. n., Parsonnet, J. n., Singh, U. n. 2021; 12 (1): 1967

    Abstract

    Type III interferons have been touted as promising therapeutics in outpatients with coronavirus disease 2019 (COVID-19). We conducted a randomized, single-blind, placebo-controlled trial (NCT04331899) in 120 outpatients with mild to moderate COVID-19 to determine whether a single, 180 mcg subcutaneous dose of Peginterferon Lambda-1a (Lambda) within 72 hours of diagnosis could shorten the duration of viral shedding (primary endpoint) or symptoms (secondary endpoint). In both the 60 patients receiving Lambda and 60 receiving placebo, the median time to cessation of viral shedding was 7 days (hazard ratio [HR] = 0.81; 95% confidence interval [CI] 0.56 to 1.19). Symptoms resolved in 8 and 9 days in Lambda and placebo, respectively, and symptom duration did not differ significantly between groups (HR 0.94; 95% CI 0.64 to 1.39). Both Lambda and placebo were well-tolerated, though liver transaminase elevations were more common in the Lambda vs. placebo arm (15/60 vs 5/60; p = 0.027). In this study, a single dose of subcutaneous Peginterferon Lambda-1a neither shortened the duration of SARS-CoV-2 viral shedding nor improved symptoms in outpatients with uncomplicated COVID-19.

    View details for DOI 10.1038/s41467-021-22177-1

    View details for PubMedID 33785743

  • Illuminating the Fc dependence of SARS-CoV-2 neutralization. Immunity González, J. C., Wang, T. T. 2021

    Abstract

    Monoclonal antibodies show efficacy in treating COVID-19, but the functional requirements for protection are unclear. In this issue of Immunity, Ullah et al. (2021) develop a stable SARS-CoV-2 reporter virus and use bioluminescence imaging to longitudinally monitor infection and assess neutralizing monoclonal antibody interventions in mice. They find that antibody-mediated protection depends on the Fc domain and Fc-gamma receptor-expressing immune cells.

    View details for DOI 10.1016/j.immuni.2021.08.024

    View details for PubMedID 34464594

  • Proinflammatory IgG Fc structures in patients with severe COVID-19 Nature Immunology Chakraborty, S., Gonzalez, J., Edwards, K., ..., Wang, T. T. 2021
  • SARS-CoV-2 vaccines in advanced clinical trials: where do we stand. Advanced drug delivery reviews Chakraborty, S. n., Mallajosyula, V. n., Tato, C. M., Tan, G. S., Wang, T. T. 2021

    Abstract

    The ongoing SARS-CoV-2 pandemic has led to the focused application of resources and scientific expertise toward the goal of developing investigational vaccines to prevent COVID-19. The highly collaborative global efforts by private industry, governments and non-governmental organizations have resulted in a number of SARS-CoV-2 vaccine candidates moving to Phase III trials in a period of only months since the start of the pandemic. In this review, we provide an overview of the preclinical and clinical data on SARS-CoV-2 vaccines that are currently in Phase III clinical trials and in few cases authorized for emergency use. We further discuss relevant vaccine platforms and provide a discussion of SARS-CoV-2 antigens that may be targeted to increase the breadth and durability of vaccine responses.

    View details for DOI 10.1016/j.addr.2021.01.014

    View details for PubMedID 33482248

    View details for PubMedCentralID PMC7816567

  • Immunoglobulin E sialylation regulates allergic responses. Immunology and cell biology Xie, M. M., Bertozzi, C. R., Wang, T. T. 2020

    Abstract

    Shade et al. demonstrate that people with peanut allergies produce IgE antibodies that are enriched for sialic acid-containing glycoforms. The sialylated IgE triggered significantly more degranulation by basophils and mast cells, suggesting intrinsic functional differences between IgEs from allergic and nonallergic subjects.

    View details for DOI 10.1111/imcb.12368

    View details for PubMedID 32632971

  • Maternal Anti-Dengue IgG Fucosylation Predicts Susceptibility to Dengue Disease in Infants. Cell reports Thulin, N. K., Brewer, R. C., Sherwood, R., Bournazos, S., Edwards, K. G., Ramadoss, N. S., Taubenberger, J. K., Memoli, M., Gentles, A. J., Jagannathan, P., Zhang, S., Libraty, D. H., Wang, T. T. 2020; 31 (6): 107642

    Abstract

    Infant mortality from dengue disease is a devastating global health burden that could be minimized with the ability to identify susceptibility for severe disease prior to infection. Although most primary infant dengue infections are asymptomatic, maternally derived anti-dengue immunoglobulin G (IgGs) present during infection can trigger progression to severe disease through antibody-dependent enhancement mechanisms. Importantly, specific characteristics of maternal IgGs that herald progression to severe infant dengue are unknown. Here, we define ≥10% afucosylation of maternal anti-dengue IgGs as a risk factor for susceptibility of infants to symptomatic dengue infections. Mechanistic experiments show that afucosylation of anti-dengue IgGs promotes FcgammaRIIIa signaling during infection, in turn enhancing dengue virus replication in FcgammaRIIIa+ monocytes. These studies identify a post-translational modification of anti-dengue IgGs that correlates with risk for symptomatic infant dengue infections and define a mechanism by which afucosylated antibodies and FcgammaRIIIa enhance dengue infections.

    View details for DOI 10.1016/j.celrep.2020.107642

    View details for PubMedID 32402275

  • Defining the features and duration of antibody responses to SARS-CoV-2 infection associated with disease severity and outcome. Science immunology Röltgen, K. n., Powell, A. E., Wirz, O. F., Stevens, B. A., Hogan, C. A., Najeeb, J. n., Hunter, M. n., Wang, H. n., Sahoo, M. K., Huang, C. n., Yamamoto, F. n., Manohar, M. n., Manalac, J. n., Otrelo-Cardoso, A. R., Pham, T. D., Rustagi, A. n., Rogers, A. J., Shah, N. H., Blish, C. A., Cochran, J. R., Jardetzky, T. S., Zehnder, J. L., Wang, T. T., Narasimhan, B. n., Gombar, S. n., Tibshirani, R. n., Nadeau, K. C., Kim, P. S., Pinsky, B. A., Boyd, S. D. 2020; 5 (54)

    Abstract

    SARS-CoV-2-specific antibodies, particularly those preventing viral spike receptor binding domain (RBD) interaction with host angiotensin-converting enzyme 2 (ACE2) receptor, can neutralize the virus. It is, however, unknown which features of the serological response may affect clinical outcomes of COVID-19 patients. We analyzed 983 longitudinal plasma samples from 79 hospitalized COVID-19 patients and 175 SARS-CoV-2-infected outpatients and asymptomatic individuals. Within this cohort, 25 patients died of their illness. Higher ratios of IgG antibodies targeting S1 or RBD domains of spike compared to nucleocapsid antigen were seen in outpatients who had mild illness versus severely ill patients. Plasma antibody increases correlated with decreases in viral RNAemia, but antibody responses in acute illness were insufficient to predict inpatient outcomes. Pseudovirus neutralization assays and a scalable ELISA measuring antibodies blocking RBD-ACE2 interaction were well correlated with patient IgG titers to RBD. Outpatient and asymptomatic individuals' SARS-CoV-2 antibodies, including IgG, progressively decreased during observation up to five months post-infection.

    View details for DOI 10.1126/sciimmunol.abe0240

    View details for PubMedID 33288645

  • Engineering luminescent biosensors for point-of-care SARS-CoV-2 antibody detection. medRxiv : the preprint server for health sciences Elledge, S. K., Zhou, X. X., Byrnes, J. R., Martinko, A. J., Lui, I. n., Pance, K. n., Lim, S. A., Glasgow, J. E., Glasgow, A. A., Turcios, K. n., Iyer, N. n., Torres, L. n., Peluso, M. J., Henrich, T. J., Wang, T. T., Tato, C. M., Leung, K. K., Greenhouse, B. n., Wells, J. A. 2020

    Abstract

    Current serology tests for SARS-CoV-2 antibodies mainly take the form of enzyme-linked immunosorbent assays or lateral flow assays, with the former being laborious and the latter being expensive and often lacking sufficient sensitivity and scalability. Here we present the development and validation of a rapid, low-cost solution-based assay to detect antibodies in serum, plasma, whole blood, and saliva, using rationally designed split luciferase antibody biosensors (spLUC). This new assay, which generates quantitative results in as short as 5 minutes, substantially reduces the complexity and improves the scalability of COVID-19 antibody tests for point-of-care and broad population testing.

    View details for DOI 10.1101/2020.08.17.20176925

    View details for PubMedID 32839788

    View details for PubMedCentralID PMC7444307

  • FcRn, but not FcγRs, drives maternal-fetal transplacental transport of human IgG antibodies. Proceedings of the National Academy of Sciences of the United States of America Borghi, S. n., Bournazos, S. n., Thulin, N. K., Li, C. n., Gajewski, A. n., Sherwood, R. W., Zhang, S. n., Harris, E. n., Jagannathan, P. n., Wang, L. X., Ravetch, J. V., Wang, T. T. 2020

    Abstract

    The IgG Fc domain has the capacity to interact with diverse types of receptors, including the neonatal Fc receptor (FcRn) and Fcγ receptors (FcγRs), which confer pleiotropic biological activities. Whereas FcRn regulates IgG epithelial transport and recycling, Fc effector activities, such as antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis, are mediated by FcγRs, which upon cross-linking transduce signals that modulate the function of effector leukocytes. Despite the well-defined and nonoverlapping functional properties of FcRn and FcγRs, recent studies have suggested that FcγRs mediate transplacental IgG transport, as certain Fc glycoforms were reported to be enriched in fetal circulation. To determine the contribution of FcγRs and FcRn to the maternal-fetal transport of IgG, we characterized the IgG Fc glycosylation in paired maternal-fetal samples from patient cohorts from Uganda and Nicaragua. No differences in IgG1 Fc glycan profiles and minimal differences in IgG2 Fc glycans were noted, whereas the presence or absence of galactose on the Fc glycan of IgG1 did not alter FcγRIIIa or FcRn binding, half-life, or their ability to deplete target cells in FcγR/FcRn humanized mice. Modeling maternal-fetal transport in FcγR/FcRn humanized mice confirmed that only FcRn contributed to transplacental transport of IgG; IgG selectively enhanced for FcRn binding resulted in enhanced accumulation of maternal antibody in the fetus. In contrast, enhancing FcγRIIIa binding did not result in enhanced maternal-fetal transport. These results argue against a role for FcγRs in IgG transplacental transport, suggesting Fc engineering of maternally administered antibody to enhance only FcRn binding as a means to improve maternal-fetal transport of IgG.

    View details for DOI 10.1073/pnas.2004325117

    View details for PubMedID 32461366

  • Competitive SARS-CoV-2 Serology Reveals Most Antibodies Targeting the Spike Receptor-Binding Domain Compete for ACE2 Binding. mSphere Byrnes, J. R., Zhou, X. X., Lui, I. n., Elledge, S. K., Glasgow, J. E., Lim, S. A., Loudermilk, R. P., Chiu, C. Y., Wang, T. T., Wilson, M. R., Leung, K. K., Wells, J. A. 2020; 5 (5)

    Abstract

    As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread around the world, there is an urgent need for new assay formats to characterize the humoral response to infection. Here, we present an efficient, competitive serological assay that can simultaneously determine an individual's seroreactivity against the SARS-CoV-2 Spike protein and determine the proportion of anti-Spike antibodies that block interaction with the human angiotensin-converting enzyme 2 (ACE2) required for viral entry. In this approach based on the use of enzyme-linked immunosorbent assays (ELISA), we present natively folded viral Spike protein receptor-binding domain (RBD)-containing antigens via avidin-biotin interactions. Sera are then competed with soluble ACE2-Fc, or with a higher-affinity variant thereof, to determine the proportion of ACE2 blocking anti-RBD antibodies. Assessment of sera from 144 SARS-CoV-2 patients ultimately revealed that a remarkably consistent and high proportion of antibodies in the anti-RBD pool targeted the epitope responsible for ACE2 engagement (83% ± 11%; 50% to 107% signal inhibition in our largest cohort), further underscoring the importance of tailoring vaccines to promote the development of such antibodies.IMPORTANCE With the emergence and continued spread of the SARS-CoV-2 virus, and of the associated disease, coronavirus disease 2019 (COVID-19), there is an urgent need for improved understanding of how the body mounts an immune response to the virus. Here, we developed a competitive SARS-CoV-2 serological assay that can simultaneously determine whether an individual has developed antibodies against the SARS-CoV-2 Spike protein receptor-binding domain (RBD) and measure the proportion of these antibodies that block interaction with the human angiotensin-converting enzyme 2 (ACE2) required for viral entry. Using this assay and 144 SARS-CoV-2 patient serum samples, we found that a majority of anti-RBD antibodies compete for ACE2 binding. These results not only highlight the need to design vaccines to generate such blocking antibodies but also demonstrate the utility of this assay to rapidly screen patient sera for potentially neutralizing antibodies.

    View details for DOI 10.1128/mSphere.00802-20

    View details for PubMedID 32938700

  • Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell Blanco-Melo, D. n., Nilsson-Payant, B. E., Liu, W. C., Uhl, S. n., Hoagland, D. n., Møller, R. n., Jordan, T. X., Oishi, K. n., Panis, M. n., Sachs, D. n., Wang, T. T., Schwartz, R. E., Lim, J. K., Albrecht, R. A., tenOever, B. R. 2020; 181 (5): 1036–45.e9

    Abstract

    Viral pandemics, such as the one caused by SARS-CoV-2, pose an imminent threat to humanity. Because of its recent emergence, there is a paucity of information regarding viral behavior and host response following SARS-CoV-2 infection. Here we offer an in-depth analysis of the transcriptional response to SARS-CoV-2 compared with other respiratory viruses. Cell and animal models of SARS-CoV-2 infection, in addition to transcriptional and serum profiling of COVID-19 patients, consistently revealed a unique and inappropriate inflammatory response. This response is defined by low levels of type I and III interferons juxtaposed to elevated chemokines and high expression of IL-6. We propose that reduced innate antiviral defenses coupled with exuberant inflammatory cytokine production are the defining and driving features of COVID-19.

    View details for DOI 10.1016/j.cell.2020.04.026

    View details for PubMedID 32416070

    View details for PubMedCentralID PMC7227586

  • Human B Cell Clonal Expansion and Convergent Antibody Responses to SARS-CoV-2. Cell host & microbe Nielsen, S. C., Yang, F. n., Jackson, K. J., Hoh, R. A., Röltgen, K. n., Jean, G. H., Stevens, B. A., Lee, J. Y., Rustagi, A. n., Rogers, A. J., Powell, A. E., Hunter, M. n., Najeeb, J. n., Otrelo-Cardoso, A. R., Yost, K. E., Daniel, B. n., Nadeau, K. C., Chang, H. Y., Satpathy, A. T., Jardetzky, T. S., Kim, P. S., Wang, T. T., Pinsky, B. A., Blish, C. A., Boyd, S. D. 2020

    Abstract

    B cells are critical for the production of antibodies and protective immunity to viruses. Here we show that patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) who develop coronavirus disease 2019 (COVID-19) display early recruitment of B cells expressing a limited subset of IGHV genes, progressing to a highly polyclonal response of B cells with broader IGHV gene usage and extensive class switching to IgG and IgA subclasses with limited somatic hypermutation in the initial weeks of infection. We identify convergence of antibody sequences across SARS-CoV-2-infected patients, highlighting stereotyped naive responses to this virus. Notably, sequence-based detection in COVID-19 patients of convergent B cell clonotypes previously reported in SARS-CoV infection predicts the presence of SARS-CoV/SARS-CoV-2 cross-reactive antibody titers specific for the receptor-binding domain. These findings offer molecular insights into shared features of human B cell responses to SARS-CoV-2 and SARS-CoV.

    View details for DOI 10.1016/j.chom.2020.09.002

    View details for PubMedID 32941787

  • Proinflammatory IgG Fc structures in patients with severe COVID-19 Nature Immunology Chakraborty, S., Gonzales, J., Edwards, K., Mallajosyulla, V., Buzzanco, A. S., Sherwood, R., Buffone, C., Kathale, N., Providenza, S., Xie, M. M., Andrews, J. R., Blish, C. A., Singh, U., Dugan, H., Wilson, P. C., Pham, T. D., Boyd, S. D., Nadeau, K. C., Pinsky, B. A., Zhang, S., Memoli, M. J., Taubenberger, J. K., Morales, T., Schapiro, J. M., Tan, G. S., et al 2020
  • Functional diversification of IgGs through Fc glycosylation JOURNAL OF CLINICAL INVESTIGATION Wang, T. T., Ravetch, J. 2019; 129 (9): 3492–98

    Abstract

    IgG antibodies are secreted from B cells and bind to a variety of pathogens to control infections as well as contribute to inflammatory diseases. Many of the functions of IgGs are mediated through Fcγ receptors (FcγRs), which transduce interactions with immune complexes, leading to a variety of cellular outcomes depending on the FcγRs and cell types engaged. Which FcγRs and cell types will be engaged during an immune response depends on the structure of Fc domains within immune complexes that are formed when IgGs bind to cognate antigen(s). Recent studies have revealed an unexpected degree of structural variability in IgG Fc domains among people, driven primarily by differences in IgG subclasses and N-linked glycosylation of the CH2 domain. This translates, in turn, to functional immune diversification through type I and type II FcγR-mediated cellular functions. For example, Fc domain sialylation triggers conformational changes of IgG1 that enable interactions with type II FcγRs; these receptors mediate cellular functions including antiinflammatory activity or definition of thresholds for B cell selection based on B cell receptor affinity. Similarly, presence or absence of a core fucose alters type I FcγR binding of IgG1 by modulating the Fc's affinity for FcγRIIIa, thereby altering its proinflammatory activity. How heterogeneity in IgG Fc domains contributes to human immune diversity is now being elucidated, including impacts on vaccine responses and susceptibility to disease and its sequelae during infections. Here, we discuss how Fc structures arising from sialylation and fucosylation impact immunity, focusing on responses to vaccination and infection. We also review work defining individual differences in Fc glycosylation, regulation of Fc glycosylation, and clinical implications of these pathways.

    View details for DOI 10.1172/JCI130029

    View details for Web of Science ID 000484368600006

    View details for PubMedID 31478910

    View details for PubMedCentralID PMC6715372

  • IgG Fc Glycosylation in Human Immunity. Current topics in microbiology and immunology Wang, T. T. 2019

    Abstract

    Glycosylation of IgG Fc domains is a central mechanism in the diversification of antibody function. Modifications to the core Fc glycan impact antibody function by shifting the balance of Type I and Type II Fc gamma receptors (FcγR) that will be engaged by immune complexes. This, in turn, modulates the effector cells and functions that can be recruited during immune activation. Critically, humans have evolved to regulate Fc glycan modifications for immune homeostasis. Dysregulation in Fc glycan modifications can lead to loss of immune tolerance, symptomatic autoimmunity, and susceptibility to infectious diseases. Here, we discuss IgG Fc glycosylation and its role in human health and disease.

    View details for DOI 10.1007/82_2019_152

    View details for PubMedID 30805712

  • Immunity by Design. Cell host & microbe Wang, T. T. 2018; 23 (4): 430–31

    Abstract

    One outcome of the many advances in basic sciences that have been made over the last decades is the prospect of rational vaccine design. A recent publication by Du et al. (2018) describes a screening method for selection of live-attenuated viral vaccine platforms with enhanced immune-stimulatory properties.

    View details for DOI 10.1016/j.chom.2018.03.017

    View details for PubMedID 29649438

  • The Role of Fc Gamma Receptors in Broad Protection against Influenza Viruses. Vaccines Thulin, N. K., Wang, T. T. 2018; 6 (3)

    Abstract

    Recent studies have revealed multiple roles for Fc gamma receptors (FcγRs) in broad immunity against influenza viruses. Activating FcγR pathways can be harnessed to confer protection mediated by non-neutralizing anti-HA IgGs and to increase the potency of broadly neutralizing anti-HA IgGs and of anti-NA IgGs. Separate FcγR pathways can be targeted to enhance the breadth of antibody responses elicited by seasonal influenza virus vaccines. Here, we review the current understanding of FcγR pathways in broad influenza immunity and suggest mechanisms to bypass FcγR signaling heterogeneity among people that arises from distinctions in structural repertoires of IgG Fc domains.

    View details for DOI 10.3390/vaccines6030036

    View details for PubMedID 29966222

  • Immunological responses to influenza vaccination: lessons for improving vaccine efficacy. Current opinion in immunology Wang, T. T., Bournazos, S. n., Ravetch, J. V. 2018; 53: 124–29

    Abstract

    A critical factor in the maturation of influenza vaccine responses is the nearly inevitable binding of vaccine antigens by exiting anti-influenza IgGs. These antigen-IgG immune complexes direct the response to immunization by modulating cellular processes that determine antibody and T-cell repertoires: maturation of dendritic cells, processing and presentation of antigens to T cells, trafficking of antigens to the germinal center, and selection of B cells for antibody production. By focusing on the recent advances in the study of the immunomodulatory processes mediated by IgG immune complexes upon influenza vaccination, we discuss a pathway that is critical for modulating the breadth and potency of anti-HA antibody responses and has previously led to the development of strategies to improve influenza vaccine efficacy.

    View details for DOI 10.1016/j.coi.2018.04.026

    View details for PubMedID 29753885

  • IgG antibodies to dengue enhanced for FcγRIIIA binding determine disease severity. Science (New York, N.Y.) Wang, T. T., Sewatanon, J., Memoli, M. J., Wrammert, J., Bournazos, S., Bhaumik, S. K., Pinsky, B. A., Chokephaibulkit, K., Onlamoon, N., Pattanapanyasat, K., Taubenberger, J. K., Ahmed, R., Ravetch, J. V. 2017; 355 (6323): 395-398

    Abstract

    Dengue virus (DENV) infection in the presence of reactive, non-neutralizing immunoglobulin G (IgG) (RNNIg) is the greatest risk factor for dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). Progression to DHF/DSS is attributed to antibody-dependent enhancement (ADE); however, because only a fraction of infections occurring in the presence of RNNIg advance to DHF/DSS, the presence of RNNIg alone cannot account for disease severity. We discovered that DHF/DSS patients respond to infection by producing IgGs with enhanced affinity for the activating Fc receptor FcγRIIIA due to afucosylated Fc glycans and IgG1 subclass. RNNIg enriched for afucosylated IgG1 triggered platelet reduction in vivo and was a significant risk factor for thrombocytopenia. Thus, therapeutics and vaccines restricting production of afucosylated, IgG1 RNNIg during infection may prevent ADE of DENV disease.

    View details for DOI 10.1126/science.aai8128

    View details for PubMedID 28126818

  • Signaling by Antibodies: Recent Progress Annual Review of Immunology Bournazos, S., Wang, T. T., Dahan, R., Maamary, J., Ravetch, J. V. 2017; 35 (April 26): 285-311
  • Signaling by Antibodies: Recent Progress ANNUAL REVIEW OF IMMUNOLOGY, VOL 35 Bournazos, S., Wang, T. T., Dahan, R., Maamary, J., Ravetch, J. V., Littman, D. R., Yokoyama, W. M. 2017; 35: 285–311

    Abstract

    IgG antibodies mediate a diversity of immune functions by coupling of antigen specificity through the Fab domain to signal transduction via Fc-Fc receptor interactions. Indeed, balanced IgG signaling through type I and type II Fc receptors is required for the control of proinflammatory, anti-inflammatory, and immunomodulatory processes. In this review, we discuss the mechanisms that govern IgG-Fc receptor interactions, highlighting the diversity of Fc receptor-mediated effector functions that regulate immunity and inflammation as well as determine susceptibility to infection and autoimmunity and responsiveness to antibody-based therapeutics and vaccines.

    View details for DOI 10.1146/annurev-immunol-051116-052433

    View details for Web of Science ID 000401334800011

    View details for PubMedID 28446061

    View details for PubMedCentralID PMC5613280

  • Sex Differences in Autoimmune Diseases HORMONES, BRAIN AND BEHAVIOR, VOL 4: CLINICALLY IMPORTANT HORMONE EFFECTS ON BRAIN AND BEHAVIOR, 3RD EDITION Voskuhl, R., Wang, T. T., Lightman, S., Pfaff, D. W., Joels, M. 2017: 445–72
  • Increasing the breadth and potency of response to the seasonal influenza virus vaccine by immune complex immunization. Proceedings of the National Academy of Sciences of the United States of America Maamary, J. n., Wang, T. T., Tan, G. S., Palese, P. n., Ravetch, J. V. 2017

    Abstract

    The main barrier to reduction of morbidity caused by influenza is the absence of a vaccine that elicits broad protection against different virus strains. Studies in preclinical models of influenza virus infections have shown that antibodies alone are sufficient to provide broad protection against divergent virus strains in vivo. Here, we address the challenge of identifying an immunogen that can elicit potent, broadly protective, antiinfluenza antibodies by demonstrating that immune complexes composed of sialylated antihemagglutinin antibodies and seasonal inactivated flu vaccine (TIV) can elicit broadly protective antihemagglutinin antibodies. Further, we found that an Fc-modified, bispecific monoclonal antibody against conserved epitopes of the hemagglutinin can be combined with TIV to elicit broad protection, thus setting the stage for a universal influenza virus vaccine.

    View details for DOI 10.1073/pnas.1707950114

    View details for PubMedID 28874545

  • The Role and Function of Fcγ Receptors on Myeloid Cells. Microbiology spectrum Bournazos, S. n., Wang, T. T., Ravetch, J. V. 2016; 4 (6)

    Abstract

    A key determinant for the survival of organisms is their capacity to recognize and respond efficiently to foreign antigens. This is largely accomplished by the orchestrated activity of the innate and adaptive branches of the immune system. Antibodies are specifically generated in response to foreign antigens, facilitating thereby the specific recognition of antigens of almost infinite diversity. Receptors specific for the Fc domain of antibodies, Fc receptors, are expressed on the surface of the various myeloid leukocyte populations and mediate the binding and recognition of antibodies by innate leukocytes. By directly linking the innate and the adaptive components of immunity, Fc receptors play a central role in host defense and the maintenance of tissue homeostasis through the induction of diverse proinflammatory, anti-inflammatory, and immunomodulatory processes that are initiated upon engagement by the Fc domain. In this chapter, we discuss the mechanisms that regulate Fc domain binding to the various types of Fc receptors and provide an overview of the astonishing diversity of effector functions that are mediated through Fc-FcR interactions on myeloid cells. Lastly, we discuss the impact of FcR-mediated interactions in the context of IgG-mediated inflammation, autoimmunity, susceptibility to infection, and responsiveness to antibody-based therapeutics.

    View details for DOI 10.1128/microbiolspec.MCHD-0045-2016

    View details for PubMedID 28087938

    View details for PubMedCentralID PMC5240797

  • Sex Differences in Autoimmune Disease Hormones, Brain and Behavior Voskuhl, R., Wang, T. T. Academic Press. 2016; 3: 445–465
  • Anti-HA Glycoforms Drive B Cell Affinity Selection and Determine Influenza Vaccine Efficacy CELL Wang, T. T., Maamary, J., Tan, G. S., Bournazos, S., Davis, C. W., Krammer, F., Schlesinger, S. J., Palese, P., Ahmed, R., Ravetch, J. V. 2015; 162 (1): 160-169

    Abstract

    Protective vaccines elicit high-affinity, neutralizing antibodies by selection of somatically hypermutated B cell antigen receptors (BCR) on immune complexes (ICs). This implicates Fc-Fc receptor (FcR) interactions in affinity maturation, which, in turn, are determined by IgG subclass and Fc glycan composition within ICs. Trivalent influenza virus vaccination elicited regulation of anti-hemagglutinin (HA) IgG subclass and Fc glycans, with abundance of sialylated Fc glycans (sFc) predicting quality of vaccine response. We show that sFcs drive BCR affinity selection by binding the Type-II FcR CD23, thus upregulating the inhibitory FcγRIIB on activated B cells. This elevates the threshold requirement for BCR signaling, resulting in B cell selection for higher affinity BCR. Immunization with sFc HA ICs elicited protective, high-affinity IgGs against the conserved stalk of the HA. These results reveal a novel, endogenous pathway for affinity maturation that can be exploited for eliciting high-affinity, broadly neutralizing antibodies through immunization with sialylated immune complexes.

    View details for DOI 10.1016/j.cell.2015.06.026

    View details for Web of Science ID 000357542300017

    View details for PubMedID 26140596

    View details for PubMedCentralID PMC4594835

  • IgG anti-HA Fc glycoform modulation is predictive of influenza vaccine efficacy Wang, T., Maamary, J., Schlesinger, S., Ravetch, J. AMER ASSOC IMMUNOLOGISTS. 2015
  • Immune Complexes: Not Just an Innocent Bystander in Chronic Viral Infection IMMUNITY Wang, T. T., Ravetch, J. V. 2015; 42 (2): 213-215

    Abstract

    Understanding of how persistent viral infection impacts humoral immunity is incomplete. In this issue of Immunity, Wieland et al. (2015) and Yamada et al. (2015) find that high amounts of IgG-antigen complexes formed during chronic lymphocytic choriomeningitis infection can interfere with Fcγ-receptor-mediated effector activities, potentially contributing to immune dysfunction.

    View details for DOI 10.1016/j.immuni.2015.01.022

    View details for Web of Science ID 000349916400006

    View details for PubMedID 25692698

  • Type I and type II Fc receptors regulate innate and adaptive immunity NATURE IMMUNOLOGY Pincetic, A., Bournazos, S., DiLillo, D. J., Maamary, J., Wang, T. T., Dahan, R., Fiebiger, B., Ravetch, J. V. 2014; 15 (8): 707-716

    Abstract

    Antibodies produced in response to a foreign antigen are characterized by polyclonality, not only in the diverse epitopes to which their variable domains bind but also in the various effector molecules to which their constant regions (Fc domains) engage. Thus, the antibody's Fc domain mediates diverse effector activities by engaging two distinct classes of Fc receptors (type I and type II) on the basis of the two dominant conformational states that the Fc domain may adopt. These conformational states are regulated by the differences among antibody subclasses in their amino acid sequence and by the complex, biantennary Fc-associated N-linked glycan. Here we discuss the diverse downstream proinflammatory, anti-inflammatory and immunomodulatory consequences of the engagement of type I and type II Fc receptors in the context of infectious, autoimmune, and neoplastic disorders.

    View details for DOI 10.1038/ni.2939

    View details for Web of Science ID 000339323000006

    View details for PubMedID 25045879

  • Emergence and evolution of the 1918, 1957, 1968, and 2009 pandemic virus strains Textbook of Influenza Wang, T. T., Palese, P. John Wiley & Sons. 2013; 2
  • Emergence and evolution of the 1918, 1957, 1968, and 2009 pandemic virus strains TEXTBOOK OF INFLUENZA, 2ND EDITION Wang, T. T., Palese, P., Webster, R. G., Monto, A. S., Braciale, T. J., Lamb, R. A. 2013: 218–28
  • Seroevidence for H5N1 Influenza Infections in Humans: Meta-Analysis SCIENCE Wang, T. T., Parides, M. K., Palese, P. 2012; 335 (6075): 1463-1463

    Abstract

    The prevalence of avian H5N1 influenza A infections in humans has not been definitively determined. Cases of H5N1 infection in humans confirmed by the World Health Organization (WHO) are fewer than 600 in number, with an overall case fatality rate of >50%. We hypothesize that the stringent criteria for confirmation of a human case of H5N1 by WHO do not account for a majority of infections but rather the select few hospitalized cases that are more likely to be severe and result in poor clinical outcome. Meta-analysis shows that 1 to 2% of more than 12,500 study participants from 20 studies had seroevidence for prior H5N1 infection.

    View details for DOI 10.1126/science.1218888

    View details for Web of Science ID 000301837000037

    View details for PubMedID 22362880

  • H5N1 influenza viruses: Facts, not fear PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Palese, P., Wang, T. T. 2012; 109 (7): 2211-2213

    Abstract

    The ongoing controversy over publication of two studies involving the transmission in ferrets of H5N1 (H5) subtype influenza viruses and the recommendations of the National Science Advisory Board for Biosecurity to redact key details in the manuscripts call for an examination of relevant scientific facts. In addition, there are calls in the media to destroy the viruses, curtail future research in this area, and protect the public from such "frightening" research efforts. Fear needs to be put to rest with solid science and not speculation.

    View details for DOI 10.1073/pnas.1121297109

    View details for Web of Science ID 000300489200018

    View details for PubMedID 22308474

  • Hemagglutinin stalk antibodies elicited by the 2009 pandemic influenza virus as a mechanism for the extinction of seasonal H1N1 viruses PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Pica, N., Hai, R., Krammer, F., Wang, T. T., Maamary, J., Eggink, D., Tan, G. S., Krause, J. C., Moran, T., Stein, C. R., Banach, D., Wrammert, J., Belshe, R. B., Garcia-Sastre, A., Palese, P. 2012; 109 (7): 2573-2578

    Abstract

    After the emergence of pandemic influenza viruses in 1957, 1968, and 2009, existing seasonal viruses were observed to be replaced in the human population by the novel pandemic strains. We have previously hypothesized that the replacement of seasonal strains was mediated, in part, by a population-scale boost in antibodies specific for conserved regions of the hemagglutinin stalk and the viral neuraminidase. Numerous recent studies have shown the role of stalk-specific antibodies in neutralization of influenza viruses; the finding that stalk antibodies can effectively neutralize virus alters the existing dogma that influenza virus neutralization is mediated solely by antibodies that react with the globular head of the viral hemagglutinin. The present study explores the possibility that stalk-specific antibodies were boosted by infection with the 2009 H1N1 pandemic virus and that those antibodies could have contributed to the disappearance of existing seasonal H1N1 influenza virus strains. To study stalk-specific antibodies, we have developed chimeric hemagglutinin constructs that enable the measurement of antibodies that bind the hemagglutinin protein and neutralize virus but do not have hemagglutination inhibition activity. Using these chimeric hemagglutinin reagents, we show that infection with the 2009 pandemic H1N1 virus elicited a boost in titer of virus-neutralizing antibodies directed against the hemagglutinin stalk. In addition, we describe assays that can be used to measure influenza virus-neutralizing antibodies that are not detected in the traditional hemagglutination inhibition assay.

    View details for DOI 10.1073/pnas.1200039109

    View details for Web of Science ID 000300489200081

    View details for PubMedID 22308500

  • Why Do Influenza Virus Subtypes Die Out? A Hypothesis MBIO Palese, P., Wang, T. T. 2011; 2 (5)

    Abstract

    Novel pandemic influenza viruses enter the human population with some regularity and can cause disease that is severe and widespread. The emergence of novel viruses, historically, has often been coupled with the disappearance of existing seasonal virus strains. Here, we propose that the elimination of seasonal strains during virus pandemics is a process mediated, at the population level, by humoral immunity. Specifically, we suggest that infection with a novel virus strain, in people previously exposed to influenza viruses, can elicit a memory B cell response against conserved hemagglutinin stalk epitopes and/or neuraminidase epitopes. The anti-stalk and/or anti-neuraminidase antibodies then act to diminish the clinical severity of disease caused by novel influenza viruses and to eliminate seasonal virus strains.

    View details for DOI 10.1128/mBio.00150-11

    View details for Web of Science ID 000296844300005

    View details for PubMedID 21878571

  • Biochemistry. Catching a moving target. Science Wang, T. T., Palese, P. 2011; 333 (6044): 834-835

    View details for DOI 10.1126/science.1210724

    View details for PubMedID 21836007

  • Vaccination with a synthetic peptide from the influenza virus hemagglutinin provides protection against distinct viral subtypes PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wang, T. T., Tan, G. S., Hai, R., Pica, N., Ngai, L., Ekiert, D. C., Wilson, I. A., Garcia-Sastre, A., Moran, T. M., Palese, P. 2010; 107 (44): 18979-18984

    Abstract

    Current influenza virus vaccines protect mostly against homologous virus strains; thus, regular immunization with updated vaccine formulations is necessary to guard against the virus' hallmark remodeling of regions that mediate neutralization. Development of a broadly protective influenza vaccine would mark a significant advance in human infectious diseases research. Antibodies with broad neutralizing activity (nAbs) against multiple influenza virus strains or subtypes have been reported to bind the stalk of the viral hemagglutinin, suggesting that a vaccine based on this region could elicit a broadly protective immune response. Here we describe a hemagglutinin subunit 2 protein (HA2)-based synthetic peptide vaccine that provides protection in mice against influenza viruses of the structurally divergent subtypes H3N2, H1N1, and H5N1. The immunogen is based on the binding site of the recently described nAb 12D1, which neutralizes H3 subtype viruses, demonstrates protective activity in vivo, and, in contrast to a majority of described nAbs, appears to bind to residues within a single α-helical portion of the HA2 protein. Our data further demonstrate that the specific design of our immunogen is integral in the induction of broadly active anti-hemagglutinin antibodies. These results provide proof of concept for an HA2-based influenza vaccine that could diminish the threat of pandemic influenza disease and generally reduce the significance of influenza viruses as human pathogens.

    View details for DOI 10.1073/pnas.1013387107

    View details for Web of Science ID 000283749000045

    View details for PubMedID 20956293

  • A Nine-Segment Influenza A Virus Carrying Subtype H1 and H3 Hemagglutinins JOURNAL OF VIROLOGY Gao, Q., Lowen, A. C., Wang, T. T., Palese, P. 2010; 84 (16): 8062-8071

    Abstract

    Influenza virus genomic RNAs possess segment-specific packaging signals that include both noncoding regions (NCRs) and adjacent terminal coding region sequences. Using reverse genetics, an A/Puerto Rico/8/34 (A/PR/8/34) virus was rescued that contained a modified PB1 gene such that the PB1 packaging sequences were exchanged for those of the neuraminidase (NA) gene segment. To accomplish this, the PB1 open reading frame, in which the terminal packaging signals were inactivated by serial synonymous mutations, was flanked by the NA segment-specific packaging sequences including the NCRs and the coding region packaging signals. Next, the ATGs located on the 3' end of the NA packaging sequences of the resulting PB1 chimeric segment were mutated to allow for correct translation of the full-length PB1 protein. The virus containing this chimeric PB1 segment was viable and able to stably carry a ninth, green fluorescent protein (GFP), segment flanked by PB1 packaging signals. Utilizing this method, we successfully generated an influenza virus that contained the genes coding for both the H1 hemagglutinin (HA) from A/PR/8/34 and the H3 HA from A/Hong Kong/1/68 (A/HK/1/68); both subtypes of HA protein were also incorporated into the viral envelope. Immunization of mice with this recombinant virus conferred complete protection from lethal challenge with recombinant A/PR/8/34 virus and with X31 virus that expresses the A/HK/1/68 HA and NA. Using the described methodology, we show that a ninth segment can also be incorporated by manipulation of the PB2 or PA segment-specific packaging signals. This approach offers a means of generating a bivalent influenza virus vaccine.

    View details for DOI 10.1128/JVI.00722-10

    View details for Web of Science ID 000279983200014

    View details for PubMedID 20519387

  • PB1-F2 Expression by the 2009 Pandemic H1N1 Influenza Virus Has Minimal Impact on Virulence in Animal Models JOURNAL OF VIROLOGY Hai, R., Schmolke, M., Varga, Z. T., Manicassamy, B., Wang, T. T., Belser, J. A., Pearce, M. B., Garcia-Sastre, A., Tumpey, T. M., Palese, P. 2010; 84 (9): 4442-4450

    Abstract

    Unlike previous pandemic viruses, the 2009 H1N1 pandemic influenza virus does not code for the virulence factor PB1-F2. The genome of the 2009 H1N1 virus contains three stop codons preventing PB1-F2 expression; however, PB1-F2 production could occur following genetic mutation or reassortment. Thus, it is of great interest to understand the impact that expression of the PB1-F2 protein might have in the context of the 2009 pandemic influenza virus, A/California/04/2009 (Cal/09). We have addressed this question by generating two Cal/09 viruses with productive PB1-F2 open reading frames containing either an asparagine at position 66 of PB1-F2 (66N) or a serine at position 66 (66S): this N66S change has previously been shown to be associated with increased virulence in mice. We used these viruses to investigate the effect on virulence conferred by expression of the 66N or the 66S PB1-F2 protein in both in vitro and in vivo systems. Our results show enhanced replication of the 66S virus in A549 cells, while studies of BALB/c and DBA/2 mice and ferrets revealed no significant differences in symptoms of infection with wild-type Cal/09 versus the 66N or 66S virus variant. Also, coinfection of mice with Streptococcus pneumoniae and the different viruses (recombinant wild-type [rWT] Cal/09 and the 66N and 66S viruses) did not result in significant differences in mortality. Mice infected with either PB1-F2-expressing virus did demonstrate altered protein levels of proinflammatory cytokines; differences were observed to be greater in infection caused by the 66S virus. In summary, our study demonstrates that PB1-F2 expression by the Cal/09 virus modulates the immune response to infection while having a minimal effect on virus virulence in two mammalian models.

    View details for DOI 10.1128/JVI.02717-09

    View details for Web of Science ID 000276358000030

    View details for PubMedID 20181699

  • Influenza Virus Vaccine Based on the Conserved Hemagglutinin Stalk Domain MBIO Steel, J., Lowen, A. C., Wang, T. T., Yondola, M., Gao, Q., Haye, K., Garcia-Sastre, A., Palese, P. 2010; 1 (1)

    Abstract

    Although highly effective in the general population when well matched to circulating influenza virus strains, current influenza vaccines are limited in their utility due to the narrow breadth of protection they provide. The strain specificity of vaccines presently in use mirrors the exquisite specificity of the neutralizing antibodies that they induce, that is, antibodies which bind to the highly variable globular head domain of hemagglutinin (HA). Herein, we describe the construction of a novel immunogen comprising the conserved influenza HA stalk domain and lacking the globular head. Vaccination of mice with this headless HA construct elicited immune sera with broader reactivity than those obtained from mice immunized with a full-length HA. Furthermore, the headless HA vaccine provided full protection against death and partial protection against disease following lethal viral challenge. Our results suggest that the response induced by headless HA vaccines is sufficiently potent to warrant their further development toward a universal influenza virus vaccine.

    View details for DOI 10.1128/mBio.00018-10

    View details for Web of Science ID 000284716600002

    View details for PubMedID 20689752

  • Broadly Protective Monoclonal Antibodies against H3 Influenza Viruses following Sequential Immunization with Different Hemagglutinins PLOS PATHOGENS Wang, T. T., Tan, G. S., Hai, R., Pica, N., Petersen, E., Moran, T. M., Palese, P. 2010; 6 (2)

    Abstract

    As targets of adaptive immunity, influenza viruses are characterized by the fluidity with which they respond to the selective pressure applied by neutralizing antibodies. This mutability of structural determinants of protective immunity is the obstacle in developing universal influenza vaccines. Towards the development of such vaccines and other immune therapies, our studies are designed to identify regions of influenza viruses that are conserved and that mediate virus neutralization. We have specifically focused on viruses of the H3N2 subtype, which have persisted as a principal source of influenza-related morbidity and mortality in humans since the pandemic of 1968. Three monoclonal antibodies have been identified that are broadly-neutralizing against H3 influenza viruses spanning 40 years. The antibodies react with the hemagglutinin glycoprotein and appear to bind in regions that are refractory to the structural variation required for viral escape from neutralization. The antibodies demonstrate therapeutic efficacy in mice against H3N2 virus infection and have potential for use in the treatment of human influenza disease. By mapping the binding region of one antibody, 12D1, we have identified a continuous region of the hemagglutinin that may act as an immunogen to elicit broadly protective immunity to H3 viruses. The anti-H3 monoclonal antibodies were identified after immunization of mice with the hemagglutinin of four different viruses (A/Hong Kong/1/1968, A/Alabama/1/1981, A/Beijing/47/1992, A/Wyoming/3/2003). This immunization schedule was designed to boost B cells specific for conserved regions of the hemagglutinin from distinct antigenic clusters. Importantly, our antibodies are of naturally occurring specificity rather than selected from cloned libraries, demonstrating that broad-spectrum humoral immunity to influenza viruses can be elicited in vivo.

    View details for DOI 10.1371/journal.ppat.1000796

    View details for Web of Science ID 000275295900042

    View details for PubMedID 20195520

  • Unraveling the Mystery of Swine Influenza Virus CELL Wang, T. T., Palese, P. 2009; 137 (6): 983-985

    Abstract

    Influenza virus outbreaks occur with regularity, but the severity of outbreaks is not consistent. The recent flu epidemic caused by an H1N1 swine influenza virus presents an opportunity to examine what is known about virulence factors and the spread of infection to better prepare for major influenza outbreaks in the future.

    View details for DOI 10.1016/j.cell.2009.05.032

    View details for Web of Science ID 000266916400006

    View details for PubMedID 19524497

  • Universal epitopes of influenza virus hemagglutinins? NATURE STRUCTURAL & MOLECULAR BIOLOGY Wang, T. T., Palese, P. 2009; 16 (3): 233-234

    View details for DOI 10.1038/nsmb.1574

    View details for Web of Science ID 000263906000003

    View details for PubMedID 19234464

  • The capsule of Bacillus anthracis behaves as a thymus-independent type 2 antigen INFECTION AND IMMUNITY Wang, T. T., Lucas, A. H. 2004; 72 (9): 5460-5463

    Abstract

    Bacillus anthracis elaborates a homopolymeric capsule composed of gamma-D-glutamic acid residues. Mice were immunized with formalin-fixed encapsulated B. anthracis bacilli, and the serum antibody response to a gamma-D-glutamyl capsular epitope was measured. Antiglutamyl antibodies were elicited in athymic BALB/c Nu/Nu, BALB/c Nu/+, and CBA/J mice but not in CBA/N xid mice. These response patterns define the capsule of B. anthracis as a thymus-independent type 2 antigen.

    View details for DOI 10.1128/IAI.72.9.5460-5463.2004

    View details for Web of Science ID 000223580400062

    View details for PubMedID 15322045

  • Induction of opsonic antibodies to the gamma-D-glutamic acid capsule of Bacillus anthracis by immunization with a synthetic peptide-carrier protein conjugate FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY Wang, T. T., Fellows, P. F., Leighton, T. J., Lucas, A. H. 2004; 40 (3): 231-237

    Abstract

    The capsule of Bacillus anthracis, a polymer of gamma-D-glutamic acid, functions as a virulence determinant and is a poor immunogen. In this study we show that antibodies reactive with the B. anthracis capsule can be elicited in mice by immunization with a conjugate consisting of a synthetic gamma-D-glutamic acid nonamer peptide (gamma-D-glu9) covalently coupled to keyhole limpet hemocyanin. The serum response to gamma-D-glu9 was comprised primarily of IgG antibodies that recognized an epitope requiring a minimum of four gamma-linked D-glutamic acid residues. Antibodies to (gamma-D-glu9) bound to the surface of encapsulated B. anthracis cells and mediated opsonophagoctosis. These findings suggest that anti-capsular antibodies could mediate the clearance of vegetative B. anthracis cells in vivo. Thus, inclusion of an immunogenic capsular component as well as protective antigen in new anthrax vaccines would generate immune responses targeting both the bacteremic and toxigenic aspects of anthrax infection and thus may increase protective efficacy.

    View details for DOI 10.1016/S0928-8244(03)00366-3

    View details for Web of Science ID 000220582800008

    View details for PubMedID 15039099