Network


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

Hotspot


Dive into the research topics where Lingshu Wang is active.

Publication


Featured researches published by Lingshu Wang.


Nature | 2013

Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies.

Masaru Kanekiyo; Chih Jen Wei; Hadi M. Yassine; Patrick McTamney; Jeffrey C. Boyington; James R. R. Whittle; Srinivas S. Rao; Wing Pui Kong; Lingshu Wang; Gary J. Nabel

Influenza viruses pose a significant threat to the public and are a burden on global health systems. Each year, influenza vaccines must be rapidly produced to match circulating viruses, a process constrained by dated technology and vulnerable to unexpected strains emerging from humans and animal reservoirs. Here we use knowledge of protein structure to design self-assembling nanoparticles that elicit broader and more potent immunity than traditional influenza vaccines. The viral haemagglutinin was genetically fused to ferritin, a protein that naturally forms nanoparticles composed of 24 identical polypeptides. Haemagglutinin was inserted at the interface of adjacent subunits so that it spontaneously assembled and generated eight trimeric viral spikes on its surface. Immunization with this influenza nanoparticle vaccine elicited haemagglutination inhibition antibody titres more than tenfold higher than those from the licensed inactivated vaccine. Furthermore, it elicited neutralizing antibodies to two highly conserved vulnerable haemagglutinin structures that are targets of universal vaccines: the stem and the receptor binding site on the head. Antibodies elicited by a 1999 haemagglutinin–nanoparticle vaccine neutralized H1N1 viruses from 1934 to 2007 and protected ferrets from an unmatched 2007 H1N1 virus challenge. This structure-based, self-assembling synthetic nanoparticle vaccine improves the potency and breadth of influenza virus immunity, and it provides a foundation for building broader vaccine protection against emerging influenza viruses and other pathogens.


Nature Medicine | 2014

Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge

Daphne Stanley; Anna N. Honko; Clement Asiedu; John C. Trefry; Annie W. Lau-Kilby; Joshua C. Johnson; Lisa Hensley; Virginia Ammendola; Adele Abbate; Fabiana Grazioli; Kathryn E. Foulds; Cheng Cheng; Lingshu Wang; Mitzi Donaldson; Stefano Colloca; Antonella Folgori; Mario Roederer; Gary J. Nabel; John R. Mascola; Alfredo Nicosia; Riccardo Cortese; Richard A. Koup; Nancy J. Sullivan

Ebolavirus disease causes high mortality, and the current outbreak has spread unabated through West Africa. Human adenovirus type 5 vectors (rAd5) encoding ebolavirus glycoprotein (GP) generate protective immunity against acute lethal Zaire ebolavirus (EBOV) challenge in macaques, but fail to protect animals immune to Ad5, suggesting natural Ad5 exposure may limit vaccine efficacy in humans. Here we show that a chimpanzee-derived replication-defective adenovirus (ChAd) vaccine also rapidly induced uniform protection against acute lethal EBOV challenge in macaques. Because protection waned over several months, we boosted ChAd3 with modified vaccinia Ankara (MVA) and generated, for the first time, durable protection against lethal EBOV challenge.


Nature Medicine | 2015

Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection

Hadi M. Yassine; Jeffrey C. Boyington; Patrick McTamney; Chih Jen Wei; Masaru Kanekiyo; Wing Pui Kong; John R. Gallagher; Lingshu Wang; Yi Zhang; M. Gordon Joyce; Daniel Lingwood; Syed M. Moin; Hanne Andersen; Yoshinobu Okuno; Srinivas S. Rao; Audray K. Harris; Peter D. Kwong; John R. Mascola; Gary J. Nabel; Barney S. Graham

The antibody response to influenza is primarily focused on the head region of the hemagglutinin (HA) glycoprotein, which in turn undergoes antigenic drift, thus necessitating annual updates of influenza vaccines. In contrast, the immunogenically subdominant stem region of HA is highly conserved and recognized by antibodies capable of binding multiple HA subtypes. Here we report the structure-based development of an H1 HA stem–only immunogen that confers heterosubtypic protection in mice and ferrets. Six iterative cycles of structure-based design (Gen1–Gen6) yielded successive H1 HA stabilized-stem (HA–SS) immunogens that lack the immunodominant head domain. Antigenic characterization, determination of two HA–SS crystal structures in complex with stem-specific monoclonal antibodies and cryo-electron microscopy analysis of HA–SS on ferritin nanoparticles (H1–SS–np) confirmed the preservation of key structural elements. Vaccination of mice and ferrets with H1–SS–np elicited broadly cross-reactive antibodies that completely protected mice and partially protected ferrets against lethal heterosubtypic H5N1 influenza virus challenge despite the absence of detectable H5N1 neutralizing activity in vitro. Passive transfer of immunoglobulin from H1–SS–np–immunized mice to naive mice conferred protection against H5N1 challenge, indicating that vaccine-elicited HA stem–specific antibodies can protect against diverse group 1 influenza strains.


Nature Medicine | 2016

Molecular-level analysis of the serum antibody repertoire in young adults before and after seasonal influenza vaccination

Jiwon Lee; Daniel R. Boutz; Veronika Chromikova; M. Gordon Joyce; Christopher Vollmers; Kwanyee Leung; Andrew P. Horton; Brandon J. DeKosky; Chang-Han Lee; Jason J. Lavinder; Ellen M. Murrin; Constantine Chrysostomou; Kam Hon Hoi; Yaroslav Tsybovsky; Paul V. Thomas; Aliaksandr Druz; Baoshan Zhang; Yi Zhang; Lingshu Wang; Wing-Pui Kong; Daechan Park; Lyubov Popova; Cornelia L. Dekker; Mark M. Davis; Chalise E. Carter; Ted M. Ross; Andrew D. Ellington; Patrick C. Wilson; Edward M. Marcotte; John R. Mascola

Molecular understanding of serological immunity to influenza has been confounded by the complexity of the polyclonal antibody response in humans. Here we used high-resolution proteomics analysis of immunoglobulin (referred to as Ig-seq) coupled with high-throughput sequencing of transcripts encoding B cell receptors (BCR-seq) to quantitatively determine the antibody repertoire at the individual clonotype level in the sera of young adults before and after vaccination with trivalent seasonal influenza vaccine. The serum repertoire comprised between 40 and 147 clonotypes that were specific to each of the three monovalent components of the trivalent influenza vaccine, with boosted pre-existing clonotypes accounting for ∼60% of the response. An unexpectedly high fraction of serum antibodies recognized both the H1 and H3 monovalent vaccines. Recombinant versions of these H1 + H3 cross-reactive antibodies showed broad binding to hemagglutinins (HAs) from previously circulating virus strains; several of these antibodies, which were prevalent in the serum of multiple donors, recognized the same conserved epitope in the HA head domain. Although the HA-head-specific H1 + H3 antibodies did not show neutralization activity in vitro, they protected mice against infection with the H1N1 and H3N2 virus strains when administered before or after challenge. Collectively, our data reveal unanticipated insights regarding the serological response to influenza vaccination and raise questions about the added benefits of using a quadrivalent vaccine instead of a trivalent vaccine.


Nature Communications | 2015

Evaluation of candidate vaccine approaches for MERS-CoV

Lingshu Wang; Wei Shi; M. Gordon Joyce; Kayvon Modjarrad; Yi Zhang; Kwanyee Leung; Christopher R. Lees; Tongqing Zhou; Hadi M. Yassine; Masaru Kanekiyo; Zhi Yong Yang; Xuejun Chen; Michelle M. Becker; Megan Culler Freeman; Leatrice Vogel; Joshua C. Johnson; Gene G. Olinger; John Paul Todd; Ulas Bagci; Jeffrey Solomon; Daniel J. Mollura; Lisa E. Hensley; Peter B. Jahrling; Mark R. Denison; Srinivas S. Rao; Kanta Subbarao; Peter D. Kwong; John R. Mascola; Wing Pui Kong; Barney S. Graham

The emergence of Middle East respiratory syndrome coronavirus (MERS-CoV) as a cause of severe respiratory disease highlights the need for effective approaches to CoV vaccine development. Efforts focused solely on the receptor-binding domain (RBD) of the viral Spike (S) glycoprotein may not optimize neutralizing antibody (NAb) responses. Here we show that immunogens based on full-length S DNA and S1 subunit protein elicit robust serum-neutralizing activity against several MERS-CoV strains in mice and non-human primates. Serological analysis and isolation of murine monoclonal antibodies revealed that immunization elicits NAbs to RBD and, non-RBD portions of S1 and S2 subunit. Multiple neutralization mechanisms were demonstrated by solving the atomic structure of a NAb-RBD complex, through sequencing of neutralization escape viruses and by constructing MERS-CoV S variants for serological assays. Immunization of rhesus macaques confers protection against MERS-CoV-induced radiographic pneumonia, as assessed using computerized tomography, supporting this strategy as a promising approach for MERS-CoV vaccine development. Supplementary information The online version of this article (doi:10.1038/ncomms8712) contains supplementary material, which is available to authorized users.


Cell | 2016

Induction of HIV Neutralizing Antibody Lineages in Mice with Diverse Precursor Repertoires

Ming Tian; Cheng Cheng; Xuejun Chen; Hongying Duan; Hwei-Ling Cheng; Mai Dao; Zizhang Sheng; Michael Kimble; Lingshu Wang; Sherry Lin; Stephen D. Schmidt; Zhou Du; M. Gordon Joyce; Yiwei Chen; Brandon J. DeKosky; Yimin Chen; Erica Normandin; Elizabeth Cantor; Rita E. Chen; Nicole A. Doria-Rose; Yi Zhang; Wei Shi; Wing-Pui Kong; Misook Choe; Amy R. Henry; Farida Laboune; Ivelin S. Georgiev; Pei-Yi Huang; Suvi Jain; Andrew T. McGuire

The design of immunogens that elicit broadly reactive neutralizing antibodies (bnAbs) has been a major obstacle to HIV-1 vaccine development. One approach to assess potential immunogens is to use mice expressing precursors of human bnAbs as vaccination models. The bnAbs of the VRC01-class derive from the IGHV1-2 immunoglobulin heavy chain and neutralize a wide spectrum of HIV-1 strains via targeting the CD4 binding site of the envelope glycoprotein gp120. We now describe a mouse vaccination model that allows a germline human IGHV1-2(∗)02 segment to undergo normal V(D)J recombination and, thereby, leads to the generation of peripheral B cells that express a highly diverse repertoire of VRC01-related receptors. When sequentially immunized with modified gp120 glycoproteins designed to engage VRC01 germline and intermediate antibodies, IGHV1-2(∗)02-rearranging mice, which also express a VRC01-antibody precursor light chain, can support the affinity maturation of VRC01 precursor antibodies into HIV-neutralizing antibody lineages.


Cell | 2016

Vaccine-Induced Antibodies that Neutralize Group 1 and Group 2 Influenza A Viruses.

M. Gordon Joyce; Adam K. Wheatley; Paul V. Thomas; Gwo-Yu Chuang; Cinque Soto; Robert T. Bailer; Aliaksandr Druz; Ivelin S. Georgiev; Rebecca A. Gillespie; Masaru Kanekiyo; Wing-Pui Kong; Kwanyee Leung; Sandeep N. Narpala; Madhu Prabhakaran; Eun Sung Yang; Baoshan Zhang; Yi Zhang; Mangaiarkarasi Asokan; Jeffrey C. Boyington; Tatsiana Bylund; Sam Darko; Christopher R. Lees; Amy Ransier; Chen-Hsiang Shen; Lingshu Wang; James R. R. Whittle; Xueling Wu; Hadi M. Yassine; Celia Santos; Yumiko Matsuoka

Antibodies capable of neutralizing divergent influenza A viruses could form the basis of a universal vaccine. Here, from subjects enrolled in an H5N1 DNA/MIV-prime-boost influenza vaccine trial, we sorted hemagglutinin cross-reactive memory B cells and identified three antibody classes, each capable of neutralizing diverse subtypes of group 1 and group 2 influenza A viruses. Co-crystal structures with hemagglutinin revealed that each class utilized characteristic germline genes and convergent sequence motifs to recognize overlapping epitopes in the hemagglutinin stem. All six analyzed subjects had sequences from at least one multidonor class, and-in half the subjects-multidonor-class sequences were recovered from >40% of cross-reactive B cells. By contrast, these multidonor-class sequences were rare in published antibody datasets. Vaccination with a divergent hemagglutinin can thus increase the frequency of B cells encoding broad influenza A-neutralizing antibodies. We propose the sequence signature-quantified prevalence of these B cells as a metric to guide universal influenza A immunization strategies.


Journal of Immunology | 2013

Comparative Analysis of the Magnitude, Quality, Phenotype, and Protective Capacity of Simian Immunodeficiency Virus Gag-Specific CD8+ T Cells following Human-, Simian-, and Chimpanzee-Derived Recombinant Adenoviral Vector Immunization

Kylie M. Quinn; Andreia Costa; Ayako Yamamoto; Dana Berry; Ross W. B. Lindsay; Patricia A. Darrah; Lingshu Wang; Cheng Cheng; Wing Pui Kong; Jason G. D. Gall; Alfredo Nicosia; Antonella Folgori; Stefano Colloca; Riccardo Cortese; Emma Gostick; David A. Price; Carmen Elena Gómez; Mariano Esteban; Linda S. Wyatt; Bernard Moss; Cecilia Morgan; Mario Roederer; Robert T. Bailer; Gary J. Nabel; Richard A. Koup; Robert A. Seder

Recombinant adenoviral vectors (rAds) are the most potent recombinant vaccines for eliciting CD8+ T cell–mediated immunity in humans; however, prior exposure from natural adenoviral infection can decrease such responses. In this study we show low seroreactivity in humans against simian- (sAd11, sAd16) or chimpanzee-derived (chAd3, chAd63) compared with human-derived (rAd5, rAd28, rAd35) vectors across multiple geographic regions. We then compared the magnitude, quality, phenotype, and protective capacity of CD8+ T cell responses in mice vaccinated with rAds encoding SIV Gag. Using a dose range (1 × 107–109 particle units), we defined a hierarchy among rAd vectors based on the magnitude and protective capacity of CD8+ T cell responses, from most to least, as: rAd5 and chAd3, rAd28 and sAd11, chAd63, sAd16, and rAd35. Selection of rAd vector or dose could modulate the proportion and/or frequency of IFN-γ+TNF-α+IL-2+ and KLRG1+CD127−CD8+ T cells, but strikingly ∼30–80% of memory CD8+ T cells coexpressed CD127 and KLRG1. To further optimize CD8+ T cell responses, we assessed rAds as part of prime-boost regimens. Mice primed with rAds and boosted with NYVAC generated Gag-specific responses that approached ∼60% of total CD8+ T cells at peak. Alternatively, priming with DNA or rAd28 and boosting with rAd5 or chAd3 induced robust and equivalent CD8+ T cell responses compared with prime or boost alone. Collectively, these data provide the immunologic basis for using specific rAd vectors alone or as part of prime-boost regimens to induce CD8+ T cells for rapid effector function or robust long-term memory, respectively.


Journal of Virology | 2015

Broadly Neutralizing Human Immunodeficiency Virus Type 1 Antibody Gene Transfer Protects Nonhuman Primates from Mucosal Simian-Human Immunodeficiency Virus Infection

Kevin O. Saunders; Lingshu Wang; M. Gordon Joyce; Zhi Yong Yang; Alejandro B. Balazs; Cheng Cheng; Sung Youl Ko; Wing Pui Kong; Rebecca S. Rudicell; Ivelin S. Georgiev; Lijie Duan; Kathryn E. Foulds; Mitzi Donaldson; Ling Xu; Stephen D. Schmidt; John Paul Todd; David Baltimore; Mario Roederer; Ashley T. Haase; Peter D. Kwong; Srinivas S. Rao; John R. Mascola; Gary J. Nabel

ABSTRACT Broadly neutralizing antibodies (bnAbs) can prevent lentiviral infection in nonhuman primates and may slow the spread of human immunodeficiency virus type 1 (HIV-1). Although protection by passive transfer of human bnAbs has been demonstrated in monkeys, durable expression is essential for its broader use in humans. Gene-based expression of bnAbs provides a potential solution to this problem, although immune responses to the viral vector or to the antibody may limit its durability and efficacy. Here, we delivered an adeno-associated viral vector encoding a simianized form of a CD4bs bnAb, VRC07, and evaluated its immunogenicity and protective efficacy. The expressed antibody circulated in macaques for 16 weeks at levels up to 66 μg/ml, although immune suppression with cyclosporine (CsA) was needed to sustain expression. Gene-delivered simian VRC07 protected against simian-human immunodeficiency virus (SHIV) infection in monkeys 5.5 weeks after treatment. Gene transfer of an anti-HIV antibody can therefore protect against infection by viruses that cause AIDS in primates when the host immune responses are controlled. IMPORTANCE Sustained interventions that can prevent HIV-1 infection are needed to halt the spread of the HIV-1 pandemic. The protective capacity of anti-HIV antibody gene therapy has been established in mouse models of HIV-1 infection but has not been established for primates. We show here a proof-of-concept that gene transfer of anti-HIV antibody genes can protect against infection by viruses that cause AIDS in primates when host immune responses are controlled.


Nature Communications | 2015

Junctional and allele-specific residues are critical for MERS-CoV neutralization by an exceptionally potent germline-like antibody.

Tianlei Ying; Ponraj Prabakaran; Lanying Du; Wei Shi; Yang Feng; Yanping Wang; Lingshu Wang; Wei Li; Shibo Jiang; Dimiter S. Dimitrov; Tongqing Zhou

The MERS-CoV is an emerging virus, which already infected more than 1,300 humans with high (∼36%) mortality. Here, we show that m336, an exceptionally potent human anti-MERS-CoV antibody, is almost germline with only one somatic mutation in the heavy chain. The structure of Fab m336 in complex with the MERS-CoV receptor-binding domain reveals that its IGHV1-69-derived heavy chain provides more than 85% binding surface and that its epitope almost completely overlaps with the receptor-binding site. Analysis of antibodies from 69 healthy humans suggests an important role of the V(D)J recombination-generated junctional and allele-specific residues for achieving high affinity of binding at such low levels of somatic hypermutation. Our results also have important implications for development of vaccine immunogens based on the newly identified m336 epitope as well as for elucidation of mechanisms of neutralization by m336-like antibodies and their elicitation in vivo. Supplementary information The online version of this article (doi:10.1038/ncomms9223) contains supplementary material, which is available to authorized users.

Collaboration


Dive into the Lingshu Wang's collaboration.

Top Co-Authors

Avatar

Wing-Pui Kong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cheng Cheng

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Mascola

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Masaru Kanekiyo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wei Shi

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

M. Gordon Joyce

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wing Pui Kong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yi Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Barney S. Graham

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge