Network


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

Hotspot


Dive into the research topics where Yinghui Rong is active.

Publication


Featured researches published by Yinghui Rong.


Journal of Biological Chemistry | 2017

Structural Analysis of Single Domain Antibodies Bound to a Second Neutralizing Hot Spot on Ricin Toxin's Enzymatic Subunit.

Michael J. Rudolph; David J. Vance; Michael S. Cassidy; Yinghui Rong; Nicholas J. Mantis

Ricin toxin is a heterodimer consisting of RTA, a ribosome-inactivating protein, and RTB, a lectin that facilitates receptor-mediated uptake into mammalian cells. In previous studies, we demonstrated that toxin-neutralizing antibodies target four spatially distinct hot spots on RTA, which we refer to as epitope clusters I–IV. In this report, we identified and characterized three single domain camelid antibodies (VHH) against cluster II. One of these VHHs, V5E1, ranks as one of the most potent ricin-neutralizing antibodies described to date. We solved the X-ray crystal structures of each of the three VHHs (E1, V1C7, and V5E1) in complex with RTA. V5E1 buries a total of 1,133 Å2 of surface area on RTA and makes primary contacts with α-helix A (residues 18–32), α-helix F (182–194), as well as the F-G loop. V5E1, by virtue of complementarity determining region 3 (CDR3), may also engage with RTB and potentially interfere with the high affinity galactose-recognition element that plays a critical role in toxin attachment to cell surfaces and intracellular trafficking. The two other VHHs, E1 and V1C7, bind epitopes adjacent to V5E1 but display only weak toxin neutralizing activity, thereby providing structural insights into specific residues within cluster II that may be critical contact points for toxin inactivation.


Vaccine | 2015

Combination of two candidate subunit vaccine antigens elicits protective immunity to ricin and anthrax toxin in mice

David J. Vance; Yinghui Rong; Robert N. Brey; Nicholas J. Mantis

In an effort to develop combination vaccines for biodefense, we evaluated a ricin subunit antigen, RiVax, given in conjunction with an anthrax protective antigen, DNI. The combination led to high endpoint titer antibody response, neutralizing antibodies, and protective immunity against ricin and anthrax lethal toxin. This is a natural combination vaccine, since both antigens are recombinant subunit proteins that would be given to the same target population.


Proteins | 2016

Structural analysis of nested neutralizing and non-neutralizing B cell epitopes on ricin toxin's enzymatic subunit.

Michael J. Rudolph; David J. Vance; Michael S. Cassidy; Yinghui Rong; Charles B. Shoemaker; Nicholas J. Mantis

In this report, we describe the X‐ray crystal structures of two single domain camelid antibodies (VHH), F5 and F8, each in complex with ricin toxins enzymatic subunit (RTA). F5 has potent toxin‐neutralizing activity, while F8 has weak neutralizing activity. F5 buried a total of 1760 Å2 in complex with RTA and made contact with three prominent secondary structural elements: α‐helix B (Residues 98–106), β‐strand h (Residues 113–117), and the C‐terminus of α‐helix D (Residues 154–156). F8 buried 1103 Å2 in complex with RTA that was centered primarily on β‐strand h. As such, the structural epitope of F8 is essentially nested within that of F5. All three of the F5 complementarity determining regions CDRs were involved in RTA contact, whereas F8 interactions were almost entirely mediated by CDR3, which essentially formed a seventh β‐strand within RTAs centrally located β‐sheet. A comparison of the two structures reported here to several previously reported (RTA‐VHH) structures identifies putative contact sites on RTA, particularly α‐helix B, associated with potent toxin‐neutralizing activity. This information has implications for rational design of RTA‐based subunit vaccines for biodefense. Proteins 2016; 84:1162–1172.


Proteins | 2017

Using Homology Modeling to Interrogate Binding Affinity in Neutralization of Ricin Toxin by a Family of Single Domain Antibodies

Andrea Bazzoli; David J. Vance; Michael J. Rudolph; Yinghui Rong; Siva Krishna Angalakurthi; Ronald T. Toth; C. Russell Middaugh; David B. Volkin; David D. Weis; John Karanicolas; Nicholas J. Mantis

In this report we investigated, within a group of closely related single domain camelid antibodies (VHHs), the relationship between binding affinity and neutralizing activity as it pertains to ricin, a fast‐acting toxin and biothreat agent. The V1C7‐like VHHs (V1C7, V2B9, V2E8, and V5C1) are similar in amino acid sequence, but differ in their binding affinities and toxin‐neutralizing activities. Using the X‐ray crystal structure of V1C7 in complex with ricins enzymatic subunit (RTA) as a template, Rosetta‐based homology modeling coupled with energetic decomposition led us to predict that a single pairwise interaction between Arg29 on V5C1 and Glu67 on RTA was responsible for the difference in ricin toxin binding affinity between V1C7, a weak neutralizer, and V5C1, a moderate neutralizer. This prediction was borne out experimentally: substitution of Arg for Gly at position 29 enhanced V1C7s binding affinity for ricin, whereas the reverse (ie, Gly for Arg at position 29) diminished V5C1s binding affinity by >10 fold. As expected, the V5C1R29G mutant was largely devoid of toxin‐neutralizing activity (TNA). However, the TNA of the V1C7G29R mutant was not correspondingly improved, indicating that in the V1C7 family binding affinity alone does not account for differences in antibody function. V1C7 and V5C1, as well as their respective point mutants, recognized indistinguishable epitopes on RTA, at least at the level of sensitivity afforded by hydrogen‐deuterium mass spectrometry. The results of this study have implications for engineering therapeutic antibodies because they demonstrate that even subtle differences in epitope specificity can account for important differences in antibody function.


Clinical and Vaccine Immunology | 2017

High-Resolution Epitope Positioning of a Large Collection of Neutralizing and Non-Neutralizing Single Domain Antibodies on Ricin Toxin's Enzymatic and Binding Subunits

David J. Vance; Jacqueline M. Tremblay; Yinghui Rong; Siva Krishna Angalakurthi; David B. Volkin; C. Russell Middaugh; David D. Weis; Charles B. Shoemaker; Nicholas J. Mantis

ABSTRACT We previously produced a heavy-chain-only antibody (Ab) VH domain (VHH)-displayed phage library from two alpacas that had been immunized with ricin toxoid and nontoxic mixtures of the enzymatic ricin toxin A subunit (RTA) and binding ricin toxin B subunit (RTB) (D. J. Vance, J. M. Tremblay, N. J. Mantis, and C. B. Shoemaker, J Biol Chem 288:36538–36547, 2013, https://doi.org/10.1074/jbc.M113.519207 ). Initial and subsequent screens of that library by direct enzyme-linked immunosorbent assay (ELISA) yielded more than two dozen unique RTA- and RTB-specific VHHs, including 10 whose structures were subsequently solved in complex with RTA. To generate a more complete antigenic map of ricin toxin and to define the epitopes associated with toxin-neutralizing activity, we subjected the VHH-displayed phage library to additional “pannings” on both receptor-bound ricin and antibody-captured ricin. We now report the full-length DNA sequences, binding affinities, and neutralizing activities of 68 unique VHHs: 31 against RTA, 33 against RTB, and 4 against ricin holotoxin. Epitope positioning was achieved through cross-competition ELISAs performed with a panel of monoclonal antibodies (MAbs) and verified, in some instances, with hydrogen-deuterium exchange mass spectrometry. The 68 VHHs grouped into more than 20 different competition bins. The RTA-specific VHHs with strong toxin-neutralizing activities were confined to bins that overlapped two previously identified neutralizing hot spots, termed clusters I and II. The four RTB-specific VHHs with potent toxin-neutralizing activity grouped within three adjacent bins situated at the RTA-RTB interface near cluster II. These results provide important insights into epitope interrelationships on the surface of ricin and delineate regions of vulnerability that can be exploited for the purpose of vaccine and therapeutic development.


Clinical and Vaccine Immunology | 2015

Comparative Adjuvant Effects of Type II Heat-Labile Enterotoxins in Combination with Two Different Candidate Ricin Toxin Vaccine Antigens

David J. Vance; Christopher J. Greene; Yinghui Rong; Lorrie Mandell; Terry D. Connell; Nicholas J. Mantis

ABSTRACT Type II heat-labile enterotoxins (HLTs) constitute a promising set of adjuvants that have been shown to enhance humoral and cellular immune responses when coadministered with an array of different proteins, including several pathogen-associated antigens. However, the adjuvant activities of the four best-studied HLTs, LT-IIa, LT-IIb, LT-IIbT13I, and LT-IIc, have never been compared side by side. We therefore conducted immunization studies in which LT-IIa, LT-IIb, LT-IIbT13I, and LT-IIc were coadministered by the intradermal route to mice with two clinically relevant protein subunit vaccine antigens derived from the enzymatic A subunit (RTA) of ricin toxin, RiVax and RVEc. The HLTs were tested with low and high doses of antigen and were assessed for their abilities to stimulate antigen-specific serum IgG titers, ricin toxin-neutralizing activity (TNA), and protective immunity. We found that all four HLTs tested were effective adjuvants when coadministered with RiVax or RVEc. LT-IIa was of particular interest because as little as 0.03 μg when coadministered with RiVax or RVEc proved effective at augmenting ricin toxin-specific serum antibody titers with nominal evidence of local inflammation. Collectively, these results justify the need for further studies into the mechanism(s) underlying LT-IIa adjuvant activity, with the long-term goal of evaluating LT-IIas activity in humans.


PLOS ONE | 2017

Spatial location of neutralizing and non-neutralizing B cell epitopes on domain 1 of ricin toxin’s binding subunit

Yinghui Rong; Greta Van Slyke; David J. Vance; Jennifer Westfall; Dylan J. Ehrbar; Nicholas J. Mantis

Ricin toxin’s binding subunit (RTB) is a galactose-/N-acetylgalactosamine (Gal/GalNac)-specific lectin that mediates uptake and intracellular trafficking of ricin within mammalian cells. Structurally, RTB consists of two globular domains, each divided into three homologous sub-domains (α, β, γ). In this report, we describe five new murine IgG monoclonal antibodies (mAbs) against RTB: MH3, 8A1, 8B3, LF1, and LC5. The mAbs have similar binding affinities (KD) for ricin holotoxin, but displayed a wide range of in vitro toxin-neutralizing activities. Competition ELISAs indicate that the two most potent toxin-neutralizing mAbs (MH3, 8A1), as well as one of the moderate toxin-neutralizing mAbs (LF1), recognize distinct epitopes near the low affinity Gal recognition domain in RTB subdomain 1α. Evaluated in a mouse model of systemic ricin challenge, all five mAbs afforded some benefit against intoxication, but only MH3 was protective. However, neither MH3 nor 24B11, another well-characterized mAb against RTB subdomain 1α, could passively protect mice against a mucosal (intranasal) ricin challenge. This is in contrast to SylH3, a previously characterized mAb directed against an epitope near RTB’s high affinity Gal/GalNac recognition element in sub-domain 2γ, which protected animals against systemic and mucosal ricin exposure. SylH3 was significantly more effective than MH3 and 24B11 at blocking ricin attachment to host cell receptors, suggesting that mucosal immunity to ricin is best imparted by antibodies that target RTB’s high affinity Gal/GalNac recognition element in subdomain 2γ, not the low affinity Gal recognition domain in subdomain 1α.


Toxins | 2017

A Supercluster of Neutralizing Epitopes at the Interface of Ricin’s Enzymatic (RTA) and Binding (RTB) Subunits

Amanda Poon; David J. Vance; Yinghui Rong; Dylan J. Ehrbar; Nicholas J. Mantis

As part of an effort to engineer ricin antitoxins and immunotherapies, we previously produced and characterized a collection of phage-displayed, heavy chain-only antibodies (VHHs) from alpacas that had been immunized with ricin antigens. In our initial screens, we identified nine VHHs directed against ricin toxin’s binding subunit (RTB), but only one, JIZ-B7, had toxin-neutralizing activity. Linking JIZ-B7 to different VHHs against ricin’s enzymatic subunit (RTA) resulted in several bispecific antibodies with potent toxin-neutralizing activity in vitro and in vivo. JIZ-B7 may therefore be an integral component of a future VHH-based neutralizing agent (VNA) for ricin toxin. In this study, we now localize, using competitive ELISA, JIZ-B7’s epitope to a region of RTB’s domain 2 sandwiched between the high-affinity galactose/N-acetylgalactosamine (Gal/GalNAc)-binding site and the boundary of a neutralizing hotspot on RTA known as cluster II. Analysis of additional RTB (n = 8)- and holotoxin (n = 4)-specific VHHs from a recent series of screens identified a “supercluster” of neutralizing epitopes at the RTA-RTB interface. Among the VHHs tested, toxin-neutralizing activity was most closely associated with epitope proximity to RTA, and not interference with RTB’s ability to engage Gal/GalNAc receptors. We conclude that JIZ-B7 is representative of a larger group of potent toxin-neutralizing antibodies, possibly including many described in the literature dating back several decades, that recognize tertiary and possibly quaternary epitopes located at the RTA-RTB interface and that target a region of vulnerability on ricin toxin.


bioRxiv | 2018

Humanized Monoclonal Antibody Rescues Rhesus macaques from Aerosolized Ricin Toxin Exposure

Chad J. Roy; Dylan J. Ehrbar; Natasha Bohorova; Ognian Bohorov; Do Han Kim; Michael Pauley; Kevin J. Whaley; Yinghui Rong; Fernando Torres-Velez; Ellen S. Vitetta; Peter J. Didier; Lara A. Doyle-Meyers; Larry Zeitlin; Nicholas J. Mantis

Ricin toxin (RT) ranks at the top of the list of potential bioweapons of concern to civilian and military personnel alike due to its high potential for morbidity and mortality after inhalation. In non-human primates, aerosolized ricin triggers a severe acute respiratory distress characterized by perivascular and alveolar edema, neutrophilic infiltration, and severe necrotizing bronchiolitis and alveolitis. There are currently no approved countermeasures for ricin intoxication. In this report, we demonstrate the therapeutic potential of huPB10, a toxin-neutralizing humanized monoclonal antibody (MAb) against an immunodominant epitope on ricin’s enzymatic A chain (RTA). Five rhesus macaques that received intravenous huPB10 (10 mg/kg) four hours after lethal dose ricin aerosol exposure all survived the toxin challenge, as compared to control animals, which succumbed to ricin intoxication within 30 h. Antibody treatment at 12 h after ricin exposure resulted in the survival of only one of five monkeys, indicating that, in the majority of animals, ricin intoxication and local tissue damage had progressed beyond the point where huPB10 intervention was beneficial. Change in pro-inflammatory cytokine/chemokines levels in bronchial alveolar lavage fluids before and after toxin challenge successfully clustered monkeys based on survival, as well as treatment group. IL-6 was the most apparent marker of ricin intoxication. This study represents the first demonstration in nonhuman primates that the lethal effects of inhalational ricin exposure can be negated by a drug candidate and opens up a path forward for product development.Ricin toxin ranks at the top of the list of potential bioweapons of concern to civilian and military personnel alike due to the high potential for morbidity and mortality after inhalation. In non-human primates, aerosolized ricin triggers a severe acute respiratory distress characterized by perivascular and alveolar edema, neutrophilic infiltration, and severe necrotizing bronchiolitis and alveolitis. Despite heightened public health and military concerns, there are currently no approved countermeasures for ricin. In this report, we demonstrate the therapeutic potential of huPB10, a toxin-neutralizing humanized monoclonal antibody (MAb) against an immunodominant epitope on ricin s enzymatic subunit. Five rhesus macaques that received intravenous huPB10 (10 mg/kg) four hours after lethal dose ricin aerosol exposure all survived the toxin challenge, as compared to control animals, which succumbed to ricin intoxication within 30 h. Antibody treatment at 12 h post-ricin exposure resulted in the survival of only one of five monkeys, signifying that, in the majority of animals, ricin intoxication and local tissue damage had progressed beyond the point where huPB10 intervention was beneficial. Principle coordinate analysis of fold change in pro-inflammatory cytokine/chemokines levels in bronchial alveolar lavage fluids before and after toxin challenge successfully clustered monkeys based on survival, as well as treatment group. IL-6 was the most apparent marker of ricin intoxication. This study represents the first demonstration in nonhuman primates that the lethal effects of inhalational ricin exposure can be negated by a drug candidate and opens up a path forward for product development.


bioRxiv | 2018

Fine-Specificity Epitope Analysis Identifies Contact Points on Ricin Toxin Recognized by Protective Monoclonal Antibodies

Greta Van Slyke; Siva Krishna Angalakurthi; Ronald T. Toth; David J. Vance; Yinghui Rong; Dylan J. Ehrbar; Yuqi Shi; C. Russell Middaugh; David B. Volkin; David D. Weis; Nicholas J. Mantis

Ricin is a fast-acting protein toxin classified by the Centers for Disease Control and Prevention as a biothreat agent. In this report, we describe five new mouse mAbs directed against an immunodominant region, so-called epitope cluster II, on the surface of ricin’s ribosome-inactivating enzymatic subunit A (RTA). The five mAbs were tested alongside four previously described cluster II–specific mAbs for their capacity to passively protect mice against 10× LD50 ricin challenge by injection. Only three of the mAbs (LE4, PH12, and TB12) afforded protection over the 7-d study period. Neither binding affinity nor in vitro toxin-neutralizing activity could fully account for LE4, PH12, and TB12’s potent in vivo activity relative to the other six mAbs. However, epitope mapping studies by hydrogen exchange–mass spectrometry revealed that LE4, PH12, and TB12 shared common contact points on RTA corresponding to RTA α-helices D and E and β-strands d and e located on the back side of RTA relative to the active site. The other six mAbs recognized overlapping epitopes on RTA, but none shared the same hydrogen exchange–mass spectrometry profile as LE4, PH12, and TB12. A high-density competition ELISA with a panel of ricin-specific, single-domain camelid Abs indicated that even though LE4, PH12, and TB12 make contact with similar secondary motifs, they likely approach RTA from different angles. These results underscore how subtle differences in epitope specificity can significantly impact Ab functionality in vivo.

Collaboration


Dive into the Yinghui Rong's collaboration.

Top Co-Authors

Avatar

Nicholas J. Mantis

New York State Department of Health

View shared research outputs
Top Co-Authors

Avatar

David J. Vance

New York State Department of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dylan J. Ehrbar

New York State Department of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge