Network


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

Hotspot


Dive into the research topics where Lingyi Huang is active.

Publication


Featured researches published by Lingyi Huang.


Journal of Virology | 2003

Comparative Immunogenicity in Rhesus Monkeys of DNA Plasmid, Recombinant Vaccinia Virus, and Replication-Defective Adenovirus Vectors Expressing a Human Immunodeficiency Virus Type 1 gag Gene

Danilo R. Casimiro; Ling Chen; Tong-Ming Fu; Robert K. Evans; Michael J. Caulfield; Mary-Ellen Davies; Aimin Tang; Minchun Chen; Lingyi Huang; Virginia Harris; Daniel C. Freed; Keith A. Wilson; Sheri A. Dubey; De-Min Zhu; Denise K. Nawrocki; Henryk Mach; Robert Troutman; Lynne Isopi; Donna M. Williams; William Hurni; Zheng Xu; Jeffrey G. Smith; Su Wang; Xu Liu; Liming Guan; Romnie Long; Wendy L. Trigona; Gwendolyn J. Heidecker; Helen C. Perry; Natasha Persaud

ABSTRACT Cellular immune responses, particularly those associated with CD3+ CD8+ cytotoxic T lymphocytes (CTL), play a primary role in controlling viral infection, including persistent infection with human immunodeficiency virus type 1 (HIV-1). Accordingly, recent HIV-1 vaccine research efforts have focused on establishing the optimal means of eliciting such antiviral CTL immune responses. We evaluated several DNA vaccine formulations, a modified vaccinia virus Ankara vector, and a replication-defective adenovirus serotype 5 (Ad5) vector, each expressing the same codon-optimized HIV-1 gag gene for immunogenicity in rhesus monkeys. The DNA vaccines were formulated with and without one of two chemical adjuvants (aluminum phosphate and CRL1005). The Ad5-gag vector was the most effective in eliciting anti-Gag CTL. The vaccine produced both CD4+ and CD8+ T-cell responses, with the latter consistently being the dominant component. To determine the effect of existing antiadenovirus immunity on Ad5-gag-induced immune responses, monkeys were exposed to adenovirus subtype 5 that did not encode antigen prior to immunization with Ad5-gag. The resulting anti-Gag T-cell responses were attenuated but not abolished. Regimens that involved priming with different DNA vaccine formulations followed by boosting with the adenovirus vector were also compared. Of the formulations tested, the DNA-CRL1005 vaccine primed T-cell responses most effectively and provided the best overall immune responses after boosting with Ad5-gag. These results are suggestive of an immunization strategy for humans that are centered on use of the adenovirus vector and in which existing adenovirus immunity may be overcome by combined immunization with adjuvanted DNA and adenovirus vector boosting.


Journal of Virology | 2005

Attenuation of Simian Immunodeficiency Virus SIVmac239 Infection by Prophylactic Immunization with DNA and Recombinant Adenoviral Vaccine Vectors Expressing Gag

Danilo R. Casimiro; Fubao Wang; William A. Schleif; Xiaoping Liang; Zhi Qiang Zhang; Timothy W. Tobery; Mary-Ellen Davies; Adrian B. McDermott; David H. O'Connor; Arthur Fridman; Ansu Bagchi; Lynda Tussey; Andrew J. Bett; Adam C. Finnefrock; Tong-Ming Fu; Aimin Tang; Keith A. Wilson; Minchun Chen; Helen C. Perry; Gwendolyn J. Heidecker; Daniel C. Freed; Anthony Carella; Kara Punt; Kara J. Sykes; Lingyi Huang; Virginia I. Ausensi; Margaret Bachinsky; Usha Sadasivan-Nair; David I. Watkins; Emilio A. Emini

ABSTRACT The prophylactic efficacy of DNA and replication-incompetent adenovirus serotype 5 (Ad5) vaccine vectors expressing simian immunodeficiency virus (SIV) Gag was examined in rhesus macaques using an SIVmac239 challenge. Cohorts of either Mamu-A*01(+) or Mamu-A*01(−) macaques were immunized with a DNA prime-Ad5 boost regimen; for comparison, a third cohort consisting of Mamu-A*01(+) monkeys was immunized using the Ad5 vector alone for both prime and boost. All animals, along with unvaccinated control cohorts of Mamu-A*01(+) and Mamu-A*01(−) macaques, were challenged intrarectally with SIVmac239. Viral loads were measured in both peripheral and lymphoid compartments. Only the DNA prime-Ad5-boosted Mamu-A*01(+) cohort exhibited a notable reduction in peak plasma viral load (sevenfold) as well as in early set-point viral burdens in both plasma and lymphoid tissues (10-fold) relative to those observed in the control monkeys sharing the same Mamu-A*01 allele. The degree of control in each animal correlated with the levels of Gag-specific immunity before virus challenge. However, virus control was short-lived, and indications of viral escape were evident as early as 6 months postinfection. The implications of these results in vaccine design and clinical testing are discussed.


Journal of Virology | 2006

Vaccine-Induced Cellular Immune Responses Reduce Plasma Viral Concentrations after Repeated Low-Dose Challenge with Pathogenic Simian Immunodeficiency Virus SIVmac239

Nancy A. Wilson; Jason S. Reed; Gnankang Napoé; Shari M. Piaskowski; Andy Szymanski; Jessica Furlott; Edna J. Gonzalez; Levi Yant; Nicholas J. Maness; Gemma E. May; Taeko Soma; Matthew R. Reynolds; Eva G. Rakasz; Richard Rudersdorf; Adrian B. McDermott; David H. O'Connor; Thomas C. Friedrich; David B. Allison; Amit Patki; Louis J. Picker; Dennis R. Burton; Jing Lin; Lingyi Huang; Deepa Patel; Gwendolyn Heindecker; Jiang Fan; Michael Citron; Melanie Horton; Fubao Wang; Xiaoping Liang

ABSTRACT The goal of an AIDS vaccine regimen designed to induce cellular immune responses should be to reduce the viral set point and preserve memory CD4 lymphocytes. Here we investigated whether vaccine-induced cellular immunity in the absence of any Env-specific antibodies can control viral replication following multiple low-dose challenges with the highly pathogenic SIVmac239 isolate. Eight Mamu-A*01-positive Indian rhesus macaques were vaccinated with simian immunodeficiency virus (SIV) gag, tat, rev, and nef using a DNA prime-adenovirus boost strategy. Peak viremia (P = 0.007) and the chronic phase set point (P = 0.0192) were significantly decreased in the vaccinated cohort, out to 1 year postinfection. Loss of CD4+ memory populations was also ameliorated in vaccinated animals. Interestingly, only one of the eight vaccinees developed Env-specific neutralizing antibodies after infection. The control observed was significantly improved over that observed in animals vaccinated with SIV gag only. Vaccine-induced cellular immune responses can, therefore, exert a measure of control over replication of the AIDS virus in the complete absence of neutralizing antibody and give us hope that a vaccine designed to induce cellular immune responses might control viral replication.


Journal of Lipid Research | 2011

A PCSK9-binding antibody that structurally mimics the EGF(A) domain of LDL-receptor reduces LDL cholesterol in vivo

Yan G. Ni; Di Marco S; Jon H. Condra; Laurence B. Peterson; Weirong Wang; Fubao Wang; Shilpa Pandit; Holly A. Hammond; Ray Rosa; Cummings Rt; Dana D Wood; Xiaomei Liu; Bottomley Mj; Xun Shen; Cubbon Rm; Wang Sp; Douglas G. Johns; Volpari C; Hamuro L; Jayne Chin; Lingyi Huang; Jing Zhang Zhao; Salvatore Vitelli; Peter Haytko; Douglas Wisniewski; Lyndon J. Mitnaul; Carl P. Sparrow; Brian K. Hubbard; Andrea Carfi; Ayesha Sitlani

Proprotein convertase subtilisin-like/kexin type 9 (PCSK9) regulates LDL cholesterol levels by inhibiting LDL receptor (LDLr)-mediated cellular LDL uptake. We have identified a fragment antigen-binding (Fab) 1D05 which binds PCSK9 with nanomolar affinity. The fully human antibody 1D05-IgG2 completely blocks the inhibitory effects of wild-type PCSK9 and two gain-of-function human PCSK9 mutants, S127R and D374Y. The crystal structure of 1D05-Fab bound to PCSK9 reveals that 1D05-Fab binds to an epitope on the PCSK9 catalytic domain which includes the entire LDLr EGF(A) binding site. Notably, the 1D05-Fab CDR-H3 and CDR-H2 loops structurally mimic the EGF(A) domain of LDLr. In a transgenic mouse model (CETP/LDLr-hemi), in which plasma lipid and PCSK9 profiles are comparable to those of humans, 1D05-IgG2 reduces plasma LDL cholesterol to 40% and raises hepatic LDLr protein levels approximately fivefold. Similarly, in healthy rhesus monkeys, 1D05-IgG2 effectively reduced LDL cholesterol 20%–50% for over 2 weeks, despite its relatively short terminal half-life (t1/2 = 3.2 days). Importantly, the decrease in circulating LDL cholesterol corresponds closely to the reduction in free PCSK9 levels. Together these results clearly demonstrate that the LDL-lowering effect of the neutralizing anti-PCSK9 1D05-IgG2 antibody is mediated by reducing the amount of PCSK9 that can bind to the LDLr.


PLOS ONE | 2010

Characterization of notch1 antibodies that inhibit signaling of both normal and mutated notch1 receptors

Miguel Aste-Amezaga; Ningyan Zhang; Janet Lineberger; Beth Anne Arnold; Timothy J. Toner; Mingcheng Gu; Lingyi Huang; Salvatore Vitelli; Kim Vo; Peter Haytko; Jing Zhang Zhao; Frederic Baleydier; Sarah L'heureux; Hongfang Wang; Wendy R. Gordon; Elizabeth Thoryk; Marie Blanke Andrawes; Kittichoat Tiyanont; Kimberly Stegmaier; Giovanni Roti; Kenneth N. Ross; Laura L. Franlin; Hui Wang; Fubao Wang; Michael Chastain; Andrew J. Bett; Laurent P. Audoly; Stephen C. Blacklow; Hans E. Huber

Background Notch receptors normally play a key role in guiding a variety of cell fate decisions during development and differentiation of metazoan organisms. On the other hand, dysregulation of Notch1 signaling is associated with many different types of cancer as well as tumor angiogenesis, making Notch1 a potential therapeutic target. Principal Findings Here we report the in vitro activities of inhibitory Notch1 monoclonal antibodies derived from cell-based and solid-phase screening of a phage display library. Two classes of antibodies were found, one directed against the EGF-repeat region that encompasses the ligand-binding domain (LBD), and the second directed against the activation switch of the receptor, the Notch negative regulatory region (NRR). The antibodies are selective for Notch1, inhibiting Jag2-dependent signaling by Notch1 but not by Notch 2 and 3 in reporter gene assays, with EC50 values as low as 5±3 nM and 0.13±0.09 nM for the LBD and NRR antibodies, respectively, and fail to recognize Notch4. While more potent, NRR antibodies are incomplete antagonists of Notch1 signaling. The antagonistic activity of LBD, but not NRR, antibodies is strongly dependent on the activating ligand. Both LBD and NRR antibodies bind to Notch1 on human tumor cell lines and inhibit the expression of sentinel Notch target genes, including HES1, HES5, and DTX1. NRR antibodies also strongly inhibit ligand-independent signaling in heterologous cells transiently expressing Notch1 receptors with diverse NRR “class I” point mutations, the most common type of mutation found in human T-cell acute lymphoblastic leukemia (T-ALL). In contrast, NRR antibodies failed to antagonize Notch1 receptors bearing rare “class II” or “class III” mutations, in which amino acid insertions generate a duplicated or constitutively sensitive metalloprotease cleavage site. Signaling in T-ALL cell lines bearing class I mutations is partially refractory to inhibitory antibodies as compared to cell-penetrating gamma-secretase inhibitors. Conclusions/Significance Antibodies that compete with Notch1 ligand binding or that bind to the negative regulatory region can act as potent inhibitors of Notch1 signaling. These antibodies may have clinical utility for conditions in which inhibition of signaling by wild-type Notch1 is desired, but are likely to be of limited value for treatment of T-ALLs associated with aberrant Notch1 activation.


Journal of Virology | 2009

Vaccine-Induced Cellular Responses Control Simian Immunodeficiency Virus Replication after Heterologous Challenge

Nancy A. Wilson; Brandon F. Keele; Jason S. Reed; Shari M. Piaskowski; Caitlin E. MacNair; Andrew J. Bett; Xiaoping Liang; Fubao Wang; Elizabeth Thoryk; Gwendolyn J. Heidecker; Michael Citron; Lingyi Huang; Jing Lin; Salvatore Vitelli; Chanook D. Ahn; Masahiko Kaizu; Nicholas J. Maness; Matthew R. Reynolds; Thomas C. Friedrich; John T. Loffredo; Eva G. Rakasz; Stephen Erickson; David B. Allison; Michael Piatak; Jeffrey D. Lifson; John W. Shiver; Danilo R. Casimiro; George M. Shaw; Beatrice H. Hahn; David I. Watkins

ABSTRACT All human immunodeficiency virus (HIV) vaccine efficacy trials to date have ended in failure. Structural features of the Env glycoprotein and its enormous variability have frustrated efforts to induce broadly reactive neutralizing antibodies. To explore the extent to which vaccine-induced cellular immune responses, in the absence of neutralizing antibodies, can control replication of a heterologous, mucosal viral challenge, we vaccinated eight macaques with a DNA/Ad5 regimen expressing all of the proteins of SIVmac239 except Env. Vaccinees mounted high-frequency T-cell responses against 11 to 34 epitopes. We challenged the vaccinees and eight naïve animals with the heterologous biological isolate SIVsmE660, using a regimen intended to mimic typical HIV exposures resulting in infection. Viral loads in the vaccinees were significantly less at both the peak (1.9-log reduction; P < 0.03) and at the set point (2.6-log reduction; P < 0.006) than those in control naïve animals. Five of eight vaccinated macaques controlled acute peak viral replication to less than 80,000 viral RNA (vRNA) copy eq/ml and to less than 100 vRNA copy eq/ml in the chronic phase. Our results demonstrate that broad vaccine-induced cellular immune responses can effectively control replication of a pathogenic, heterologous AIDS virus, suggesting that T-cell-based vaccines may have greater potential than previously appreciated.


Journal of Virology | 2003

Vaccine-Induced Immunity in Baboons by Using DNA and Replication-Incompetent Adenovirus Type 5 Vectors Expressing a Human Immunodeficiency Virus Type 1 gag Gene

Danilo R. Casimiro; Aimin Tang; Ling Chen; Tong-Ming Fu; Robert K. Evans; Mary-Ellen Davies; Daniel C. Freed; William Hurni; Jose M. Aste-Amezaga; Liming Guan; Romnie Long; Lingyi Huang; Virginia Harris; Denise K. Nawrocki; Henryk Mach; Robert Troutman; Lynne Isopi; Krishna K. Murthy; Karen Rice; Keith A. Wilson; David B. Volkin; Emilio A. Emini; John W. Shiver

ABSTRACT The cellular immunogenicity of formulated plasmid DNA and replication-defective human adenovirus serotype 5 (Ad5) vaccine vectors expressing a codon-optimized human immunodeficiency virus type 1 gag gene was examined in baboons. The Ad5 vaccine was capable of inducing consistently strong, long-lived CD8+-biased T-cell responses and in vitro cytotoxic activities. The DNA vaccine-elicited immune responses were weaker than those elicited by the Ad5 vaccine and highly variable; formulation with chemical adjuvants led to moderate increases in the levels of Gag-specific T cells. Increasing the DNA-primed responses with booster doses of either Ad5 or modified vaccinia virus Ankara vaccines suggests a difference in the relative levels of cytotoxic and helper responses. The implications of these results are discussed.


Journal of Biological Chemistry | 2010

Generation and selection of novel fully human monoclonal antibodies that neutralize Dickkopf-1 (DKK1) inhibitory function in vitro and increase bone mass in vivo

Helmut Glantschnig; Richard Hampton; Ping Lu; Jing Zhang Zhao; Salvatore Vitelli; Lingyi Huang; Peter Haytko; Tara E. Cusick; Cheryl Ireland; Stephen W. Jarantow; Robin Ernst; Nan Wei; Pascale Nantermet; Kevin Scott; John E. Fisher; Fabio Talamo; Laura Orsatti; Alfred A. Reszka; Punam Sandhu; Donald Kimmel; Osvaldo Flores; William R. Strohl; Zhiqiang An; Fubao Wang

Wnt/LRP5 signaling is a central regulatory component of bone formative and resorptive activities, and the pathway inhibitor DKK1 is a suppressor of bone formation and bone mass accrual in mice. In addition, augmented DKK1 levels are associated with high bone turnover in diverse low bone mass states in rodent models and disease etiologies in human. However, examination of the precise role of DKK1 in the normal skeleton and in higher species requires the development of refined DKK1-specific pharmacological tools. Here, we report the strategy resulting in isolation of a panel of fully human anti-DKK1 antibodies applicable to studies interrogating the roles of mouse, rhesus, and human DKK1. Selected anti-DKK1 antibodies bind primate and human DKK-1 with picomolar affinities yet do not appreciably bind to DKK2 or DKK4. Epitopes mapped within the DKK1 C-terminal domain necessary for interaction with LRP5/6 and consequently effectively neutralized DKK1 function in vitro. When introduced into naïve normal growing female mice, IgGs significantly improved trabecular bone volume and structure and increased both trabecular and cortical bone mineral densities in a dose-related fashion. Furthermore, fully human DKK1-IgG displayed favorable pharmacokinetic parameters in non-human primates. In summary, we demonstrate here a rate-limiting function of physiologic DKK1 levels in the regulation of bone mass in intact female mice, amendable to specific pharmacologic neutralization by newly identified DKK1-IgGs. Importantly the fully human IgGs display a profile of attributes that recommends their testing in higher species and their use in evaluating DKK1 function in relevant disease models.


Journal of Virology | 2005

Vectored Gag and Env but Not Tat Show Efficacy against Simian-Human Immunodeficiency Virus 89.6P Challenge in Mamu-A*01-Negative Rhesus Monkeys

Xiaoping Liang; Danilo R. Casimiro; William A. Schleif; Fubao Wang; Mary-Ellen Davies; Zhi Qiang Zhang; Tong-Ming Fu; Adam C. Finnefrock; Larry Handt; Michael Citron; Gwendolyn J. Heidecker; Aimin Tang; Minchun Chen; Keith A. Wilson; Lori Gabryelski; Michael McElhaugh; Anthony Carella; Cheryl Moyer; Lingyi Huang; Salvatore Vitelli; Deepa Patel; Jing Lin; Emilio A. Emini; John W. Shiver

ABSTRACT Simian-human immunodeficiency virus (SHIV) challenge studies in rhesus macaques were conducted to evaluate the efficacy of adenovirus-based vaccines in the context of different major histocompatibility complex class I genetic backgrounds and different vaccine compositions. Mamu-A*01 allele-negative rhesus monkeys were immunized with one of the following vaccine constructs: (i) replication-defective recombinant adenovirus type 5 (Ad5) expressing human immunodeficiency virus type 1 (HIV-1) Tat (Ad5/HIVTat); (ii) Ad5 vector expressing simian immunodeficiency virus (SIV) Gag (Ad5/SIVGag); (iii) Ad5 vector expressing the truncated HIV-1jrfl Env, gp140 (Ad5/gp140_jrfl); (iv) Ad5 vector expressing the SHIV-89.6P gp140 (Ad5/gp140_89.6P); or (v) the combination of Ad5/SIVGag and Ad5/gp140_jrfl. Following intravenous challenge with SHIV-89.6P, only those cohorts that received vaccines expressing Gag or Env exhibited an attenuation of the acute viremia and associated CD4-cell lymphopenia. While no prechallenge neutralizing antibody titers were detectable in either Ad5/gp140-vaccinated group, an accelerated neutralizing antibody response was observed in the Ad5/gp140_89.6P-vaccinated group upon viral challenge. The set-point viral loads in the Ad5/SIVGag- and Ad5/gp140_jrfl-vaccinated groups were associated with the overall strength of the induced cellular immune responses. To examine the contribution of Mamu-A*01 allele in vaccine efficacy against SHIV-89.6P challenge, Mamu-A*01-positive monkeys were immunized with Ad5/SIVGag. Vaccine-mediated protection was significantly more pronounced in the Mamu-A*01-positive monkeys than in Mamu-A*01-negative monkeys, suggesting the strong contributions of T-cell epitopes restricted by the Mamu-A*01 molecule. The implications of these results in the development of an HIV-1 vaccine will be discussed.


International Journal of Biological Sciences | 2012

An Anti-PCSK9 Antibody Reduces LDL-Cholesterol On Top Of A Statin And Suppresses Hepatocyte SREBP-Regulated Genes

Liwen Zhang; Timothy Mccabe; Jon H. Condra; Yan G. Ni; Laurence B. Peterson; Weirong Wang; Alison M. Strack; Fubao Wang; Shilpa Pandit; Holly A. Hammond; Dana D Wood; Dale Lewis; Ray Rosa; Vivienne Mendoza; Anne Marie Cumiskey; Douglas G. Johns; Barbara C. Hansen; Xun Shen; Neil S. Geoghagen; Kristian K. Jensen; Lei Zhu; Karol Wietecha; Douglas Wisniewski; Lingyi Huang; Jing Zhang Zhao; Robin Ernst; Richard Hampton; Peter Haytko; Frances Ansbro; Shannon Chilewski

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a promising therapeutic target for treating coronary heart disease. We report a novel antibody 1B20 that binds to PCSK9 with sub-nanomolar affinity and antagonizes PCSK9 function in-vitro. In CETP/LDLR-hemi mice two successive doses of 1B20, administered 14 days apart at 3 or 10 mpk, induced dose dependent reductions in LDL-cholesterol (≥ 25% for 7-14 days) that correlated well with the extent of PCSK9 occupancy by the antibody. In addition, 1B20 induces increases in total plasma antibody-bound PCSK9 levels and decreases in liver mRNA levels of SREBP-regulated genes PCSK9 and LDLR, with a time course that parallels decreases in plasma LDL-cholesterol (LDL-C). Consistent with this observation in mice, in statin-responsive human primary hepatocytes, 1B20 lowers PCSK9 and LDLR mRNA levels and raises serum steady-state levels of antibody-bound PCSK9. In addition, mRNA levels of several SREBP regulated genes involved in cholesterol and fatty-acid synthesis including ACSS2, FDPS, IDI1, MVD, HMGCR, and CYP51A1 were decreased significantly with antibody treatment of primary human hepatocytes. In rhesus monkeys, subcutaneous (SC) dosing of 1B20 dose-dependently induces robust LDL-C lowering (maximal ~70%), which is correlated with increases in target engagement and total antibody-bound PCSK9 levels. Importantly, a combination of 1B20 and Simvastatin in dyslipidemic rhesus monkeys reduced LDL-C more than either agent alone, consistent with a mechanism of action that predicts additive effects of anti-PCSK9 agents with statins. Our results suggest that antibodies targeting PCSK9 could provide patients powerful LDL lowering efficacy on top of statins, and lower cardiovascular risk.

Collaboration


Dive into the Lingyi Huang's collaboration.

Top Co-Authors

Avatar

Aimin Tang

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Keith A. Wilson

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Mary-Ellen Davies

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge