Network


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

Hotspot


Dive into the research topics where Yaoxing Huang is active.

Publication


Featured researches published by Yaoxing Huang.


Proceedings of the National Academy of Sciences of the United States of America | 2008

The microbial mimic poly IC induces durable and protective CD4+ T cell immunity together with a dendritic cell targeted vaccine

Christine Trumpfheller; Marina Caskey; Godwin Nchinda; Maria Paula Longhi; Olga Mizenina; Yaoxing Huang; Sarah J. Schlesinger; Marco Colonna; Ralph M. Steinman

CD4+ Th1 type immunity is implicated in resistance to global infectious diseases. To improve the efficacy of T cell immunity induced by human immunodeficiency virus (HIV) vaccines, we are developing a protein-based approach that directly harnesses the function of dendritic cells (DCs) in intact lymphoid tissues. Vaccine proteins are selectively delivered to DCs by antibodies to DEC-205/CD205, a receptor for antigen presentation. We find that polyriboinosinic:polyribocytidylic acid (poly IC) independently serves as an adjuvant to allow a DC-targeted protein to induce protective CD4+ T cell responses at a mucosal surface, the airway. After two doses of DEC-targeted, HIV gag p24 along with poly IC, responder CD4+ T cells have qualitative features that have been correlated with protective function. The T cells simultaneously make IFN-γ, tumor necrosis factor (TNF)-α, and IL-2, and in high amounts for prolonged periods. The T cells also proliferate and continue to secrete IFN-γ in response to HIV gag p24. The adjuvant role of poly IC requires Toll-like receptor (TLR) 3 and melanoma differentiation-associated gene-5 (MDA5) receptors, but its analog poly IC12U requires only TLR3. We suggest that poly IC be tested as an adjuvant with DC-targeted vaccines to induce numerous multifunctional CD4+ Th1 cells with proliferative capacity.


Journal of Experimental Medicine | 2006

Intensified and protective CD4+ T cell immunity in mice with anti-dendritic cell HIV gag fusion antibody vaccine.

Christine Trumpfheller; Jennifer S. Finke; Carolina B. López; Thomas M. Moran; Bruno Moltedo; Helena Soares; Yaoxing Huang; Sarah J. Schlesinger; Chae Gyu Park; Michel C. Nussenzweig; Angela Granelli-Piperno; Ralph M. Steinman

Current human immunodeficiency virus (HIV) vaccine approaches emphasize prime boost strategies comprising multiple doses of DNA vaccine and recombinant viral vectors. We are developing a protein-based approach that directly harnesses principles for generating T cell immunity. Vaccine is delivered to maturing dendritic cells in lymphoid tissue by engineering protein antigen into an antibody to DEC-205, a receptor for antigen presentation. Here we characterize the CD4+ T cell immune response to HIV gag and compare efficacy with other vaccine strategies in a single dose. DEC-205–targeted HIV gag p24 or p41 induces stronger CD4+ T cell immunity relative to high doses of gag protein, HIV gag plasmid DNA, or recombinant adenovirus-gag. High frequencies of interferon (IFN)-γ– and interleukin 2–producing CD4+ T cells are elicited, including double cytokine-producing cells. In addition, the response is broad because the primed mice respond to an array of peptides in different major histocompatibility complex haplotypes. Long-lived T cell memory is observed. After subcutaneous vaccination, CD4+ and IFN-γ–dependent protection develops to a challenge with recombinant vaccinia-gag virus at a mucosal surface, the airway. We suggest that a DEC-targeted vaccine, in part because of an unusually strong and protective CD4+ T cell response, will improve vaccine efficacy as a stand-alone approach or with other modalities.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A

Juliana Idoyaga; Ashira Lubkin; Christopher Fiorese; Mireille H. Lahoud; Irina Caminschi; Yaoxing Huang; Anthony Rodriguez; Björn E. Clausen; Chae Gyu Park; Christine Trumpfheller; Ralph M. Steinman

Improved protein-based vaccines should facilitate the goal of effective vaccines against HIV and other pathogens. With respect to T cells, the efficiency of immunization, or “immunogenicity,” is improved by targeting vaccine proteins to maturing dendritic cells (DCs) within mAbs to DC receptors. Here, we compared the capacity of Langerin/CD207, DEC205/CD205, and Clec9A receptors, each expressed on the CD8+ DC subset in mice, to bring about immunization of microbial-specific T cells from the polyclonal repertoire, using HIV gag-p24 protein as an antigen. α-Langerin mAb targeted splenic CD8+ DCs selectively in vivo, whereas α-DEC205 and α-Clec9A mAbs targeted additional cell types. When the mAb heavy chains were engineered to express gag-p24, the α-Langerin, α-DEC205, and α-Clec9A fusion mAbs given along with a maturation stimulus induced comparable levels of gag-specific T helper 1 (Th1) and CD8+ T cells in BALB/c × C57BL/6 F1 mice. These immune T cells were more numerous than targeting the CD8− DC subset with α-DCIR2-gag-p24. In an in vivo assay in which gag-primed T cells were used to report the early stages of T-cell responses, α-Langerin, α-DEC205, and α-Clec9A also mediated cross-presentation to primed CD8+ T cells if, in parallel to antigen uptake, the DCs were stimulated with α-CD40. α-Langerin, α-DEC205, and α-Clec9A targeting greatly enhanced T-cell immunization relative to nonbinding control mAb or nontargeted HIV gag-p24 protein. Therefore, when the appropriate subset of DCs is targeted with a vaccine protein, several different receptors expressed by that subset are able to initiate combined Th1 and CD8+ immunity.


Journal of Clinical Investigation | 2008

The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells

Godwin Nchinda; Janelle My Kuroiwa; Margarita Oks; Christine Trumpfheller; Chae Gyu Park; Yaoxing Huang; Drew Hannaman; Sarah J. Schlesinger; Olga Mizenina; Michel C. Nussenzweig; Klaus Überla; Ralph M. Steinman

DNA vaccines promote an immune response by providing antigen-encoding DNA to the recipient, but the efficacy of such vaccines needs improving. Many approaches have considerable potential but currently induce relatively weak immune responses despite multiple high doses of DNA vaccine. Here, we asked whether targeting vaccine antigens to DCs would increase the immunity and protection that result from DNA vaccines. To determine this, we generated a DNA vaccine encoding a fusion protein comprised of the vaccine antigen and a single-chain Fv antibody (scFv) specific for the DC-restricted antigen-uptake receptor DEC205. Following vaccination of mice, the vaccine antigen was expressed selectively by DCs, which were required for the increased efficacy of MHC class I and MHC class II antigen presentation relative to a control scFv DNA vaccine. In addition, a DNA vaccine encoding an HIV gag p41-scFv DEC205 fusion protein induced 10-fold higher antibody levels and increased numbers of IFN-gamma-producing CD4+ and CD8+ T cells. After a single i.m. injection of the DNA vaccine encoding an HIV gag p41-scFv DEC205 fusion protein, mice were protected from an airway challenge with a recombinant vaccinia virus expressing the HIV gag p41, even with 1% of the dose of nontargeted DNA vaccine. The efficacy of DNA vaccines therefore may be enhanced by inclusion of sequences such as single-chain antibodies to target the antigen to DCs.


Proceedings of the National Academy of Sciences of the United States of America | 2008

A consensus–hemagglutinin-based DNA vaccine that protects mice against divergent H5N1 influenza viruses

Ming-Wei Chen; Ting-Jen R. Cheng; Yaoxing Huang; Jia-Tsrong Jan; Shiou-Hwa Ma; Alice L. Yu; Chi-Huey Wong; David D. Ho

H5N1 influenza viruses have spread extensively among wild birds and domestic poultry. Cross-species transmission of these viruses to humans has been documented in over 380 cases, with a mortality rate of ≈60%. There is great concern that a H5N1 virus would acquire the ability to spread efficiently between humans, thereby becoming a pandemic threat. An H5N1 influenza vaccine must, therefore, be an integral part of any pandemic preparedness plan. However, traditional methods of making influenza vaccines have yet to produce a candidate that could induce potently neutralizing antibodies against divergent strains of H5N1 influenza viruses. To address this need, we generated a consensus H5N1 hemagglutinin (HA) sequence based on data available in early 2006. This sequence was then optimized for protein expression before being inserted into a DNA plasmid (pCHA5). Immunizing mice with pCHA5, delivered intramuscularly via electroporation, elicited antibodies that neutralized a panel of virions that have been pseudotyped with the HA from various H5N1 viruses (clades 1, 2.1, 2.2, 2.3.2, and 2.3.4). Moreover, immunization with pCHA5 in mice conferred complete (clades 1 and 2.2) or significant (clade 2.1) protection from H5N1 virus challenges. We conclude that this vaccine, based on a consensus HA, could induce broad protection against divergent H5N1 influenza viruses and thus warrants further study.


PLOS ONE | 2011

In Vivo Electroporation Enhances the Immunogenicity of an HIV-1 DNA Vaccine Candidate in Healthy Volunteers

Sandhya Vasan; Arlene Hurley; Sarah J. Schlesinger; Drew Hannaman; David F. Gardiner; Daniel Dugin; Mar Boente-Carrera; Roselle Vittorino; Marina Caskey; Johanne Andersen; Yaoxing Huang; Josephine H. Cox; Tony Tarragona-Fiol; Dilbinder K. Gill; Hannah Cheeseman; Lorna Clark; Len Dally; Carol Smith; Claudia Schmidt; Harriet Park; Jakub Kopycinski; Jill Gilmour; Patricia Fast; Robert M. Bernard; David D. Ho

Background DNA-based vaccines have been safe but weakly immunogenic in humans to date. Methods and Findings We sought to determine the safety, tolerability, and immunogenicity of ADVAX, a multigenic HIV-1 DNA vaccine candidate, injected intramuscularly by in vivo electroporation (EP) in a Phase-1, double-blind, randomized placebo-controlled trial in healthy volunteers. Eight volunteers each received 0.2 mg, 1 mg, or 4 mg ADVAX or saline placebo via EP, or 4 mg ADVAX via standard intramuscular injection at weeks 0 and 8. A third vaccination was administered to eleven volunteers at week 36. EP was safe, well-tolerated and considered acceptable for a prophylactic vaccine. EP delivery of ADVAX increased the magnitude of HIV-1-specific cell mediated immunity by up to 70-fold over IM injection, as measured by gamma interferon ELISpot. The number of antigens to which the response was detected improved with EP and increasing dosage. Intracellular cytokine staining analysis of ELISpot responders revealed both CD4+ and CD8+ T cell responses, with co-secretion of multiple cytokines. Conclusions This is the first demonstration in healthy volunteers that EP is safe, tolerable, and effective in improving the magnitude, breadth and durability of cellular immune responses to a DNA vaccine candidate. Trial Registration ClinicalTrials.gov NCT00545987


AIDS | 1992

Identification and quantitation of HIV-1 in the liver of patients with AIDS

Yunzhen Cao; Douglas T. Dieterich; Patricia A. Thomas; Yaoxing Huang; Michael Mirabile; David D. Ho

ObjectiveTo detect and quantify HIV-1 in the liver in vivo.Design: Fourteen liver biopsy samples and corresponding blood lymphocytes and monocytes from patients with AIDS were studied for HIV-1 using quantitative polymerase chain reaction (PCR). In addition, expression of HIV-1 antigen and messenger (m) RNA in 10 autopsy liver specimens was examined by immunohistochemistry and in situ hybridization ResultsThe amount of HIV-1 DNA in nine liver samples ranged from 850 to 27000 copies per 106 cells, with mean and median values of 8150 and 3500 copies per 106 cells, respectively. Five other samples had no detectable HIV-1 DNA by PCR. Intracellular expression of HIV-1 antigen and mRNA was also detected in both Kupffer cells and hepatocytes by in situ studies. ConclusionThese findings strongly indicate that HIV-1 could replicate in the liver of a majority of patients with AIDS.


Journal of Virology | 2000

Enhanced Infectivity of an R5-Tropic Simian/Human Immunodeficiency Virus Carrying Human Immunodeficiency Virus Type 1 Subtype C Envelope after Serial Passages in Pig-Tailed Macaques (Macaca nemestrina)

Zhiwei Chen; Yaoxing Huang; Xiuqing Zhao; Eva Skulsky; Dorothy Lin; James Ip; Agegnehu Gettie; David D. Ho

ABSTRACT The increasing prevalence of human immunodeficiency virus type 1 (HIV-1) subtype C infection worldwide calls for efforts to develop a relevant animal model for evaluating strategies against the transmission of the virus. A chimeric simian/human immunodeficiency virus (SHIV), SHIVCHN19, was generated with a primary, non-syncytium-inducing HIV-1 subtype C envelope from a Chinese strain in the background of SHIV33. Unlike R5-tropic SHIV162, SHIVCHN19 was not found to replicate in rhesus CD4+ T lymphocytes. SHIVCHN19 does, however, replicate in CD4+ T lymphocytes of pig-tailed macaques (Macaca nemestrina). The observed replication competence of SHIVCHN19 requires the fulltat/rev genes and partial gp41 region derived from SHIV33. To evaluate in vivo infectivity, SHIVCHN19 was intravenously inoculated, at first, into two pig-tailed and two rhesus macaques. Although all four animals became infected, the virus replicated preferentially in pig-tailed macaques with an earlier plasma viral peak and a faster seroconversion. To determine whether in vivo adaptation would enhance the infectivity of SHIVCHN19, passages were carried out serially in three groups of two pig-tailed macaques each, via intravenous blood-bone marrow transfusion. The passages greatly enhanced the infectivity of the virus as shown by the increasingly elevated viral loads during acute infection in animals with each passage. Moreover, the doubling time of plasma virus during acute infection became much shorter in passage 4 (P4) animals (0.2 day) in comparison to P1 animals (1 to 2 days). P2 to P4 animals all became seropositive around 2 to 3 weeks postinoculation and had a decline in CD4/CD8 T-cell ratio during the early phase of infection. In P4 animals, a profound depletion of CD4 T cells in the lamina propria of the jejunum was observed. Persistent plasma viremia has been found in most of the infected animals with sustained viral loads ranging from 103 to 105per ml up to 6 months postinfection. Serial passages did not change the viral phenotype as confirmed by the persistence of the R5 tropism of SHIVCHN19 isolated from P4 animals. In addition, the infectivity of SHIVCHN19 in rhesus peripheral blood mononuclear cells was also increased after in vivo passages. Our data indicate that SHIVCHN19 has adapted well to grow in macaque cells. This established R5-tropic SHIVCHN19/macaque model would be very useful for HIV-1 subtype C vaccine and pathogenesis studies.


Vaccine | 2008

Enhancement of HIV DNA vaccine immunogenicity by the NKT cell ligand, α-galactosylceramide

Yaoxing Huang; Alex Chen; Xiangming Li; Zhiwei Chen; Wenyong Zhang; Yang Song; Deborah Gurner; David F. Gardiner; Sankha S. Basu; David D. Ho; Moriya Tsuji

A number of studies have shown that the natural killer T cell (NKT) ligand alpha-galactosylceramide (alpha-GalCer) serves as an adjuvant for various vaccines, including viral vaccines, parasite vaccines and protein vaccines. In this report, we investigated the adjuvant activity of alpha-GalCer on HIV-1 DNA vaccines in mice. This is a first study to show that alpha-GalCer can enhance the immunogenicity of DNA vaccines, since co-administration of alpha-GalCer with suboptimal doses of DNA vaccines greatly enhanced antigen-specific CD4+ T-cell and CD8+ T-cell responses. Differently from other vaccines, alpha-GalCer was also able to enhance HIV-specific antibody response 10-fold. It is of practical importance to find out that, in a DNA prime-DNA boost regimen, the adjuvant activity of alpha-GalCer was most profound when co-administered at the priming, but not at the boosting phase. In a dose-sparing experiment, we found that the level of cell-mediated immune responses in mice vaccinated with 5 microg of DNA in the presence of alpha-GalCer was equivalent to that of mice vaccinated with 50 microg of DNA in the absence of alpha-GalCer. Finally, results from CD1d and interferon-gamma receptor knockout mice confirm our previous data and determine the mechanistic dependence upon these molecules. These results illustrate that alpha-GalCer enhances the immunogenicity of DNA vaccines in a mechanism-based fashion. Since both mice and humans share the CD1d molecule, this information may aid in designing more effective DNA vaccines and vaccine adjuvants against HIV-1.


Cell | 2002

The HIV-1 Vaccine Race

David D. Ho; Yaoxing Huang

There is an urgent need to come up with a vaccine that will curtail the spread of HIV-1. To be successful, the scientific community must work in concert to develop sound strategies based on a deeper understanding of the virus. Significant technological advances are also required to overcome the unique obstacles posed by HIV-1. Some of the challenges we face in this endeavor are discussed here.

Collaboration


Dive into the Yaoxing Huang's collaboration.

Top Co-Authors

Avatar

David D. Ho

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhiwei Chen

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Jian Yu

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandhya Vasan

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yang Song

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar

Yunzhen Cao

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge