Network


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

Hotspot


Dive into the research topics where Douglass Wu is active.

Publication


Featured researches published by Douglass Wu.


Nature | 2005

Recognition of bacterial glycosphingolipids by natural killer T cells

Yuki Kinjo; Douglass Wu; Gisen Kim; Guo-Wen Xing; Michael A. Poles; David D. Ho; Moriya Tsuji; Kazuyoshi Kawahara; Chi-Huey Wong; Mitchell Kronenberg

Natural killer T (NKT) cells constitute a highly conserved T lymphocyte subpopulation that has the potential to regulate many types of immune responses through the rapid secretion of cytokines. NKT cells recognize glycolipids presented by CD1d, a class I-like antigen-presenting molecule. They have an invariant T-cell antigen receptor (TCR) α-chain, but whether this invariant TCR recognizes microbial antigens is still controversial. Here we show that most mouse and human NKT cells recognize glycosphingolipids from Sphingomonas, Gram-negative bacteria that do not contain lipopolysaccharide. NKT cells are activated in vivo after exposure to these bacterial antigens or bacteria, and mice that lack NKT cells have a marked defect in the clearance of Sphingomonas from the liver. These data suggest that NKT cells are T lymphocytes that provide an innate-type immune response to certain microorganisms through recognition by their antigen receptor, and that they might be useful in providing protection from bacteria that cannot be detected by pattern recognition receptors such as Toll-like receptor 4.


Nature Immunology | 2006

Natural killer T cells recognize diacylglycerol antigens from pathogenic bacteria.

Yuki Kinjo; Emmanuel Tupin; Douglass Wu; Masakazu Fujio; Raquel Garcia-Navarro; Mohammed Rafii El Idrissi Benhnia; Dirk M. Zajonc; Gil Ben-Menachem; Gary D. Ainge; Gavin F. Painter; Archana Khurana; Kasper Hoebe; Samuel M. Behar; Bruce Beutler; Ian A. Wilson; Moriya Tsuji; Timothy J. Sellati; Chi-Huey Wong; Mitchell Kronenberg

Natural killer T (NKT) cells recognize glycosphingolipids presented by CD1d molecules and have been linked to defense against microbial infections. Previously defined foreign glycosphingolipids recognized by NKT cells are uniquely found in nonpathogenic sphingomonas bacteria. Here we show that mouse and human NKT cells also recognized glycolipids, specifically a diacylglycerol, from Borrelia burgdorferi, which causes Lyme disease. The B. burgdorferi–derived, glycolipid-induced NKT cell proliferation and cytokine production and the antigenic potency of this glycolipid was dependent on acyl chain length and saturation. These data indicate that NKT cells recognize categories of glycolipids beyond those in sphingomonas and suggest that NKT cell responses driven by T cell receptor–mediated glycolipid recognition may provide protection against diverse pathogens.


Journal of Experimental Medicine | 2005

Structural basis for CD1d presentation of a sulfatide derived from myelin and its implications for autoimmunity

Dirk M. Zajonc; Igor Maricic; Douglass Wu; Ramesh Halder; Keshab Roy; Chi-Huey Wong; Vipin Kumar; Ian A. Wilson

Sulfatide derived from the myelin stimulates a distinct population of CD1d-restricted natural killer T (NKT) cells. Cis-tetracosenoyl sulfatide is one of the immunodominant species in myelin as identified by proliferation, cytokine secretion, and CD1d tetramer staining. The crystal structure of mouse CD1d in complex with cis-tetracosenoyl sulfatide at 1.9 Å resolution reveals that the longer cis-tetracosenoyl fatty acid chain fully occupies the A′ pocket of the CD1d binding groove, whereas the sphingosine chain fills up the F′ pocket. A precise hydrogen bond network in the center of the binding groove orients and positions the ceramide backbone for insertion of the lipid tails in their respective pockets. The 3′-sulfated galactose headgroup is highly exposed for presentation to the T cell receptor and projects up and away from the binding pocket due to its β linkage, compared with the more intimate binding of the α-glactosyl ceramide headgroup to CD1d. These structure and binding data on sulfatide presentation by CD1d have important implications for the design of therapeutics that target T cells reactive for myelin glycolipids in autoimmune diseases of the central nervous system.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Potent immune-modulating and anticancer effects of NKT cell stimulatory glycolipids

Ya-Jen Chang; Jing-Rong Huang; Yi-Chien Tsai; Jung-Tung Hung; Douglass Wu; Masakazu Fujio; Chi-Huey Wong; Alice L. Yu

α-Galactosylceramide (α-GalCer), a glycolipid that stimulates natural killer T (NKT) cells to produce both T helper (Th)1 and Th2 cytokines, has shown antitumor effects in mice but failed in clinical trials. We evaluated 16 analogs of α-GalCer for their CD1-mediated T cell receptor (TCR) activation of naïve human NKT cells and their anticancer efficacy. In vitro, glycolipids containing an aromatic ring in their acyl tail or sphingosine tail were more effective than α-GalCer in inducing Th1 cytokines/chemokines, TCR activation, and human NKT cell expansion. None of these glycolipids could directly stimulate human dendritic cell maturation, except for a glycolipid with an aromatic ring on the sphingosine tail. Here, we show that glycolipids activated the TCR of NKT cells with phosphorylation of CD3ε, ERK1/2, or CREB, which correlated with their induction of Th1 cytokines. Notably, the extent of NKT cell activation when glycolipid was presented by antigen-presenting cells was greater than when glycolipid was presented by non-antigen-presenting cells. In vivo, in mice bearing breast or lung cancers, the glycolipids that induced more Th1-biased cytokines and CD8/CD4 T cells displayed significantly greater anticancer potency than α-GalCer. These findings indicate that α-GalCer analogs can be designed to favor Th1-biased immunity, with greater anticancer efficacy and other immune-enhancing activities than α-GalCer itself.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Design of a potent CD1d-binding NKT cell ligand as a vaccine adjuvant

Xiangming Li; Masakazu Fujio; Masakazu Imamura; Douglass Wu; Sandhya Vasan; Chi-Huey Wong; David D. Ho; Moriya Tsuji

The glycolipid α-galactosylceramide (α-GalCer) has been shown to bind CD1d molecules to activate invariant natural killer T (iNKT) cells, and subsequently induce activation of various immune-competent cells, including dendritic cells, thereby providing a significant adjuvant effect for various vaccines. However, in phase I clinical trials, α-GalCer was shown to display only marginal biological activity. In our search for a glycolipid that can exert more potent stimulatory activity against iNKT cells and dendritic cells and produce an adjuvant effect superior to α-GalCer, we performed step-wise screening assays on a focused library of 25 α-GalCer analogues. Assays included quantification of the magnitude of stimulatory activity against human iNKT cells in vitro, binding affinity to human and murine CD1d molecules, and binding affinity to the invariant t cell receptor of human iNKT cells. Through this rigorous and iterative screening process, we have identified a lead candidate glycolipid, 7DW8-5, that exhibits a superior adjuvant effect than α-GalCer on HIV and malaria vaccines in mice.


Journal of Immunology | 2006

Conserved and Heterogeneous Lipid Antigen Specificities of CD1d-Restricted NKT Cell Receptors

Manfred Brigl; Peter van den Elzen; Xiuxu Chen; Jennifer Hartt Meyers; Douglass Wu; Chi-Huey Wong; Faye Reddington; Petr A. Illarianov; Gurdyal S. Besra; Michael B. Brenner; Jenny E. Gumperz

CD1d-restricted NKT cells use structurally conserved TCRs and recognize both self and foreign glycolipids, but the TCR features that determine these Ag specificities remain unclear. We investigated the TCR structures and lipid Ag recognition properties of five novel Vα24-negative and 13 canonical Vα24-positive/Vβ11-positive human NKT cell clones generated using α-galactosylceramide (α-GalCer)-loaded CD1d tetramers. The Vα24-negative clones expressed Vβ11 paired with Vα10, Vα2, or Vα3. Strikingly, their Vα-chains had highly conserved rearrangements to Jα18, resulting in CDR3α loop sequences that are nearly identical to those of canonical TCRs. Vα24-positive and Vα24-negative clones responded similarly to α-GalCer and a closely related bacterial analog, suggesting that conservation of the CDR3α loop is sufficient for recognition of α-GalCer despite CDR1α and CDR2α sequence variation. Unlike Vα24-positive clones, the Vα24-negative clones responded poorly to a glucose-linked glycolipid (α-glucosylceramide), which correlated with their lack of a conserved CDR1α amino acid motif, suggesting that fine specificity for α-linked glycosphingolipids is influenced by Vα-encoded TCR regions. Vα24-negative clones showed no response to isoglobotrihexosylceramide, indicating that recognition of this mammalian lipid is not required for selection of Jα18-positive TCRs that can recognize α-GalCer. One α-GalCer-reactive, Vα24-positive clone differed from the others in responding specifically to mammalian phospholipids, demonstrating that semi-invariant NKT TCRs have a capacity for private Ag specificities that are likely conferred by individual TCR β-chain rearrangements. These results highlight the variation in Ag recognition among CD1d-restricted TCRs and suggest that TCR α-chain elements contribute to α-linked glycosphingolipid specificity, whereas TCR β-chains can confer heterogeneous additional reactivities.


Chemistry & Biology | 2008

Natural Sphingomonas Glycolipids Vary Greatly in Their Ability to Activate Natural Killer T Cells

Yuki Kinjo; Bo Pei; Simone Bufali; Ravinder Raju; Stewart K. Richardson; Masakazu Imamura; Masakazu Fujio; Douglass Wu; Archana Khurana; Kazuyoshi Kawahara; Chi-Huey Wong; Amy R. Howell; Peter H. Seeberger; Mitchell Kronenberg

Mouse natural killer T (NKT) cells expressing an invariant T cell antigen receptor (TCR) recognize glycosphingolipids (GSLs) from Sphingomonas bacteria. The synthetic antigens previously tested, however, were designed to closely resemble the potent synthetic agonist alpha-galactosyl ceramide (alphaGalCer), which contains a monosaccharide and a C18:0 sphingosine lipid. Some Sphingomonas bacteria, however, also have oligosaccharide-containing GSLs, and they normally synthesize several GSLs with different sphingosine chains including one with a cyclopropyl ring-containing C21:0 (C21cycl) sphingosine. Here we studied the stimulation of NKT cells with synthetic GSL antigens containing natural tetrasaccharide sugars, or the C21cycl sphingosine. Our results indicate that there is a great degree of variability in the antigenic potency of different natural Sphingomonas glycolipids, with the C21cycl sphingosine having intermediate potency and the oligosaccharide-containing antigens exhibiting limited or no stimulatory capacity.


PLOS ONE | 2013

A Glycolipid Adjuvant, 7DW8-5, Enhances CD8+ T Cell Responses Induced by an Adenovirus-Vectored Malaria Vaccine in Non-Human Primates

Neal N. Padte; Mar Boente-Carrera; Chasity D. Andrews; Jenny McManus; Brooke Grasperge; Agegnehu Gettie; Jordana G. A. Coelho-dos-Reis; Xiangming Li; Douglass Wu; Joseph T. Bruder; Martha Sedegah; Noelle B. Patterson; Thomas L. Richie; Chi-Huey Wong; David D. Ho; Sandhya Vasan; Moriya Tsuji

A key strategy to a successful vaccine against malaria is to identify and develop new adjuvants that can enhance T-cell responses and improve protective immunity. Upon co-administration with a rodent malaria vaccine in mice, 7DW8-5, a recently identified novel analog of α-galactosylceramide (α-GalCer), enhances the level of malaria-specific protective immune responses more strongly than the parent compound. In this study, we sought to determine whether 7DW8-5 could provide a similar potent adjuvant effect on a candidate human malaria vaccine in the more relevant non-human primate (NHP) model, prior to committing to clinical development. The candidate human malaria vaccine, AdPfCA (NMRC-M3V-Ad-PfCA), consists of two non-replicating recombinant adenoviral (Ad) vectors, one expressing the circumsporozoite protein (CSP) and another expressing the apical membrane antigen-1 (AMA1) of Plasmodium falciparum. In several phase 1 clinical trials, AdPfCA was well tolerated and demonstrated immunogenicity for both humoral and cell-mediated responses. In the study described herein, 25 rhesus macaques received prime and boost intramuscular (IM) immunizations of AdPfCA alone or with an ascending dose of 7DW8-5. Our results indicate that 7DW8-5 is safe and well-tolerated and provides a significant enhancement (up to 9-fold) in malaria-specific CD8+ T-cell responses after both priming and boosting phases, supporting further clinical development.


Journal of Immunology | 2015

Colocalization of a CD1d-Binding Glycolipid with a Radiation-Attenuated Sporozoite Vaccine in Lymph Node–Resident Dendritic Cells for a Robust Adjuvant Effect

Xiangming Li; Akira Kawamura; Chasity D. Andrews; Jessica L. Miller; Douglass Wu; Tiffany Tsao; Min Zhang; Deena A. Oren; Neal N. Padte; Steven A. Porcelli; Chi-Huey Wong; Stefan H. I. Kappe; David D. Ho; Moriya Tsuji

A CD1d-binding glycolipid, α-Galactosylceramide (αGalCer), activates invariant NK T cells and acts as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying nearly 100-fold stronger CD1d binding affinity. In the current study, 7DW8-5 was found to exert a more potent adjuvant effect than αGalCer for a vaccine based on radiation-attenuated sporozoites of a rodent malaria parasite, Plasmodium yoelii, also referred to as irradiated P. yoelii sporozoites (IrPySpz). 7DW8-5 had a superb adjuvant effect only when the glycolipid and IrPySpz were conjointly administered i.m. Therefore, we evaluated the effect of distinctly different biodistribution patterns of αGalCer and 7DW8-5 on their respective adjuvant activities. Although both glycolipids induce a similar cytokine response in sera of mice injected i.v., after i.m. injection, αGalCer induces a systemic cytokine response, whereas 7DW8-5 is locally trapped by CD1d expressed by dendritic cells (DCs) in draining lymph nodes (dLNs). Moreover, the i.m. coadministration of 7DW8-5 with IrPySpz results in the recruitment of DCs to dLNs and the activation and maturation of DCs. These events cause the potent adjuvant effect of 7DW8-5, resulting in the enhancement of the CD8+ T cell response induced by IrPySpz and, ultimately, improved protection against malaria. Our study is the first to show that the colocalization of a CD1d-binding invariant NK T cell–stimulatory glycolipid and a vaccine, like radiation-attenuated sporozoites, in dLN-resident DCs upon i.m. conjoint administration governs the potency of the adjuvant effect of the glycolipid.


Proceedings of the National Academy of Sciences of the United States of America | 2005

Bacterial glycolipids and analogs as antigens for CD1d-restricted NKT cells

Douglass Wu; Guo-Wen Xing; Michael A. Poles; Amir Horowitz; Yuki Kinjo; Barbara A. Sullivan; Vera Bodmer-Narkevitch; Oliver Plettenburg; Mitchell Kronenberg; Moriya Tsuji; David D. Ho; Chi-Huey Wong

Collaboration


Dive into the Douglass Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masakazu Fujio

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Moriya Tsuji

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar

David D. Ho

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar

Mitchell Kronenberg

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Yuki Kinjo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dirk M. Zajonc

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Xiangming Li

Aaron Diamond AIDS Research Center

View shared research outputs
Top Co-Authors

Avatar

Barbara A. Sullivan

La Jolla Institute for Allergy and Immunology

View shared research outputs
Researchain Logo
Decentralizing Knowledge