Network


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

Hotspot


Dive into the research topics where Dirk M. Zajonc is active.

Publication


Featured researches published by Dirk M. Zajonc.


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.


Nature Immunology | 2011

Invariant natural killer T cells recognize glycolipids from pathogenic Gram-positive bacteria

Yuki Kinjo; Petr A. Illarionov; José Luis Vela; Bo Pei; Enrico Girardi; Xiangming Li; Yali Li; Masakazu Imamura; Yukihiro Kaneko; Akiko Okawara; Yoshitsugu Miyazaki; Anaximandro Gómez-Velasco; Paul Rogers; Samira Dahesh; Satoshi Uchiyama; Archana Khurana; Kazuyoshi Kawahara; Hasan Yesilkaya; Peter W. Andrew; Chi-Huey Wong; Kazuyoshi Kawakami; Victor Nizet; Gurdyal S. Besra; Moriya Tsuji; Dirk M. Zajonc; Mitchell Kronenberg

Natural killer T cells (NKT cells) recognize glycolipid antigens presented by CD1d. These cells express an evolutionarily conserved, invariant T cell antigen receptor (TCR), but the forces that drive TCR conservation have remained uncertain. Here we show that NKT cells recognized diacylglycerol-containing glycolipids from Streptococcus pneumoniae, the leading cause of community-acquired pneumonia, and group B Streptococcus, which causes neonatal sepsis and meningitis. Furthermore, CD1d-dependent responses by NKT cells were required for activation and host protection. The glycolipid response was dependent on vaccenic acid, which is present in low concentrations in mammalian cells. Our results show how microbial lipids position the sugar for recognition by the invariant TCR and, most notably, extend the range of microbes recognized by this conserved TCR to several clinically important bacteria.


PubMed | 2011

Invariant natural killer T cells recognize glycolipids from pathogenic Gram-positive bacteria.

Yuki Kinjo; Petr A. Illarionov; José Luis Vela; Bo Pei; Enrico Girardi; Xiangming Li; Yali Li; Masakazu Imamura; Yukihiro Kaneko; Akiko Okawara; Yoshitsugu Miyazaki; Anaximandro Gómez-Velasco; Paul Rogers; Samira Dahesh; Satoshi Uchiyama; Archana Khurana; Kazuyoshi Kawahara; Hasan Yesilkaya; Peter W. Andrew; Chi-Huey Wong; Kazuyoshi Kawakami; Nizet; Gurdyal S. Besra; Moriya Tsuji; Dirk M. Zajonc; Mitchell Kronenberg

Natural killer T cells (NKT cells) recognize glycolipid antigens presented by CD1d. These cells express an evolutionarily conserved, invariant T cell antigen receptor (TCR), but the forces that drive TCR conservation have remained uncertain. Here we show that NKT cells recognized diacylglycerol-containing glycolipids from Streptococcus pneumoniae, the leading cause of community-acquired pneumonia, and group B Streptococcus, which causes neonatal sepsis and meningitis. Furthermore, CD1d-dependent responses by NKT cells were required for activation and host protection. The glycolipid response was dependent on vaccenic acid, which is present in low concentrations in mammalian cells. Our results show how microbial lipids position the sugar for recognition by the invariant TCR and, most notably, extend the range of microbes recognized by this conserved TCR to several clinically important bacteria.


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.


Nature Reviews Immunology | 2005

Anatomy of CD1–lipid antigen complexes

D. Branch Moody; Dirk M. Zajonc; Ian A. Wilson

CD1 proteins bind lipids to form antigen complexes that contact T-cell receptors and activate T cells. Recent crystal structures of CD1 proteins show that their antigen-binding grooves are composed of up to four pockets (A′, C′, F′ and T′) and two antigen portals (C′ and F′). Although certain structural features are conserved among CD1 proteins, the grooves of CD1a, CD1b and CD1d differ in the number, shape and connectivity of their antigen-binding pockets. Here, we outline how the portals and pockets of CD1 antigen-binding grooves influence ligand specificity and facilitate the presentation of a surprisingly diverse set of antigenic lipids, glycolipids, lipopeptides and even small, non-lipidic molecules.


Immunity | 2014

The identification of the endogenous ligands of natural killer T cells reveals the presence of mammalian α-linked glycosylceramides.

Lisa Kain; Bill Webb; Brian L. Anderson; Shenglou Deng; Marie Holt; Anne Costanzo; Meng Zhao; Kevin Self; Anais Teyton; Chris Everett; Mitchell Kronenberg; Dirk M. Zajonc; Albert Bendelac; Paul B. Savage; Luc Teyton

Glycosylceramides in mammalian species are thought to be present in the form of β-anomers. This conclusion was reinforced by the identification of only one glucosylceramide and one galactosylceramide synthase, both β-transferases, in mammalian genomes. Thus, the possibility that small amounts of α-anomers could be produced by an alternative enzymatic pathway, by an unfaithful enzyme, or spontaneously in unusual cellular compartments has not been examined in detail. We approached the question by taking advantage of the exquisite specificity of T and B lymphocytes and combined it with the specificity of catabolic enzymes of the sphingolipid pathway. Here, we demonstrate that mammalian immune cells produce constitutively very small quantities of α-glycosylceramides, which are the major endogenous ligands of natural killer T cells. Catabolic enzymes of the ceramide and glycolipid pathway tightly control the amount of these α-glycosylceramides. The exploitation of this pathway to manipulate the immune response will create new therapeutic opportunities.


Nature | 2005

Anatomy of CD1|[ndash]|lipid antigen complexes

D. Branch Moody; Dirk M. Zajonc; Ian A. Wilson

CD1 proteins bind lipids to form antigen complexes that contact T-cell receptors and activate T cells. Recent crystal structures of CD1 proteins show that their antigen-binding grooves are composed of up to four pockets (A′, C′, F′ and T′) and two antigen portals (C′ and F′). Although certain structural features are conserved among CD1 proteins, the grooves of CD1a, CD1b and CD1d differ in the number, shape and connectivity of their antigen-binding pockets. Here, we outline how the portals and pockets of CD1 antigen-binding grooves influence ligand specificity and facilitate the presentation of a surprisingly diverse set of antigenic lipids, glycolipids, lipopeptides and even small, non-lipidic molecules.


Journal of Immunology | 2010

Mechanisms for Glycolipid Antigen-Driven Cytokine Polarization by Vα14i NKT Cells

Barbara A. Sullivan; Niranjana Nagarajan; Gerhard Wingender; Jing Wang; Iain Scott; Moriya Tsuji; Richard W. Franck; Steven A. Porcelli; Dirk M. Zajonc; Mitchell Kronenberg

Certain glycolipid Ags for Vα14i NKT cells can direct the overall cytokine balance of the immune response. Th2-biasing OCH has a lower TCR avidity than the most potent agonist known, α-galactosylceramide. Although the CD1d-exposed portions of OCH and α-galactosylceramide are identical, structural analysis indicates that there are subtle CD1d conformational differences due to differences in the buried lipid portion of these two Ags, likely accounting for the difference in antigenic potency. Th1-biasing C-glycoside/CD1d has even weaker TCR interactions than OCH/CD1d. Despite this, C-glycoside caused a greater downstream activation of NK cells to produce IFN-γ, accounting for its promotion of Th1 responses. We found that this difference correlated with the finding that C-glycoside/CD1d complexes survive much longer in vivo. Therefore, we suggest that the pharmacokinetic properties of glycolipids are a major determinant of cytokine skewing, suggesting a pathway for designing therapeutic glycolipids for modulating invariant NKT cell responses.


Nature Immunology | 2012

Type II natural killer T cells use features of both innate-like and conventional T cells to recognize sulfatide self antigens.

Enrico Girardi; Igor Maricic; Jing Wang; Thien-Thi Mac; Pooja Iyer; Vipin Kumar; Dirk M. Zajonc

Glycolipids presented by the major histocompatibility complex class I (MHC I) homolog CD1d are recognized by natural killer T (NKT) cells characterized by either a semi-invariant (type I or iNKT) or a relatively variable (type II) T cell receptor (TCR) repertoire. Here we describe the first structure of a type II NKT TCR complexed with CD1d-lysosulfatide (LSF). Both TCR α and β chains contacted the CD1d molecule with a diagonal footprint, typical of MHC-TCR interactions, while the antigen was recognized exclusively with a single TCR chain, similar to the iNKT TCR. Type II NKT cells, therefore, recognize CD1d-sulfatide complexes with a distinct recognition mechanism characterized by features of both iNKT cells as well as conventional peptide-reactive T cells.Glycolipids presented by the major histocompatibility complex (MHC) class I homolog CD1d are recognized by natural killer T cells (NKT cells) characterized by either a semi-invariant T cell antigen receptor (TCR) repertoire (type I NKT cells or iNKT cells) or a relatively variable TCR repertoire (type II NKT cells). Here we describe the structure of a type II NKT cell TCR in complex with CD1d-lysosulfatide. Both TCR α-chains and TCR β-chains made contact with the CD1d molecule with a diagonal footprint, typical of MHC-TCR interactions, whereas the antigen was recognized exclusively with a single TCR chain, similar to the iNKT cell TCR. Type II NKT cell TCRs, therefore, recognize CD1d-sulfatide complexes by a distinct recognition mechanism characterized by the TCR-binding features of both iNKT cells and conventional peptide-reactive T cells.


The EMBO Journal | 2011

Galactose-modified iNKT cell agonists stabilized by an induced fit of CD1d prevent tumour metastasis

Sandrine Aspeslagh; Yali Li; Esther Dawen Yu; Nora Pauwels; Matthias Trappeniers; Enrico Girardi; Tine Decruy; Katrien Van Beneden; Koen Venken; Michael Drennan; Luc Leybaert; Jing Wang; Richard W. Franck; Serge Van Calenbergh; Dirk M. Zajonc; Dirk Elewaut

Invariant natural killer T (iNKT) cells are known to have marked immunomodulatory capacity due to their ability to produce copious amounts of effector cytokines. Here, we report the structure and function of a novel class of aromatic α‐galactosylceramide structurally related glycolipids with marked Th1 bias in both mice and men, leading to superior tumour protection in vivo. The strength of the Th1 response correlates well with enhanced lipid binding to CD1d as a result of an induced fit mechanism that binds the aromatic substitution as a third anchor, in addition to the two lipid chains. This induced fit is in contrast to another Th1‐biasing glycolipid, α‐C‐GalCer, whose CD1d binding follows a conventional key‐lock principle. These findings highlight the previously unexploited flexibility of CD1d in accommodating galactose‐modified glycolipids and broaden the range of glycolipids that can stimulate iNKT cells. We speculate that glycolipids can be designed that induce a similar fit, thereby leading to superior and more sustained iNKT cell responses in vivo.

Collaboration


Dive into the Dirk M. Zajonc's collaboration.

Top Co-Authors

Avatar

Enrico Girardi

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Jing Wang

La Jolla Institute for Allergy and Immunology

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

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bjoern Peters

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Yali Li

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Michael H. Matho

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

D. Branch Moody

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Yan Xiang

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge