Network


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

Hotspot


Dive into the research topics where Jonathan D. Silk is active.

Publication


Featured researches published by Jonathan D. Silk.


Journal of Immunology | 2003

NKT Cells Enhance CD4+ and CD8+ T Cell Responses to Soluble Antigen In Vivo through Direct Interaction with Dendritic Cells

Ian F. Hermans; Jonathan D. Silk; Uzi Gileadi; Mariolina Salio; Bini Mathew; Gerd Ritter; Richard R. Schmidt; Adrian L. Harris; L J Old; Vincenzo Cerundolo

Modification in the function of dendritic cells (DC), such as that achieved by microbial stimuli or T cell help, plays a critical role in determining the quality and size of adaptive responses to Ag. NKT cells bearing an invariant TCR (iNKT cells) restricted by nonpolymorphic CD1d molecules may constitute a readily available source of help for DC. We therefore examined T cell responses to i.v. injection of soluble Ag in the presence or the absence of iNKT cell stimulation with the CD1d-binding glycolipid α-galactosylceramide (α-GalCer). Considerably enhanced CD4+ and CD8+ T cell responses were observed when α-GalCer was administered at the same time as or close to OVA injection. This enhancement was dependent on the involvement of iNKT cells and CD1d molecules and required CD40 signaling. Studies in IFN-γR−/− mice indicated that IFN-γ was not required for the adjuvant effect of α-GalCer. Consistent with this result, enhanced T cell responses were observed using OCH, an analog of α-GalCer with a truncated sphingosine chain and a reduced capacity to induce IFN-γ. Splenic DC from α-GalCer-treated animals expressed high levels of costimulatory molecules, suggesting maturation in response to iNKT cell activation. Furthermore, studies with cultured DC indicated that potentiation of T cell responses required presentation of specific peptide and α-GalCer by the same DC, implying conditioning of DC by iNKT cells. The iNKT-enhanced T cell responses resisted challenge with OVA-expressing tumors, whereas responses induced in the absence of iNKT stimulation did not. Thus, iNKT cells exert a significant influence on the efficacy of immune responses to soluble Ag by modulating DC function.


Annual Review of Immunology | 2014

Biology of CD1- and MR1-Restricted T Cells

Mariolina Salio; Jonathan D. Silk; E. Yvonne Jones; Vincenzo Cerundolo

Over the past 15 years, investigators have shown that T lymphocytes can recognize not only peptides in the context of MHC class I and class II molecules but also foreign and self-lipids in association with the nonclassical MHC class I-like molecules, CD1 proteins. In this review, we describe the most recent events in the field, with particular emphasis on (a) structural and functional aspects of lipid presentation by CD1 molecules, (b) the development of CD1d-restricted invariant natural killer T (iNKT) cells and transcription factors required for their differentiation, (c) the ability of iNKT cells to modulate innate and adaptive immune responses through their cross talk with lymphoid and myeloid cells, and (d) MR1-restricted and group I (CD1a, CD1b, and CD1c)-restricted T cells.


Journal of Immunology | 2003

High Avidity Antigen-Specific CTL Identified by CD8-Independent Tetramer Staining

Ed Man-Lik Choi; Ji-Li Chen; Linda Wooldridge; Mariolina Salio; Anna Lissina; Nikolai Lissin; Ian F. Hermans; Jonathan D. Silk; Fareed Mirza; Michael J. Palmowski; P. Rod Dunbar; Bent K. Jakobsen; Andy K. Sewell; Vincenzo Cerundolo

Tetrameric MHC/peptide complexes are important tools for enumerating, phenotyping, and rapidly cloning Ag-specific T cells. It remains however unclear whether they can reliably distinguish between high and low avidity T cell clones. In this report, tetramers with mutated CD8 binding site selectively stain higher avidity human and murine CTL capable of recognizing physiological levels of Ag. Furthermore, we demonstrate that CD8 binding significantly enhances the avidity as well as the stability of interactions between CTL and cognate tetramers. The use of CD8-null tetramers to identify high avidity CTL provides a tool to compare vaccination strategies for their ability to enhance the frequency of high avidity CTL. Using this technique, we show that DNA priming and vaccinia boosting of HHD A2 transgenic mice fail to selectively expand large numbers of high avidity NY-ESO-1157–165-specific CTL, possibly due to the large amounts of antigenic peptide delivered by the vaccinia virus. Furthermore, development of a protocol for rapid identification of high avidity human and murine T cells using tetramers with impaired CD8 binding provides an opportunity not only to monitor expansion of high avidity T cell responses ex vivo, but also to sort high avidity CTL clones for adoptive T cell transfer therapy.


Journal of Immunology | 2008

Cutting Edge: Nonglycosidic CD1d Lipid Ligands Activate Human and Murine Invariant NKT Cells

Jonathan D. Silk; Mariolina Salio; B. Gopal Reddy; Dawn Shepherd; Uzi Gileadi; James Brown; S. Hajar Masri; Paolo Polzella; Gerd Ritter; Gurdyal S. Besra; E. Yvonne Jones; Richard R. Schmidt; Vincenzo Cerundolo

Invariant NKT cells (iNKT cells) recognize CD1d/glycolipid complexes. We demonstrate that the nonglycosidic compound threitolceramide efficiently activates iNKT cells, resulting in dendritic cell (DC) maturation and the priming of Ag-specific T and B cells. Threitolceramide-pulsed DCs are more resistant to iNKT cell-dependent lysis than α-galactosylceramide-pulsed DCs due to the weaker affinity of the human iNKT TCR for CD1d/ threitolceramide than CD1d/α-galactosylceramide complexes. iNKT cells stimulated with threitolceramide also recover more quickly from activation-induced anergy. Kinetic and functional experiments showed that shortening or lengthening the threitol moiety by one hydroxymethylene group modulates ligand recognition, as human and murine iNKT cells recognize glycerolceramide and arabinitolceramide differentially. Our data broaden the range of potential iNKT cell agonists. The ability of these compounds to assist the priming of Ag-specific immune responses while minimizing iNKT cell-dependent DC lysis makes them attractive adjuvants for vaccination strategies.


Journal of Immunology | 2007

Dendritic Cell Function Can Be Modulated through Cooperative Actions of TLR Ligands and Invariant NKT Cells

Ian F. Hermans; Jonathan D. Silk; Uzi Gileadi; S. Hajar Masri; Dawn Shepherd; Kathryn J. Farrand; Mariolina Salio; Vincenzo Cerundolo

The quality of signals received by dendritic cells (DC) in response to pathogens influences the nature of the adaptive response. We show that pathogen-derived signals to DC mediated via TLRs can be modulated by activated invariant NKT (iNKT) cells. DC maturation induced in vivo with any one of a variety of TLR ligands was greatly improved through simultaneous administration of the iNKT cell ligand α-galactosylceramide. DC isolated from animals treated simultaneously with TLR and iNKT cell ligands were potent stimulators of naive T cells in vitro compared with DC from animals treated with the ligands individually. Injection of protein Ags with both stimuli resulted in significantly improved T cell and Ab responses to coadministered protein Ags over TLR stimulation alone. Ag-specific CD8+ T cell responses induced in the presence of the TLR4 ligand monophosphoryl lipid A and α-galactosylceramide showed faster proliferation kinetics, and increased effector function, than those induced with either ligand alone. Human DC exposed to TLR ligands and activated iNKT cells in vitro had enhanced expression of maturation markers, suggesting that a cooperative action of TLR ligands and iNKT cells on DC function is a generalizable phenomenon across species. These studies highlight the potential for manipulating the interactions between TLR ligands and iNKT cell activation in the design of effective vaccine adjuvants.


Current Opinion in Immunology | 2010

Recent advances in processing and presentation of CD1 bound lipid antigens.

Mariolina Salio; Jonathan D. Silk; Vincenzo Cerundolo

It is well established that different populations of alphabeta T lymphocytes can recognize not only peptides in the context of MHC class I and class II molecules, but also foreign and self-lipids in association with CD1 proteins, which share structural similarities with MHC class I molecules. CD1 molecules are comprised of five isoforms, known as group 1 (CD1a, b, c, e) and group 2 (CD1d) CD1, presenting lipid antigens to conventional T lymphocytes or innate-like T cells bearing an invariant T cell receptor (TCR) and known as invariant NKT (iNKT) cells. During the last couple of years, several papers have been published describing important aspects of the mechanisms controlling the processing and presentation of endogenous and exogenous CD1 lipid antigens, which will be the main focus of this review.


Annual Review of Cell and Developmental Biology | 2008

Structural and Functional Aspects of Lipid Binding by CD1 Molecules

Jonathan D. Silk; Mariolina Salio; James Brown; E. Yvonne Jones; Vincenzo Cerundolo

Over the past ten years, investigators have shown that T lymphocytes can recognize not only peptides in the context of MHC class I and class II molecules but also foreign and self-lipids in association with the nonclassical MHC class I molecules the CD1 proteins. We describe the events that have led to the discovery of the role of CD1 molecules, their pattern of intracellular trafficking, and their ability to sample different intracellular compartments for self- and foreign lipids. Structural and functional aspects of lipid presentation by CD1 molecules are presented in the context of the function of CD1-restricted T cells in antimicrobial responses, antitumor immunity, and the regulation of the tolerance and autoimmunity immunoregulatory axis. Particular emphasis is on invariant NKT (iNKT) cells and their ability to modulate innate and adaptive immune responses.


Journal of Virology | 2012

Pseudotyped Influenza A Virus as a Vaccine for the Induction of Heterotypic Immunity

Timothy J. Powell; Jonathan D. Silk; Jane Sharps; Ervin Fodor; Alain Townsend

ABSTRACT There is a need for vaccines that can protect broadly across all influenza A strains. We have produced a pseudotyped influenza virus based on suppression of the A/PR/8/34 hemagglutinin signal sequence (S-FLU) that can infect cells and express the viral core proteins and neuraminidase but cannot replicate. We show that when given by inhalation to mice, S-FLU is nonpathogenic but generates a vigorous T cell response in the lung associated with markedly reduced viral titers and weight loss after challenge with H1 and H3 influenza viruses. These properties of S-FLU suggest that it may have potential as a broadly protective A virus vaccine, particularly in the setting of a threatened pandemic before matched subunit vaccines become available.


Gene Therapy | 2012

Cross-presentation of tumour antigens by human induced pluripotent stem cell-derived CD141 + XCR1 + dendritic cells

K M Silk; Jonathan D. Silk; N Ichiryu; Timothy J. Davies; Kathleen F. Nolan; A J Leishman; L Carpenter; S M Watt; Vincenzo Cerundolo; Paul J. Fairchild

Monocyte-derived dendritic cells (moDC) have been widely used in cancer immunotherapy but show significant donor-to-donor variability and low capacity for the cross-presentation of tumour-associated antigens (TAA) to CD8+ T cells, greatly limiting the success of this approach. Given recent developments in induced pluripotency and the relative ease with which induced pluripotent stem (iPS) cell lines may be generated from individuals, we have succeeded in differentiating dendritic cells (DC) from human leukocyte antigen (HLA)-A*0201+ iPS cells (iPS cell-derived DC (ipDC)), using protocols compliant with their subsequent clinical application. Unlike moDC, a subset of ipDC was found to coexpress CD141 and XCR1 that have been shown previously to define the human equivalent of mouse CD8α+ DC, in which the capacity for cross-presentation has been shown to reside. Accordingly, ipDC were able to cross-present the TAA, Melan A, to a CD8+ T-cell clone and stimulate primary Melan A-specific responses among naïve T cells from an HLA-A*0201+ donor. Given that CD141+XCR1+ DC are present in peripheral blood in trace numbers that preclude their clinical application, the ability to generate a potentially unlimited source from iPS cells offers the possibility of harnessing their capacity for cross-priming of cytotoxic T lymphocytes for the induction of tumour-specific immune responses.


Journal of Immunology | 2011

IDO Induces Expression of a Novel Tryptophan Transporter in Mouse and Human Tumor Cells

Jonathan D. Silk; Samira Lakhal; Robert Laynes; Laura Vallius; Ioannis Karydis; Cornelius Marcea; C. A. Richard Boyd; Vincenzo Cerundolo

IDO is the rate-limiting enzyme in the kynurenine pathway, catabolizing tryptophan to kynurenine. Tryptophan depletion by IDO-expressing tumors is a common mechanism of immune evasion inducing regulatory T cells and inhibiting effector T cells. Because mammalian cells cannot synthesize tryptophan, it remains unclear how IDO+ tumor cells overcome the detrimental effects of local tryptophan depletion. We demonstrate that IDO+ tumor cells express a novel amino acid transporter, which accounts for ∼50% of the tryptophan uptake. The induced transporter is biochemically distinguished from the constitutively expressed tryptophan transporter System L by increased resistance to inhibitors of System L, resistance to inhibition by high concentrations of most amino acids tested, and high substrate specificity for tryptophan. Under conditions of low extracellular tryptophan, expression of this novel transporter significantly increases tryptophan entry into IDO+ tumors relative to tryptophan uptake through the low-affinity System L alone, and further decreases tryptophan levels in the microenvironment. Targeting this additional tryptophan transporter could be a way of pharmacological inhibition of IDO-mediated tumor escape. These findings highlight the ability of IDO-expressing tumor cells to thrive in a tryptophan-depleted microenvironment by expressing a novel, highly tryptophan-specific transporter, which is resistant to inhibition by most other amino acids. The additional transporter allows tumor cells to strike the ideal balance between supply of tryptophan essential for their own proliferation and survival, and depleting the extracellular milieu of tryptophan to inhibit T cell proliferation.

Collaboration


Dive into the Jonathan D. Silk's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariolina Salio

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Uzi Gileadi

John Radcliffe Hospital

View shared research outputs
Top Co-Authors

Avatar

Ian F. Hermans

Malaghan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerd Ritter

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

E. Yvonne Jones

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Fareed Mirza

John Radcliffe Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge