Network


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

Hotspot


Dive into the research topics where Kenji Chamoto is active.

Publication


Featured researches published by Kenji Chamoto.


Journal of Immunology | 2015

Specific Roles of Each TCR Hemichain in Generating Functional Chain-Centric TCR

Munehide Nakatsugawa; Yuki Yamashita; Toshiki Ochi; Shinya Tanaka; Kenji Chamoto; Tingxi Guo; Marcus O. Butler; Naoto Hirano

TCRα- and β-chains cooperatively recognize peptide–MHC complexes. It has been shown that a “chain-centric” TCR hemichain can, by itself, dictate MHC-restricted Ag specificity without requiring major contributions from the paired TCR counterchain. Little is known, however, regarding the relative contributions and roles of chain-centric and its counter, non–chain-centric, hemichains in determining T cell avidity. We comprehensively analyzed a thymically unselected T cell repertoire generated by transducing the α-chain–centric HLA-A*02:01(A2)/MART127–35 TCRα, clone SIG35α, into A2-matched and unmatched postthymic T cells. Regardless of their HLA-A2 positivity, a substantial subset of peripheral T cells transduced with SIG35α gained reactivity for A2/MART127–35. Although the generated A2/MART127–35–specific T cells used various TRBV genes, TRBV27 predominated with >102 highly diverse and unique clonotypic CDR3β sequences. T cells individually reconstituted with various A2/MART127–35 TRBV27 TCRβ genes along with SIG35α possessed a wide range (>2 log orders) of avidity. Approximately half possessed avidity higher than T cells expressing clone DMF5, a naturally occurring A2/MART127–35 TCR with one of the highest affinities. Importantly, similar findings were recapitulated with other self-Ags. Our results indicate that, although a chain-centric TCR hemichain determines Ag specificity, the paired counterchain can regulate avidity over a broad range (>2 log orders) without compromising Ag specificity. TCR chain centricity can be exploited to generate a thymically unselected Ag-specific T cell repertoire, which can be used to isolate high-avidity antitumor T cells and their uniquely encoded TCRs rarely found in the periphery because of tolerance.


Frontiers in Immunology | 2015

Adoptive T Cell Therapy Targeting CD1 and MR1

Tingxi Guo; Kenji Chamoto; Naoto Hirano

Adoptive T cell immunotherapy has demonstrated clinically relevant efficacy in treating malignant and infectious diseases. However, much of these therapies have been focused on enhancing, or generating de novo, effector functions of conventional T cells recognizing HLA molecules. Given the heterogeneity of HLA alleles, mismatched patients are ineligible for current HLA-restricted adoptive T cell therapies. CD1 and MR1 are class I-like monomorphic molecules and their restricted T cells possess unique T cell receptor specificity against entirely different classes of antigens. CD1 and MR1 molecules present lipid and vitamin B metabolite antigens, respectively, and offer a new front of targets for T cell therapies. This review will cover the recent progress in the basic research of CD1, MR1, and their restricted T cells that possess translational potential.


Cancer immunology research | 2015

Optimization of T-cell Reactivity by Exploiting TCR Chain Centricity for the Purpose of Safe and Effective Antitumor TCR Gene Therapy

Toshiki Ochi; Munehide Nakatsugawa; Kenji Chamoto; Shinya Tanaka; Yuki Yamashita; Tingxi Guo; Hiroshi Fujiwara; Masaki Yasukawa; Marcus O. Butler; Naoto Hirano

Adoptive transfer of redirected antitumor T cells can have off-target toxicities. Peptide-MHC specificity can be focused on a single TCR chain, allowing the authors to separate antitumor-reactivity from cross-reactivity, while showing that monitoring for toxicities is still necessary. Adoptive transfer of T cells redirected by a high-affinity antitumor T-cell receptor (TCR) is a promising treatment modality for cancer patients. Safety and efficacy depend on the selection of a TCR that induces minimal toxicity and elicits sufficient antitumor reactivity. Many, if not all, TCRs possess cross-reactivity to unrelated MHC molecules in addition to reactivity to target self-MHC/peptide complexes. Some TCRs display chain centricity, in which recognition of MHC/peptide complexes is dominated by one of the TCR hemi-chains. In this study, we comprehensively studied how TCR chain centricity affects reactivity to target self-MHC/peptide complexes and alloreactivity using the TCR, clone TAK1, which is specific for human leukocyte antigen-A*24:02/Wilms tumor 1235–243 (A24/WT1235) and cross-reactive with B*57:01 (B57). The TAK1β, but not the TAK1α, hemi-chain possessed chain centricity. When paired with multiple clonotypic TCRα counter-chains encoding TRAV12-2, 20, 36, or 38-2, the de novo TAK1β-containing TCRs showed enhanced, weakened, or absent reactivity to A24/WT1235 and/or to B57. T cells reconstituted with these TCRα genes along with TAK1β possessed a very broad range (>3 log orders) of functional and structural avidities. These results suggest that TCR chain centricity can be exploited to enhance desired antitumor TCR reactivity and eliminate unwanted TCR cross-reactivity. TCR reactivity to target MHC/peptide complexes and cross-reactivity to unrelated MHC molecules are not inextricably linked and are separable at the TCR sequence level. However, it is still mandatory to carefully monitor for possible harmful toxicities caused by adoptive transfer of T cells redirected by thymically unselected TCRs. Cancer Immunol Res; 3(9); 1070–81. ©2015 AACR.


Nature Communications | 2017

HLA-DP 84Gly constitutively presents endogenous peptides generated by the class I antigen processing pathway

Yuki Yamashita; Mark Anczurowski; Munehide Nakatsugawa; Makito Tanaka; Yuki Kagoya; Ankit Sinha; Kenji Chamoto; Toshiki Ochi; Tingxi Guo; Kayoko Saso; Marcus O. Butler; Mark D. Minden; Thomas Kislinger; Naoto Hirano

Classical antigen processing leads to the presentation of antigenic peptides derived from endogenous and exogenous sources for MHC class I and class II molecules, respectively. Here we show that, unlike other class II molecules, prevalent HLA-DP molecules with β-chains encoding Gly84 (DP84Gly) constitutively present endogenous peptides. DP84Gly does not bind invariant chain (Ii) via the class II-associated invariant chain peptide (CLIP) region, nor does it present CLIP. However, Ii does facilitate the transport of DP84Gly from the endoplasmic reticulum (ER) to the endosomal/lysosomal pathway by transiently binding DP84Gly via a non-CLIP region(s) in a pH-sensitive manner. Accordingly, like class I, DP84Gly constitutively presents endogenous peptides processed by the proteasome and transported to the ER by the transporter associated with antigen processing (TAP). Therefore, DP84Gly, found only in common chimpanzees and humans, uniquely uses both class I and II antigen-processing pathways to present peptides derived from intracellular and extracellular sources.


Journal of Autoimmunity | 2016

CDR3β sequence motifs regulate autoreactivity of human invariant NKT cell receptors

Kenji Chamoto; Tingxi Guo; Osamu Imataki; Makito Tanaka; Munehide Nakatsugawa; Toshiki Ochi; Yuki Yamashita; Akiko M. Saito; Toshiki I. Saito; Marcus O. Butler; Naoto Hirano

Invariant natural killer T (iNKT) cells are a subset of T lymphocytes that recognize lipid ligands presented by monomorphic CD1d. Human iNKT T cell receptor (TCR) is largely composed of invariant Vα24 (Vα24i) TCRα chain and semi-variant Vβ11 TCRβ chain, where complementarity-determining region (CDR)3β is the sole variable region. One of the characteristic features of iNKT cells is that they retain autoreactivity even after the thymic selection. However, the molecular features of human iNKT TCR CDR3β sequences that regulate autoreactivity remain unknown. Since the numbers of iNKT cells with detectable autoreactivity in peripheral blood is limited, we introduced the Vα24i gene into peripheral T cells and generated a de novo human iNKT TCR repertoire. By stimulating the transfected T cells with artificial antigen presenting cells (aAPCs) presenting self-ligands, we enriched strongly autoreactive iNKT TCRs and isolated a large panel of human iNKT TCRs with a broad range autoreactivity. From this panel of unique iNKT TCRs, we deciphered three CDR3β sequence motifs frequently encoded by strongly-autoreactive iNKT TCRs: a VD region with 2 or more acidic amino acids, usage of the Jβ2-5 allele, and a CDR3β region of 13 amino acids in length. iNKT TCRs encoding 2 or 3 sequence motifs also exhibit higher autoreactivity than those encoding 0 or 1 motifs. These data facilitate our understanding of the molecular basis for human iNKT cell autoreactivity involved in immune responses associated with human disease.


Journal of Immunology | 2016

Key Residues at Third CDR3β Position Impact Structure and Antigen Recognition of Human Invariant NK TCRs

Kenji Chamoto; Tingxi Guo; Stephen W. Scally; Yuki Kagoya; Mark Ancruzowski; Chung-Hsi Wang; Muhammed A. Rahman; Kayoko Saso; Marcus O. Butler; Priscilla P.L. Chiu; Jean-Philippe Julien; Naoto Hirano

The human invariant NK (iNK) TCR is largely composed of the invariant TCR Vα24-Jα18 chain and semivariant TCR Vβ11 chains with variable CDR3β sequences. The direct role of CDR3β in Ag recognition has been studied extensively. Although it was noted that CDR3β can interact with CDR3α, how this interaction might indirectly influence Ag recognition is not fully elucidated. We observed that the third position of Vβ11 CDR3 can encode an Arg or Ser residue as a result of somatic rearrangement. Clonotypic analysis of the two iNK TCR types with a single amino acid substitution revealed that the staining intensity by anti-Vα24 Abs depends on whether Ser or Arg is encoded. When stained with an anti–Vα24-Jα18 Ab, human primary invariant NKT cells could be divided into Vα24 low- and high-intensity subsets, and Arg-encoding TCR Vβ11 chains were more frequently isolated from the Vα24 low-intensity subpopulation compared with the Vα24 high-intensity subpopulation. The Arg/Ser substitution also influenced Ag recognition as determined by CD1d multimer staining and CD1d-restricted functional responses. Importantly, in silico modeling validated that this Ser-to-Arg mutation could alter the structure of the CDR3β loop, as well as the CDR3α loop. Collectively, these results indicate that the Arg/Ser encoded at the third CDR3β residue can effectively modulate the overall structure of, and Ag recognition by, human iNK TCRs.


Scientific Reports | 2016

CD4 + and CD8 + TCRβ repertoires possess different potentials to generate extraordinarily high-avidity T cells

Munehide Nakatsugawa; Muhammed A. Rahman; Yuki Yamashita; Toshiki Ochi; Piotr Wnuk; Shinya Tanaka; Kenji Chamoto; Yuki Kagoya; Kayoko Saso; Tingxi Guo; Mark Anczurowski; Marcus O. Butler; Naoto Hirano

Recent high throughput sequencing analysis has revealed that the TCRβ repertoire is largely different between CD8+ and CD4+ T cells. Here, we show that the transduction of SIG35α, the public chain-centric HLA-A*02:01(A2)/MART127–35 TCRα hemichain, conferred A2/MART127–35 reactivity to a substantial subset of both CD8+ and CD4+ T cells regardless of their HLA–A2 positivity. T cells individually reconstituted with SIG35α and different A2/MART127–35 TCRβ genes isolated from CD4+ or CD8+ T cells exhibited a wide range of avidity. Surprisingly, approximately half of the A2/MART127–35 TCRs derived from CD4+ T cells, but none from CD8+ T cells, were stained by A2/MART127–35 monomer and possessed broader cross-reactivity. Our results suggest that the differences in the primary structure of peripheral CD4+ and CD8+ TCRβ repertoire indeed result in the differences in their ability to form extraordinarily high avidity T cells which would otherwise have been deleted by central tolerance.


PLOS ONE | 2016

Mouse and Human CD1d-Self-Lipid Complexes Are Recognized Differently by Murine Invariant Natural Killer T Cell Receptors.

Tingxi Guo; Kenji Chamoto; Munehide Nakatsugawa; Toshiki Ochi; Yuki Yamashita; Mark Anczurowski; Marcus O. Butler; Naoto Hirano

Invariant natural killer T (iNKT) cells recognize self-lipids presented by CD1d through characteristic TCRs, which mainly consist of the invariant Vα14-Jα18 TCRα chain and Vβ8.2, 7 or 2 TCRβ chains with hypervariable CDR3β sequences in mice. The iNKT cell-CD1d axis is conserved between humans and mice, and human CD1d reactivity of murine iNKT cells have been described. However, the detailed differences between the recognition of human and mouse CD1d bound to various self-lipids by mouse iNKT TCRs are largely unknown. In this study, we generated a de novo murine iNKT TCR repertoire with a wider range of autoreactivity compared with that of naturally occurring peripheral iNKT TCRs. Vβ8.2 mouse iNKT TCRs capable of recognizing the human CD1d-self-lipid tetramer were identified, although such clones were not detectable in the Vβ7 or Vβ2 iNKT TCR repertoire. In line with previously reports, clonotypic Vβ8.2 iNKT TCRs with unique CDR3β loops did not discriminate among lipids presented by mouse CD1d. Unexpectedly, however, these iNKT TCRs showed greater ligand selectivity toward human CD1d presenting the same lipids. Our findings demonstrated that the recognition of mouse and human CD1d-self-lipid complexes by murine iNKT TCRs is not conserved, thereby further elucidating the differences between cognate and cross-species reactivity of self-antigens by mouse iNKT TCRs.


Natural Killer Cells#R##N#Basic Science and Clinical Application | 2010

Interaction of NK cells with bacteria

Kenji Chamoto; Daiko Wakita; Shin-ichi Koizumi; Kazutaka Masuko; Takayuki Ikeda; Rieko Mitamura; Takashi Nishimura

Publisher Summary Natural killer (NK) cells produce many chemokines and inflammatory cytokines in response to, and exhibit cytotoxic activity against, pathogen-infected cells. They contribute to the prevention of various infectious diseases, especially during the early phases of infection, before CD8+ cytotoxic T lymphocyte (CTL) induction. Natural killer (NK) cells, including NKT cells, are components of the innate immune system and contribute significantly to the clearance of pathogen-infected or malignant cells. Recently, it has been demonstrated that quite diverse pattern-recognition receptors expressed on dendritic cells (DCs) and macrophages (M) recognize pathogen-specific components and subsequently initiate NK cell activation through two distinct signals: soluble factors and cell-to-cell contact. Crosstalk between NK cells and DCs plays a pivotal role in bridging innate and acquired immunity. Most pathogens and some lactic acid bacteria can modulate the immune balance towards a Th1- predominance. Therefore, bacteria themselves or their components are used to improve the disrupted immune balance or to induce Th1 responses critical for the prevention of infectious diseases, cancers and allergies. The cellular and molecular mechanisms of regulating the innate and acquired immune responses by bacterial stimuli and NKT cell ligands via reciprocal interactions among DCs, NK and NKT cells has been reviewed. Sustaining healthy conditions in mice and humans require precise control of immune responses towards exogenous antigens because disruption of immune homeostasis causes various immune-associated diseases, such as allergy, autoimmune disease and cancer.


Journal of Immunology | 2014

Human peripheral invariant natural killer T are adaptive lymphocytes, whose TCR CDR3β regulates their antigen specificity and reactivity to avoid harmful autoimmune responses (BA8P.124)

Kenji Chamoto; Osamu Imataki; Tingxi Guo; Toshiki Saito; Akiko Saito; Andrew Elia; Priscilla P.L. Chiu; Marcus O. Butler; Naoto Hirano

Collaboration


Dive into the Kenji Chamoto's collaboration.

Top Co-Authors

Avatar

Naoto Hirano

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Tingxi Guo

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Marcus O. Butler

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Munehide Nakatsugawa

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Yuki Yamashita

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kayoko Saso

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Mark Anczurowski

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Muhammed A. Rahman

Princess Margaret Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge