Network


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

Hotspot


Dive into the research topics where Nicholas D. Huntington is active.

Publication


Featured researches published by Nicholas D. Huntington.


Cell | 2007

ER Stress Triggers Apoptosis by Activating BH3-Only Protein Bim

Lorraine A. O'Reilly; Priscilla Gunn; Lily Lee; Priscilla N. Kelly; Nicholas D. Huntington; Peter Hughes; Ewa M. Michalak; Jennifer L. McKimm-Breschkin; Noburo Motoyama; Tomomi Gotoh; Shizuo Akira; Andreas Strasser

Endoplasmic reticulum (ER) stress caused by misfolded proteins or cytotoxic drugs can kill cells and although activation of this pathway has been implicated in the etiology of certain degenerative disorders its mechanism remains unresolved. Bim, a proapoptotic BH3-only member of the Bcl-2 family is required for initiation of apoptosis induced by cytokine deprivation or certain stress stimuli. Its proapoptotic activity can be regulated by several transcriptional or posttranslational mechanisms, such as ERK-mediated phosphorylation, promoting its ubiquitination and proteasomal degradation. We found that Bim is essential for ER stress-induced apoptosis in a diverse range of cell types both in culture and within the whole animal. ER stress activates Bim through two novel pathways, involving protein phosphatase 2A-mediated dephosphorylation, which prevents its ubiquitination and proteasomal degradation and CHOP-C/EBPalpha-mediated direct transcriptional induction. These results define the molecular mechanisms of ER stress-induced apoptosis and identify targets for therapeutic intervention in ER stress-related diseases.


Journal of Experimental Medicine | 2009

IL-15 trans-presentation promotes human NK cell development and differentiation in vivo

Nicholas D. Huntington; Nicolas Legrand; Nuno L. Alves; Barbara Jaron; Kees Weijer; Ariane Plet; Erwan Corcuff; Erwan Mortier; Yannick Jacques; Hergen Spits; James P. Di Santo

The in vivo requirements for human natural killer (NK) cell development and differentiation into cytotoxic effectors expressing inhibitory receptors for self–major histocompatability complex class I (MHC-I; killer Ig-like receptors [KIRs]) remain undefined. Here, we dissect the role of interleukin (IL)-15 in human NK cell development using Rag2−/−γc−/− mice transplanted with human hematopoietic stem cells. Human NK cell reconstitution was intrinsically low in this model because of the poor reactivity to mouse IL-15. Although exogenous human IL-15 (hIL-15) alone made little improvement, IL-15 coupled to IL-15 receptor α (IL-15Rα) significantly augmented human NK cells. IL-15–IL-15Rα complexes induced extensive NK cell proliferation and differentiation, resulting in accumulation of CD16+KIR+ NK cells, which was not uniquely dependent on enhanced survival or preferential responsiveness of this subset to IL-15. Human NK cell differentiation in vivo required hIL-15 and progressed in a linear fashion from CD56hiCD16−KIR− to CD56loCD16+KIR−, and finally to CD56loCD16+KIR+. These data provide the first evidence that IL-15 trans-presentation regulates human NK cell homeostasis. Use of hIL-15 receptor agonists generates a robust humanized immune system model to study human NK cells in vivo. IL-15 receptor agonists may provide therapeutic tools to improve NK cell reconstitution after bone marrow transplants, enhance graft versus leukemia effects, and increase the pool of IL-15–responsive cells during immunotherapy strategies.


Nature Immunology | 2016

Targeting natural killer cells in cancer immunotherapy

Camille Guillerey; Nicholas D. Huntington; Mark J. Smyth

Alteration in the expression of cell-surface proteins is a common consequence of malignant transformation. Natural killer (NK) cells use an array of germline-encoded activating and inhibitory receptors that scan for altered protein-expression patterns, but tumor evasion of detection by the immune system is now recognized as one of the hallmarks of cancer. NK cells display rapid and potent immunity to metastasis or hematological cancers, and major efforts are now being undertaken to fully exploit NK cell anti-tumor properties in the clinic. Diverse approaches encompass the development of large-scale NK cell–expansion protocols for adoptive transfer, the establishment of a microenvironment favorable to NK cell activity, the redirection of NK cell activity against tumor cells and the release of inhibitory signals that limit NK cell function. In this Review we detail recent advances in NK cell–based immunotherapies and discuss the advantages and limitations of these strategies.


Cell Host & Microbe | 2009

Humanized mice for modeling human infectious disease: challenges, progress, and outlook.

Nicolas Legrand; Alexander Ploss; Rudi Balling; Pablo D. Becker; Chiara Borsotti; Nicolas Brezillon; Jennifer Debarry; Ype P. de Jong; Hongkui Deng; James P. Di Santo; Stephanie C. Eisenbarth; Elizabeth E. Eynon; Richard A. Flavell; Carlos A. Guzmán; Nicholas D. Huntington; Dina Kremsdorf; Michael P. Manns; Markus G. Manz; Jean-Jacques Mention; Michael Ott; Chozhavendan Rathinam; Charles M. Rice; Anthony Rongvaux; Sean Stevens; Hergen Spits; Helene Strick-Marchand; Hitoshi Takizawa; Anja U. van Lent; Chengyan Wang; Kees Weijer

Over 800 million people worldwide are infected with hepatitis viruses, human immunodeficiency virus (HIV), and malaria, resulting in more than 5 million deaths annually. Here we discuss the potential and challenges of humanized mouse models for developing effective and affordable therapies and vaccines, which are desperately needed to combat these diseases.


Journal of Immunology | 2007

NK Cell Maturation and Peripheral Homeostasis Is Associated with KLRG1 Up-Regulation

Nicholas D. Huntington; Hy Tabarias; Kirsten Fairfax; Jason Brady; Yoshihiro Hayakawa; Mariapia A. Degli-Esposti; Mark J. Smyth; David M. Tarlinton; Stephen L. Nutt

NK cells are important for the clearance of tumors, parasites, and virus-infected cells. Thus, factors that control NK cell numbers and function are critical for the innate immune response. A subset of NK cells express the inhibitory killer cell lectin-like receptor G1 (KLRG1). In this study, we identify that KLRG1 expression is acquired during periods of NK cell division such as development and homeostatic proliferation. KLRG1+ NK cells are mature in phenotype, and we show for the first time that these cells have a slower in vivo turnover rate, reduced proliferative response to IL-15, and poorer homeostatic expansion potential compared with mature NK cells lacking KLRG1. Transfer into lymphopenic recipients indicate that KLRG1− NK cells are precursors of KLRG1+ NK cells and KLRG1 expression accumulates following cell division. Furthermore, KLRG1+ NK cells represent a significantly greater proportion of NK cells in mice with enhanced NK cell numbers such as Cd45−/− mice. These data indicate that NK cells acquire KLRG1 on their surface during development, and this expression correlates with functional distinctions from other peripheral NK cells in vivo.


Immunological Reviews | 2006

Functional subsets of mouse natural killer cells

Yoshihiro Hayakawa; Nicholas D. Huntington; Stephen L. Nutt; Mark J. Smyth

Summary:  Human natural killer (NK) cells can be divided into two phenotypically distinct functional subsets based on their cell surface expression of CD56 (CD56bright and CD56dim). As mouse NK cells do not express CD56, comparable mouse NK cell subsets have proven difficult to identify. Recently, we have found that mouse NK cells can be subdivided by the expression of CD27. The CD27hi and CD27lo mouse NK cell subsets show some intriguing similarities to but also some distinct differences from the human CD56 NK cell subsets in terms of their function and phenotype. Extending our knowledge of mature NK cell heterogeneity between the species will be critical to further our understanding of the pathological role of NK cells in immune responses.


Nature Immunology | 2007

Interleukin 15–mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1

Nicholas D. Huntington; Priscilla Gunn; Edwina Naik; Ewa M. Michalak; Mark J. Smyth; Hyacinth Tabarias; Mariapia A. Degli-Esposti; Grant Dewson; Simon N. Willis; Noboru Motoyama; David C. S. Huang; Stephen L. Nutt; David M. Tarlinton; Andreas Strasser

Interleukin 15 (IL-15) promotes the survival of natural killer (NK) cells by preventing apoptosis through mechanisms unknown at present. Here we identify Bim, Noxa and Mcl-1 as key regulators of IL-15-dependent survival of NK cells. IL-15 suppressed apoptosis by limiting Bim expression through the kinases Erk1 and Erk2 and mechanisms dependent on the transcription factor Foxo3a, while promoting expression of Mcl-1, which was necessary and sufficient for the survival of NK cells. Withdrawal of IL-15 led to upregulation of Bim and, accordingly, both Bim-deficient and Foxo3a−/− NK cells were resistant to cytokine deprivation. Finally, IL-15-mediated inactivation of Foxo3a and cell survival were dependent on phosphotidylinositol-3-OH kinase. Thus, IL-15 regulates the survival of NK cells at multiple steps, with Bim and Noxa being key antagonists of Mcl-1, the critical survivor factor in this process.


Journal of Experimental Medicine | 2014

Nfil3 is required for the development of all innate lymphoid cell subsets

Cyril Seillet; Lucille C. Rankin; Joanna R. Groom; Lisa A. Mielke; Julie Tellier; Michaël Chopin; Nicholas D. Huntington; Gabrielle T. Belz; Sebastian Carotta

Loss of Nfil3 selectively reduces Peyer’s patch formation, impairing recruitment and distribution of lymphocytes and compromising immune responses to inflammatory and infectious agents.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Characterization of the thymic IL-7 niche in vivo

Nuno L. Alves; Odile Richard-Le Goff; Nicholas D. Huntington; Ana Patricia Sousa; Vera S. G. Ribeiro; Allison Bordack; Francina Langa Vives; Lucie Peduto; Ann Patricia Chidgey; Ana Cumano; Richard L. Boyd; Gérard Eberl; James P. Di Santo

The thymus represents the “cradle” for T cell development, with thymic stroma providing multiple soluble and membrane cues to developing thymocytes. Although IL-7 is recognized as an essential factor for thymopoiesis, the “environmental niche” of thymic IL-7 activity remains poorly characterized in vivo. Using bacterial artificial chromosome transgenic mice in which YFP is under control of IL-7 promoter, we identify a subset of thymic epithelial cells (TECs) that co-express YFP and high levels of Il7 transcripts (IL-7hi cells). IL-7hi TECs arise during early fetal development, persist throughout life, and co-express homeostatic chemokines (Ccl19, Ccl25, Cxcl12) and cytokines (Il15) that are critical for normal thymopoiesis. In the adult thymus, IL-7hi cells localize to the cortico-medullary junction and display traits of both cortical and medullary TECs. Interestingly, the frequency of IL-7hi cells decreases with age, suggesting a mechanism for the age-related thymic involution that is associated with declining IL-7 levels. Our temporal-spatial analysis of IL-7-producing cells in the thymus in vivo suggests that thymic IL-7 levels are dynamically regulated under distinct physiological conditions. This IL-7 reporter mouse provides a valuable tool to further dissect the mechanisms that govern thymic IL-7 expression in vivo.


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

Functional CD47/signal regulatory protein alpha (SIRPα) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo

Nicolas Legrand; Nicholas D. Huntington; Maho Nagasawa; Arjen Q. Bakker; Remko Schotte; Helene Strick-Marchand; Sandra J. de Geus; Stephan M. Pouw; Martino Böhne; Arie Voordouw; Kees Weijer; James P. Di Santo; Hergen Spits

The homeostatic control mechanisms regulating human leukocyte numbers are poorly understood. Here, we assessed the role of phagocytes in this process using human immune system (HIS) BALB/c Rag2−/−IL-2Rγc−/− mice in which human leukocytes are generated from transplanted hematopoietic progenitor cells. Interactions between signal regulatory protein alpha (SIRPα; expressed on phagocytes) and CD47 (expressed on hematopoietic cells) negatively regulate phagocyte activity of macrophages and other phagocytic cells. We previously showed that B cells develop and survive robustly in HIS mice, whereas T and natural killer (NK) cells survive poorly. Because human CD47 does not interact with BALB/c mouse SIRPα, we introduced functional CD47/SIRPα interactions in HIS mice by transducing mouse CD47 into human progenitor cells. Here, we show that this procedure resulted in a dramatic and selective improvement of progenitor cell engraftment and human T- and NK-cell homeostasis in HIS mouse peripheral lymphoid organs. The amount of engrafted human B cells also increased but much less than that of T and NK cells, and total plasma IgM and IgG concentrations increased 68- and 35-fold, respectively. Whereas T cells exhibit an activated/memory phenotype in the absence of functional CD47/SIRPα interactions, human T cells accumulated as CD4+ or CD8+ single-positive, naive, resting T cells in the presence of functional CD47/SIRPα interactions. Thus, in addition to signals mediated by T cell receptor (TCR)/MHC and/or IL/IL receptor interactions, sensing of cell surface CD47 expression by phagocyte SIRPα is a critical determinant of T- and NK-cell homeostasis under steady-state conditions in vivo.

Collaboration


Dive into the Nicholas D. Huntington's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabrielle T. Belz

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Stephen L. Nutt

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sebastian Carotta

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jai Rautela

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca B. Delconte

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Wei Shi

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Eric Vivier

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge