Network


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

Hotspot


Dive into the research topics where Emma Teixeiro is active.

Publication


Featured researches published by Emma Teixeiro.


Nature | 2006

Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling

Mark A. Daniels; Emma Teixeiro; Jason Gill; Barbara Hausmann; Dominique Roubaty; Kaisa Holmberg; Guy Werlen; Georg A. Holländer; Nicholas R. J. Gascoigne; Ed Palmer

A healthy individual can mount an immune response to exogenous pathogens while avoiding an autoimmune attack on normal tissues. The ability to distinguish between self and non-self is called ‘immunological tolerance’ and, for T lymphocytes, involves the generation of a diverse pool of functional T cells through positive selection and the removal of overtly self-reactive thymocytes by negative selection during T-cell ontogeny. To elucidate how thymocytes arrive at these cell fate decisions, here we have identified ligands that define an extremely narrow gap spanning the threshold that distinguishes positive from negative selection. We show that, at the selection threshold, a small increase in ligand affinity for the T-cell antigen receptor leads to a marked change in the activation and subcellular localization of Ras and mitogen-activated protein kinase (MAPK) signalling intermediates and the induction of negative selection. The ability to compartmentalize signalling molecules differentially in the cell endows the thymocyte with the ability to convert a small change in analogue input (affinity) into a digital output (positive versus negative selection) and provides the basis for establishing central tolerance.


Science | 2009

Different T Cell Receptor Signals Determine CD8+ Memory Versus Effector Development

Emma Teixeiro; Mark A. Daniels; Sara E. Hamilton; Adam G. Schrum; Rafael Bragado; Stephen C. Jameson; Ed Palmer

Following infection, naïve CD8+ T cells bearing pathogen-specific T cell receptors (TCRs) differentiate into a mixed population of short-lived effector and long-lived memory T cells to mediate an adaptive immune response. How the TCR regulates memory T cell development has remained elusive. Using a mutant TCR transgenic model, we found that point mutations in the TCR β transmembrane domain (βTMD) impair the development and function of CD8+ memory T cells without affecting primary effector T cell responses. Mutant T cells are deficient in polarizing the TCR and in organizing the nuclear factor κB signal at the immunological synapse. Thus, effector and memory states of CD8+ T cells are separable fates, determined by differential TCR signaling.


Journal of Histochemistry and Cytochemistry | 2001

Localization of the Lipid Receptors CD36 and CLA-1/SR-BI in the Human Gastrointestinal Tract: Towards the Identification of Receptors Mediating the Intestinal Absorption of Dietary Lipids

Maria V.T. Lobo; Lydia Huerta; Natividad Ruiz–Velasco; Emma Teixeiro; Paloma de la Cueva; Angel Celdrán; Antonia Martín–Hidalgo; Miguel A. Vega; Rafael Bragado

The scavenger receptors CLA-1/SR-BI and CD36 interact with native and modified lipoproteins and with some anionic phospholipids. In addition, CD36 binds/transports long-chain free fatty acids. Recent biochemical evidences indicates that the rabbit CLA-1/SR-BI receptor can be detected in enterocytes, and previous studies showed the presence of mRNA for both CLA-1/SR-BI and CD36 in some segments of the intestinal tract. These findings prompted us to study their respective localization and distribution from the human stomach to the colorectal segments, using immunohistochemical methods. Their expression in the colorectal carcinoma-derived cell line Caco-2 was analyzed by Northern blotting. In the human intestinal tract, CLA-1/SR-BI was found in the brush-border membrane of enterocytes from the duodenum to the rectum. However, CD36 was found only in the duodenal and jejunal epithelium, whereas enterocytes from other intestinal segments were not stained. In the duodenum and jejunum, CD36 co-localized with CLA-1/SR-BI in the apical membrane of enterocytes. The gastric epithelium was immunonegative for both glycoproteins. We also found that CLA-1/SR-BI mRNA was expressed in Caco-2 cells and that its expression levels increased concomitantly with their differentiation. In contrast, the CD36 transcript was not found in this colon cell line, in agreement with the absence of this protein in colon epithelium. The specific localization of CLA-1/SR-BI and CD36 along the human gastrointestinal tract and their ability to interact with a large variety of lipids strongly support a physiological role for them in absorption of dietary lipids.


FEBS Journal | 2010

ERK and cell death: ERK location and T cell selection

Emma Teixeiro; Mark A. Daniels

The selection of functional T cells is mediated by interactions between the T cell antigen receptor and self‐peptide major histocompatibility complex expressed on thymic epithelium. These interactions either lead to survival and development or death. The T cell antigen receptor is an unusual receptor able to signal multiple cell fates. The precise mechanism by which this is achieved has been an area of intense research effort over the years. One model proposes that the differential activation of mitogen‐activated protein kinase pathways contributes to this decision. Here, the role of extracellular signal‐regulated kinase in promoting or preventing apoptosis during thymic selection is discussed.


Cell Reports | 2013

Low-Affinity T Cells Are Programmed to Maintain Normal Primary Responses but Are Impaired in Their Recall to Low-Affinity Ligands

Karin M. Knudson; Nicholas Goplen; Cody A. Cunningham; Mark A. Daniels; Emma Teixeiro

T cell responses to low-affinity T cell receptor (TCR) ligands occur in the context of infection, tumors, and autoimmunity despite diminished TCR signal strength. The processes that enable such responses remain unclear. We show that distinct mechanisms drive effector/memory development in high- and low-affinity T cells. Low-affinity cells preferentially differentiate into memory precursors of a central memory phenotype that are interleukin (IL)-12R(lo), IL-7R(hi), and Eomes(hi). Strikingly, in contrast to naive cells, low-affinity memory cells were impaired in the response to low- but not high-affinity ligands, indicating that low-affinity cells are programmed to generate diverse immune responses while avoiding autoreactivity. Affinity and antigen dose directly correlated with IL-12R signal input and T-bet but not with Eomes expression because low- affinity signals were more potent inducers of Eomes at a high antigen dose. Our studies explain how weak antigenic signals induce complete primary immune responses and provide a framework for therapeutic intervention.


Frontiers in Immunology | 2015

TCR Signaling in T Cell Memory

Mark A. Daniels; Emma Teixeiro

T cell memory plays a critical role in our protection against pathogens and tumors. The antigen and its interaction with the T cell receptor (TCR) is one of the initiating elements that shape T cell memory together with inflammation and costimulation. Over the last decade, several transcription factors and signaling pathways that support memory programing have been identified. However, how TCR signals regulate them is still poorly understood. Recent studies have shown that the biochemical rules that govern T cell memory, strikingly, change depending on the TCR signal strength. Furthermore, TCR signal strength regulates the input of cytokine signaling, including pro-inflammatory cytokines. These highlight how tailoring antigenic signals can improve immune therapeutics. In this review, we focus on how TCR signaling regulates T cell memory and how the quantity and quality of TCR–peptide–MHC interactions impact the multiple fates a T cell can adopt in the memory pool.


Journal of Immunology | 2013

Cutting Edge: The Signals for the Generation of T Cell Memory Are Qualitatively Different Depending on TCR Ligand Strength

Karin M. Knudson; Sara E. Hamilton; Mark A. Daniels; Stephen C. Jameson; Emma Teixeiro

CD8 T cell memory critically contributes to long-term immunity. Both low- and high-affinity TCR signals are able to support the differentiation of memory CD8 T cells. However, it is unclear whether the requirements for memory development change when TCR signal strength is altered. To gain further insight into this question, we used a TCRβ transmembrane domain mutant model that is defective in the generation of memory in response to high-affinity ligands. Surprisingly, lowering TCR signal strength, by stimulation with low-affinity ligands, resulted in normal memory development. Restoration of memory correlated with recovery of TCR-dependent NF-κB signaling. Thus, these data provide novel evidence that the requirements for memory are qualitatively different depending on TCR signal strength.


Cellular and Molecular Life Sciences | 2010

The persistence of T cell memory

Mark A. Daniels; Emma Teixeiro

T cell memory is a crucial feature of the adaptive immune system in the defense against pathogens. During the last years, numerous studies have focused their efforts on uncovering the signals, inflammatory cues, and extracellular factors that support memory differentiation. This research is beginning to decipher the complex gene network that controls memory programming. However, how the different signals, that a T cell receives during the process of differentiation, interplay to trigger memory programming is still poorly defined. In this review, we focus on the most recent advances in the field and discuss how T cell receptor signaling and inflammation control CD8 memory differentiation.


Proceedings of the National Academy of Sciences of the United States of America | 2017

NFκB–Pim-1–Eomesodermin axis is critical for maintaining CD8 T-cell memory quality

Karin M. Knudson; Curtis J. Pritzl; Vikas Saxena; Amnon Altman; Mark A. Daniels; Emma Teixeiro

Significance Mice and humans whose T cells are deficient in NFκB signaling lack memory T cells, but the mechanism behind this is unclear. We found that NFκB signaling is required during the resolution phase of the immune response to maintain long-term CD8 memory. NFκB signaling is necessary for preserving expression of Eomesodermin and prosurvival Bcl-2 in memory T cells, in a cell-intrinsic process where T-cell receptor (TCR) signals and Pim-1 kinase are involved. Our study defines an unexpected role of NFκB and Pim-1 signaling in the maintenance of T-cell memory quality. Furthermore, it identifies targets and specific times of intervention where protective T-cell memory could be reinforced in vaccines and cancer immunotherapies by manipulation of the NFκB–Pim-1–Eomesodermin axis. T-cell memory is critical for long-term immunity. However, the factors involved in maintaining the persistence, function, and phenotype of the memory pool are undefined. Eomesodermin (Eomes) is required for the establishment of the memory pool. Here, we show that in T cells transitioning to memory, the expression of high levels of Eomes is not constitutive but rather requires a continuum of cell-intrinsic NFκB signaling. Failure to maintain NFκB signals after the peak of the response led to impaired Eomes expression and a defect in the maintenance of CD8 T-cell memory. Strikingly, we found that antigen receptor [T-cell receptor (TCR)] signaling regulates this process through expression of the NFκB-dependent kinase proviral integration site for Moloney murine leukemia virus-1 (PIM-1), which in turn regulates NFκB and Eomes. T cells defective in TCR-dependent NFκB signaling were impaired in late expression of Pim-1, Eomes, and CD8 memory. These defects were rescued when TCR-dependent NFκB signaling was restored. We also found that NFκB–Pim-1 signals were required at memory to maintain memory CD8 T-cell longevity, effector function, and Eomes expression. Hence, an NFκB–Pim-1–Eomes axis regulates Eomes levels to maintain memory fitness.


Journal of Immunology | 2016

IL-12 Signals through the TCR To Support CD8 Innate Immune Responses

Nicholas Goplen; Vikas Saxena; Karin M. Knudson; Adam G. Schrum; Diana Gil; Mark A. Daniels; Rose Zamoyska; Emma Teixeiro

CD8 T cells must integrate antigenic and inflammatory signals to differentiate into efficient effector and memory T cells able to protect us from infections. The mechanisms by which TCR signaling and proinflammatory cytokine receptor signaling cooperate in these processes are poorly defined. In this study, we show that IL-12 and other proinflammatory cytokines transduce signals through the TCR signalosome in a manner that requires Fyn activity and self-peptide–MHC (self-pMHC) interactions. This mechanism is crucial for CD8 innate T cell functions. Loss of Fyn activity or blockade of self-pMHC interactions severely impaired CD8 T cell IFN-γ and NKG2D expression, proliferation, and cytotoxicity upon cytokine-mediated bystander activation. Most importantly, in the absence of self-pMHC interactions, CD8 memory T cells fail to undergo bystander activation upon an unrelated infection. Thus, CD8 T cell bystander activation, although independent of cognate Ag, still requires self-pMHC and TCR signaling.

Collaboration


Dive into the Emma Teixeiro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rafael Bragado

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason Gill

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kaisa Holmberg

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge