Network


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

Hotspot


Dive into the research topics where Amanda M. Schmidt is active.

Publication


Featured researches published by Amanda M. Schmidt.


Science Signaling | 2013

The ζ Isoform of Diacylglycerol Kinase Plays a Predominant Role in Regulatory T Cell Development and TCR-Mediated Ras Signaling

Rohan P. Joshi; Amanda M. Schmidt; Jayajit Das; Dariusz Pytel; Matthew J. Riese; Lester M; J. A. Diehl; Edward M. Behrens; Taku Kambayashi; Gary A. Koretzky

Despite its relatively low abundance, DGKζ exerts a major inhibitory effect on T cell receptor signaling. Getting More Activity for Less DGK Strong antigen-dependent activation of the T cell receptor (TCR) in thymocytes leads to the development of natural regulatory T (nTreg) cells through a pathway involving diacylglycerol (DAG). Metabolism of DAG by diacylglycerol kinases (DGKs) to generate phosphatidic acid (PA) limits DAG signaling. In the second of a pair of papers, Joshi et al. characterized the relative contributions of DGKα and DGKζ to TCR-dependent signaling. Although DGKα was more abundant than DGKζ in the cytosol and at the interface between T cells and antigen-presenting cells, only loss of DGKζ enhanced TCR signaling and increased generation of nTreg cells in mice. Mathematical modeling suggested that these results might be explained by the differential catalytic activities of the DGK isoforms, and experiments in T cells showed that DGKζ produced the greatest amounts of PA. Together, these results suggest that the relatively low abundance of DGKζ in T cells belies its importance in inhibiting TCR signaling. Diacylglycerol (DAG) is a critical second messenger that mediates T cell receptor (TCR)–stimulated signaling. The abundance of DAG is reduced by the diacylglycerol kinases (DGKs), which catalyze the conversion of DAG to phosphatidic acid (PA) and thus inhibit DAG-mediated signaling. In T cells, the predominant DGK isoforms are DGKα and DGKζ, and deletion of the genes encoding either isoform enhances DAG-mediated signaling. We found that DGKζ, but not DGKα, suppressed the development of natural regulatory T (Treg) cells and predominantly mediated Ras and Akt signaling downstream of the TCR. The differential functions of DGKα and DGKζ were not attributable to differences in protein abundance in T cells or in their localization to the contact sites between T cells and antigen-presenting cells. RasGRP1, a key DAG-mediated activator of Ras signaling, associated to a greater extent with DGKζ than with DGKα; however, in silico modeling of TCR-stimulated Ras activation suggested that a difference in RasGRP1 binding affinity was not sufficient to cause differences in the functions of each DGK isoform. Rather, the model suggested that a greater catalytic rate for DGKζ than for DGKα might lead to DGKζ exhibiting increased suppression of Ras-mediated signals compared to DGKα. Consistent with this notion, experimental studies demonstrated that DGKζ was more effective than DGKα at catalyzing the metabolism of DAG to PA after TCR stimulation. The enhanced effective enzymatic production of PA by DGKζ is therefore one possible mechanism underlying the dominant functions of DGKζ in modulating Treg cell development.


Journal of Immunology | 2012

Cutting Edge: IL-2 Signals Determine the Degree of TCR Signaling Necessary To Support Regulatory T Cell Proliferation In Vivo

Tao Zou; Atsushi Satake; Evann Corbo-Rodgers; Amanda M. Schmidt; Michael A. Farrar; Jonathan S. Maltzman; Taku Kambayashi

To ensure immune tolerance, regulatory T cell (Treg) numbers must be maintained by cell division. This process has been thought to be strictly dependent on the Treg TCR interacting with MHC class II. In this study, we report that Treg division does not absolutely require cell-autonomous TCR signaling in vivo, depending on the degree of IL-2–mediated stimulation provided. At steady state IL-2 levels, Tregs require cell-autonomous TCR signaling to divide. However, when given exogenous IL-2 or when STAT5 is selectively activated in Tregs, Treg division can occur independently of MHC class II and TCR signaling. Thus, depending on the amount of IL-2R stimulation, a wide range of TCR signals supports Treg division, which may contribute to preservation of a diverse repertoire of Treg TCR specificities. These findings also have therapeutic implications, as TCR signaling by Tregs may not be required when using IL-2 to increase Treg numbers for treatment of inflammatory disorders.


Blood | 2012

Cell-autonomous role of TGFβ and IL-2 receptors in CD4+ and CD8+ inducible regulatory T-cell generation during GVHD

Norifumi Sawamukai; Atsushi Satake; Amanda M. Schmidt; Ian T. Lamborn; Priti Ojha; Yoshiya Tanaka; Taku Kambayashi

FoxP3(+) regulatory T cells (Tregs) suppress GVHD while preserving graft-versus-tumor effects, making them an attractive target for GVHD therapy. The donor-derived Treg pool can potentially be derived from the expansion of preexisting natural Tregs (nTregs) or from de novo generation of inducible Tregs (iTregs) from donor Tconvs in the transplantation recipient. Using an MHC-mismatched model of acute GVHD, in the present study we found that the Treg pool was comprised equally of donor-derived nTregs and iTregs. Experiments using various combinations of T cells from wild-type and FoxP3-deficient mice suggested that both preexisting donor nTregs and the generation of iTregs in the recipient mice contribute to protection against GVHD. Surprisingly, CD8(+)FoxP3(+) T cells represented approximately 70% of the iTreg pool. These CD8(+)FoxP3(+) T cells shared phenotypic markers with their CD4(+) counterparts and displayed suppressive activity, suggesting that they were bona fide iTregs. Both CD4(+) and CD8(+) Tregs appeared to be protective against GVHD-induced lethality and required IL-2 and TGFβ receptor expression for their generation. These data illustrate the complex makeup of the donor-derived FoxP3(+) Treg pool in allogeneic recipients and their potential role in protection against GVHD.


Journal of Immunology | 2015

Regulatory T Cells Require TCR Signaling for Their Suppressive Function

Amanda M. Schmidt; Wen Lu; Vishal J. Sindhava; Yanping Huang; Janis K. Burkhardt; Enjun Yang; Matthew J. Riese; Jonathan S. Maltzman; Martha S. Jordan; Taku Kambayashi

Regulatory T cells (Tregs) are a subset of CD4+ T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4+ T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase–like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.


PLOS ONE | 2014

Inhibition of calcineurin abrogates while inhibition of mTOR promotes regulatory T cell expansion and graft-versus-host disease protection by IL-2 in allogeneic bone marrow transplantation.

Atsushi Satake; Amanda M. Schmidt; Shosaku Nomura; Taku Kambayashi

Regulatory T cells (Treg)s attenuate excessive immune responses, making their expansion beneficial in immune-mediated diseases including allogeneic bone marrow transplantation (BMT)-associated graft-versus-host disease (GVHD). We have recently reported that Treg expansion does not require phospholipase Cγ activation when IL-2 is provided. As such, the combination of IL-2 and a calcineurin inhibitor (Cyclosporine A; CsA) expands Tregs while inhibiting Tconv proliferation and protects against a mouse model of multiple sclerosis. However, CsA inhibits Treg proliferation in the presence of a TCR stimulus, suggesting that CsA may negatively impact Treg proliferation when they receive strong allogeneic MHC-mediated TCR signals. In this study, we show that CsA inhibits Treg proliferation and inducible Treg generation in allogeneic but not in syngeneic BMT when IL-2 is provided. In contrast to CsA, the mTOR inhibitor (Rapamycin) almost completely suppressed IL-2-mediated Treg proliferation. However, CsA and Rapamycin inhibited Treg proliferation to a similar extent when TCR stimulation was provided. Furthermore, Rapamycin promoted Treg expansion and inducible Treg generation in allogeneic BMT recipients treated with IL-2. Consistent with these observations, CsA abrogated while Rapamycin promoted the protective effect of IL-2 on allogeneic BMT-induced GVHD. These results suggest that while CsA permits IL-2-induced Treg proliferation in the syngeneic setting (absence of strong TCR signals), CsA in combination with IL-2 may be detrimental for Treg proliferation in an allogeneic setting. Thus, in allogeneic settings, an mTOR inhibitor such as Rapamycin is a better choice for adjunct therapy with IL-2 in expansion of Tregs and protection against allogeneic BMT-induced GVHD.


Science Signaling | 2013

Diacylglycerol Kinase ζ Limits the Generation of Natural Regulatory T Cells

Amanda M. Schmidt; Tao Zou; Rohan P. Joshi; Theresa M. Leichner; Matthew A. Pimentel; Connie L. Sommers; Taku Kambayashi

Increasing the activation of extracellular signal–regulated kinase promotes the generation of natural regulatory T cells. ERK Signaling Promotes Treg Cells When T cell precursors (thymocytes) in the thymus receive a strong signal through their T cell receptor (TCR), they develop into natural regulatory T (nTreg) cells. TCR stimulation leads to the activation of phospholipase C–γ1 (PLC-γ1) and the production of the second messenger diacylglycerol (DAG), which, in turn, activates extracellular signal–regulated kinase (ERK) signaling. In one of a pair of papers published this week, Schmidt et al. investigated nTreg cell development in mice deficient in the ζ isoform of DAG kinase (DGKζ), an enzyme that converts DAG to phosphatidic acid, thus inhibiting DAG-dependent signaling. Compared to wild-type mice, DGKζ-deficient mice generated increased numbers of nTreg cells and their precursors. The accumulation of DAG in DGKζ-deficient thymocytes resulted in enhanced ERK activation, as well as of nuclear factor κB, and the authors found a positive correlation between the extent of ERK activation and the numbers of nTreg cells produced in vivo. Together, these data suggest that DGKζ inhibits nTreg cell generation by limiting the extent to which TCR stimulation activates ERK. Natural regulatory T (nTreg) cells are important for maintaining tolerance to self- and foreign antigens, and they are thought to develop from thymocytes that receive strong T cell receptor (TCR)–mediated signals in the thymus. TCR engagement leads to the activation of phospholipase C–γ1, which generates the lipid second messenger diacylglycerol (DAG) from phosphatidylinositol 4,5-bisphosphate. We used mice that lack the ζ isoform of DAG kinase (DGKζ), which metabolizes DAG to terminate its signaling, to enhance TCR-mediated signaling and identify critical signaling events in nTreg cell development. Loss of DGKζ resulted in increased numbers of thymic CD25+Foxp3−CD4+ nTreg cell precursors and Foxp3+CD4+ nTreg cells in a cell-autonomous manner. DGKζ-deficient T cells exhibited increased nuclear translocation of the nuclear factor κB subunit c-Rel, as well as enhanced extracellular signal–regulated kinase (ERK) phosphorylation in response to TCR stimulation, suggesting that these downstream pathways may contribute to nTreg cell development. Indeed, reducing c-Rel abundance or blocking ERK phosphorylation abrogated the increased generation of nTreg cells by DGKζ-deficient thymocytes. The extent of ERK phosphorylation correlated with TCR-mediated acquisition of Foxp3 in immature thymocytes in vitro. Furthermore, the development of nTreg cells was augmented in mice in which ERK activation was selectively enhanced in T cells. Together, these data suggest that DGKζ regulates the development of nTreg cells by limiting the extent of activation of the ERK and c-Rel signaling pathways.


Journal of Clinical Investigation | 2017

A TLR9-dependent checkpoint governs B cell responses to DNA-containing antigens

Vishal J. Sindhava; Michael A. Oropallo; Krishna Moody; Martin S. Naradikian; Lauren E. Higdon; Lin Zhou; Arpita Myles; Nathaniel M. Green; Kerstin Nundel; William Stohl; Amanda M. Schmidt; Wei Cao; Stephanie Dorta-Estremera; Taku Kambayashi; Ann Marshak-Rothstein; Michael P. Cancro

Mature B cell pools retain a substantial proportion of polyreactive and self-reactive clonotypes, suggesting that activation checkpoints exist to reduce the initiation of autoreactive B cell responses. Here, we have described a relationship among the B cell receptor (BCR), TLR9, and cytokine signals that regulate B cell responses to DNA-containing antigens. In both mouse and human B cells, BCR ligands that deliver a TLR9 agonist induce an initial proliferative burst that is followed by apoptotic death. The latter mechanism involves p38-dependent G1 cell-cycle arrest and subsequent intrinsic mitochondrial apoptosis and is shared by all preimmune murine B cell subsets and CD27– human B cells. Survival or costimulatory signals rescue B cells from this fate, but the outcome varies depending on the signals involved. B lymphocyte stimulator (BLyS) engenders survival and antibody secretion, whereas CD40 costimulation with IL-21 or IFN-&ggr; promotes a T-bet+ B cell phenotype. Finally, in vivo immunization studies revealed that when protein antigens are conjugated with DNA, the humoral immune response is blunted and acquires features associated with T-bet+ B cell differentiation. We propose that this mechanism integrating BCR, TLR9, and cytokine signals provides a peripheral checkpoint for DNA-containing antigens that, if circumvented by survival and differentiative cues, yields B cells with the autoimmune-associated T-bet+ phenotype.


Journal of Leukocyte Biology | 2015

TCR signaling intensity controls CD8+ T cell responsiveness to TGF-β

Vidhyalakshmi Arumugam; Theresa Bluemn; Erin Wesley; Amanda M. Schmidt; Taku Kambayashi; Subramaniam Malarkannan; Matthew J. Riese

DGK‐ζ is a negative regulator of TCR signaling that causes degradation of the second messenger DAG, terminating DAG‐mediated activation of Ras and PKCθ. Cytotoxic T cells deficient in DGK‐ζ demonstrate enhanced effector functions in vitro and antitumor activity in vivo, perhaps because of insensitivity to inhibitory cytokines. We sought to determine whether the enhanced responsiveness of DGK‐ζ‐deficient T cells renders them insensitive to the inhibitory cytokine TGF‐β and to determine how the loss of DGK‐ζ facilitates this insensitivity. We identified decreased transcriptional and functional responses to TGF‐β in CD8+ DGK‐ζ−/− T cells but preserved TGF‐β‐mediated conversion of naϊve DGK‐ζ−/− CD4+ T cells to a regulatory T cell phenotype. Decreased CD8+ T cell responsiveness to TGF‐β did not result from impaired canonical TGF‐β signal transduction, because similar levels of TGF‐β‐R and intracellular Smad components were identified in WT and DGK‐ζ−/− CD8+ T cells, and TGF‐β‐mediated activation of Smad2 was unchanged. Instead, an enhanced TCR signal strength was responsible for TGF‐β insensitivity, because (i) loss of DGK‐ζ conferred resistance to TGF‐β‐mediated inhibition of Erk phosphorylation, (ii) TGF‐β insensitivity could be recapitulated by exogenous addition of the DAG analog PMA, and (iii) TGF‐β sensitivity could be observed in DGK‐ζ‐deficient T cells at limiting dilutions of TCR stimulation. These data indicate that enhanced TCR signal transduction in the absence of DGK‐ζ makes T cells relatively insensitive to TGF‐β, in a manner independent of Smads, a finding with practical implications in the development of immunotherapies that target TGF‐β.


Journal of Autoimmunity | 2013

Differential targeting of IL-2 and T cell receptor signaling pathways selectively expands regulatory T cells while inhibiting conventional T cells

Atsushi Satake; Amanda M. Schmidt; Angela S. Archambault; Theresa M. Leichner; Gregory F. Wu; Taku Kambayashi


Archive | 2014

Regulatory T Cell Development from the Top Down: the Role of T Cell Receptor-Generated Second Messengers in Thymic Regulatory T Cell Development

Taku Kambayashi; Amanda M. Schmidt

Collaboration


Dive into the Amanda M. Schmidt's collaboration.

Top Co-Authors

Avatar

Taku Kambayashi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Atsushi Satake

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Riese

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Ian T. Lamborn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Priti Ojha

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rohan P. Joshi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Tao Zou

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge