Network


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

Hotspot


Dive into the research topics where Brian C. Schaefer is active.

Publication


Featured researches published by Brian C. Schaefer.


Nature Immunology | 2002

T cells down-modulate peptide-MHC complexes on APCs in vivo

Ross M. Kedl; Brian C. Schaefer; John W. Kappler; Philippa Marrack

T cells compete in the response to antigen in vivo and this competition may drive the affinity maturation of a secondary T cell response. Here we show that high-affinity T cells out-competed lower affinity T cells during a response to antigenic challenge in vivo. Although competition between T cells specific for different peptide–major histocompatibility complexes (MHC) occurred, it was less efficient than competition between T cells of the same peptide-MHC specificity. In addition, high-affinity T cells efficiently induced antigen loss from the surface of antigen-presenting cells. Thus T cells that responded to the same peptide-MHC competed with each other by lowering the amount of ligand with which the cells could react. As a result, the activation of high-affinity cells was favored. This provides a mechanism for the affinity maturation of a secondary T cell response.


Trends in Immunology | 2013

A new look at T cell receptor signaling to nuclear factor-κB

Suman Paul; Brian C. Schaefer

Antigen stimulation of T cell receptor (TCR) signaling to nuclear factor (NF)-κB is required for T cell proliferation and differentiation of effector cells. The TCR-to-NF-κB pathway is generally viewed as a linear sequence of events in which TCR engagement triggers a cytoplasmic cascade of protein-protein interactions and post-translational modifications, ultimately culminating in the nuclear translocation of NF-κB. However, recent findings suggest a more complex picture in which distinct signalosomes, previously unrecognized proteins, and newly identified regulatory mechanisms play key roles in signal transmission. In this review, we evaluate recent data and suggest areas of future emphasis in the study of this important pathway.


Journal of Immunology | 2010

Cutting Edge: TCR Ligation Triggers Digital Activation of NF-κB

Lara M. Kingeter; Suman Paul; Sean K. Maynard; Natalia G. Cartwright; Brian C. Schaefer

TCR-mediated activation of the transcription factor NF-κB is required for T cell proliferation, survival, and effector differentiation. Although this pathway is the subject of intense study, it is not known whether TCR signaling to NF-κB is digital (switch-like) or analog in nature. Through analysis of the phosphorylation and degradation of IκBα and the nuclear translocation and phosphorylation of the NF-κB subunit RelA, we show that TCR-directed NF-κB activation is digital. Furthermore, digitization occurs well upstream of the IκB kinase complex, as protein kinase C θ translocation to the immunologic synapse and activation-associated aggregation of Bcl10 and Malt1 also demonstrate both digital behavior and high correlation with RelA nuclear translocation. Thus, similar to the TCR-to-MAPK signaling cascade, analog Ag inputs are converted to digital activation outputs to NF-κB at an early step downstream of TCR ligation.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Cell surface expression of the HIV-1 envelope glycoproteins is directed from intracellular CTLA-4-containing regulated secretory granules

Luis R. Miranda; Brian C. Schaefer; Abraham Kupfer; Zixin Hu; Alex Franzusoff

The envelope glycoprotein (Env) of HIV-1 is incorporated into virions that bud from the cell surface of infected T cells. With immunofluorescence microscopy and subcellular membrane fractionation techniques, the intracellular fate of Env in the secretory pathway of HIV-1-infected T cells was evaluated. Rather than trafficking constitutively from the Golgi to the cell surface, Env is directed to intracellular CTLA-4-containing granules, whose recruitment to the cell surface is regulated. The use of the regulated pathway for intracellular Env storage before virion assembly holds implications for the staging of Env exposure at the cell surface of infected cells and of coordinating HIV virion assembly.


Frontiers in Neurology | 2012

Controlled Cortical Impact and Craniotomy Induce Strikingly Similar Profiles of Inflammatory Gene Expression, but with Distinct Kinetics

Mouna Lagraoui; Joseph R. Latoche; Natalia G. Cartwright; Gauthaman Sukumar; Clifton L. Dalgard; Brian C. Schaefer

An immediate consequence of traumatic brain injury (TBI) is the induction of an inflammatory response. Mounting data suggest that inflammation is a major contributor to TBI-induced brain damage. However, much remains unknown regarding the induction and regulation of the inflammatory response to TBI. In this study we compared the TBI-induced inflammatory response to severe parenchymal injury (controlled cortical impact) vs. mild brain injury (craniotomy) over a 21-day period. Our data show that both severe and mild brain injury induce a qualitatively similar inflammatory response, involving highly overlapping sets of effector molecules. However, kinetic analysis revealed that the inflammatory response to mild brain injury is of much shorter duration than the response to severe TBI. Specifically, the inflammatory response to severe brain injury persists for at least 21 days, whereas the response to mild brain injury returns to near baseline values within 10 days post-injury. Our data therefore imply that the development of accurate diagnostic tests of TBI severity that are based on imaging or biomarker analysis of the inflammatory response may require repeated measures over at least a 10-day period, post-injury.


Journal of Immunology | 2008

Loss of protein kinase C theta, Bcl10, or Malt1 selectively impairs proliferation and NF-kappa B activation in the CD4+ T cell subset.

Lara M. Kingeter; Brian C. Schaefer

The cytosolic proteins protein kinase Cθ (PKCθ), Bcl10, and Malt1 play critical roles in TCR signaling to the transcription factor NF-κB. Our data confirm that CD4+ T cells from PKCθ, Bcl10, and Malt1 knockout mice show severe impairment of proliferation in response to TCR stimulation. Unexpectedly, we find that knockout CD8+ T cells proliferate to a similar extent as wild-type cells in response to strong TCR signals, although a survival defect prevents their accumulation. Both CD4+ and CD8+ knockout T cells express activation markers, including CD25, following TCR stimulation. Addition of exogenous IL-2 rescues survival of knockout CD4+ and CD8+ T cells, but fails to overcome the proliferation defect of CD4+ T cells. CD4+ T cells from knockout mice are extremely deficient in TCR-induced NF-κB activation, whereas NF-κB activation is only partially impaired in CD8+ T cells. Overall, our results suggest that defects in TCR signaling through PKCθ, Bcl10, and Malt1 predominantly impair NF-κB activation and downstream functional responses of CD4+ T cells. In contrast, CD8+ T cells maintain substantial NF-κB signaling, implying the existence of a significant TCR-regulated NF-κB activation pathway in CD8+ T cells that is independent of PKCθ, Bcl10, and Malt1.


Journal of Biological Chemistry | 2008

Multiple Protein Domains Mediate Interaction between Bcl10 and MALT1

Felicia D. Langel; Nidhi A. Jain; Jeremy S. Rossman; Llara M. Kingeter; Anuj K. Kashyap; Brian C. Schaefer

Bcl10 and MALT1 are essential mediators of NF-κB activation in response to the triggering of a diverse array of transmembrane receptors, including antigen receptors. Additionally, both proteins are translocation targets in MALT lymphoma. Thus, a detailed understanding of the interaction between these mediators is of considerable biological importance. Previous studies have indicated that a 13-amino acid region downstream of the Bcl10 caspase recruitment domain (CARD) is responsible for interacting with the immunoglobulin-like domains of MALT1. We now provide evidence that the death domain of MALT1 and the CARD of Bcl10 also contribute to Bcl10-MALT1 interactions. Although a direct interaction between the MALT1 death domain and Bcl10 cannot be detected via immunoprecipitation, FRET data strongly suggest that the death domain of MALT1 contributes significantly to the association between Bcl10 and MALT1 in T cells in vivo. Furthermore, analysis of point mutants of conserved residues of Bcl10 shows that the Bcl10 CARD is essential for interaction with the MALT1 N terminus. Mutations that disrupt proper folding of the Bcl10 CARD strongly impair Bcl10-MALT1 interactions. Molecular modeling and functional analyses of Bcl10 point mutants suggest that residues Asp80 and Glu84 of helix 5 of the Bcl10 CARD directly contact MALT1. Together, these data demonstrate that the association between Bcl10 and MALT1 involves a complex interaction between multiple protein domains. Moreover, the Bcl10-MALT1 interaction is the second reported example of interactions between a CARD and a non-CARD protein region, which suggests that many signaling cascades may utilize CARD interactions with non-CARD domains.


PLOS Pathogens | 2010

Blood Fluke Exploitation of Non-Cognate CD4+ T Cell Help to Facilitate Parasite Development

Erika W. Lamb; Colleen D. Walls; John T. Pesce; Diana K. Riner; Sean K. Maynard; Emily T. Crow; Thomas A. Wynn; Brian C. Schaefer; Stephen J. Davies

Schistosoma blood flukes, which infect over 200 million people globally, co-opt CD4+ T cell-dependent mechanisms to facilitate parasite development and egg excretion. The latter requires Th2 responses, while the mechanism underpinning the former has remained obscure. Using mice that are either defective in T cell receptor (TCR) signaling or that lack TCRs that can respond to schistosomes, we show that naïve CD4+ T cells facilitate schistosome development in the absence of T cell receptor signaling. Concurrently, the presence of naïve CD4+ T cells correlates with both steady-state changes in the expression of genes that are critical for the development of monocytes and macrophages and with significant changes in the composition of peripheral mononuclear phagocyte populations. Finally, we show that direct stimulation of the mononuclear phagocyte system restores blood fluke development in the absence of CD4+ T cells. Thus we conclude that schistosomes co-opt innate immune signals to facilitate their development and that the role of CD4+ T cells in this process may be limited to the provision of non-cognate help for mononuclear phagocyte function. Our findings have significance for understanding interactions between schistosomiasis and other co-infections, such as bacterial infections and human immunodeficiency virus infection, which potently stimulate innate responses or interfere with T cell help, respectively. An understanding of immunological factors that either promote or inhibit schistosome development may be valuable in guiding the development of efficacious new therapies and vaccines for schistosomiasis.


Cellular Signalling | 2010

Malt1 and cIAP2-Malt1 as effectors of NF-κB activation: Kissing cousins or distant relatives?

Lara M. Kingeter; Brian C. Schaefer

Malt1 is a multi-domain cytosolic signaling molecule that was originally identified as the target of recurrent translocations in a large fraction of MALT lymphomas. The product of this translocation is a chimeric protein in which the N-terminus is contributed by the apoptosis inhibitor, cIAP2, and the C-terminus is contributed by Malt1. Early studies suggested that Malt1 is an essential intermediate in antigen receptor activation of NF-kappaB, and that the juxtaposition of the cIAP2 N-terminus and the Malt1 C-terminus results in deregulation of Malt1 NF-kappaB stimulatory activity. Initial experimental data further suggested that the molecular mechanisms of Malt1- and cIAP-Malt1-mediated NF-kappaB activation were quite similar. However, a number of more recent studies of both Malt1 and cIAP2-Malt1 now reveal that these proteins influence NF-kappaB activation by multiple distinct mechanisms, several of which are non-overlapping. Currently available data suggest a revised model in which cIAP2-Malt1 induces NF-kappaB activation via a mechanism that depends equally on domains contributed by cIAP2 and Malt1, which confer spontaneous oligomerization activity, polyubiquitin binding, proteolytic activity, and association with and activation of TRAF2 and TRAF6 at several independent binding sites. By contrast, emerging data suggest that the wild-type Malt1 protein uniquely contributes to NF-kappaB activation primarily through the control of two proteolytic cleavage mechanisms. Firstly, Malt1 directly cleaves and inactivates A20, a negative regulator of the antigen receptor-to-NF-kappaB pathway. Secondly, Malt1 interacts with caspase-8, inducing caspase-8 cleavage of c-FLIP(L), initiating a pathway that contributes to activation of the I kappaB kinase (IKK) complex. Furthermore, data suggest that Malt1 plays a more limited and focused role in antigen receptor activation of NF-kappaB, serving to augment weak antigen signals and stimulate a defined subset of NF-kappaB dependent responses. Thus, the potent activation of NF-kappaB by cIAP2-Malt1 contrasts with the more subtle role of Malt1 in regulating specific NF-kappaB responses downstream of antigen receptor ligation.


Science Signaling | 2014

T cell receptor signals to NF-κB are transmitted by a cytosolic p62-Bcl10-Malt1-IKK signalosome.

Suman Paul; Traver Mk; Anuj K. Kashyap; Washington Ma; Latoche; Brian C. Schaefer

Assembly of a multiprotein complex containing the adaptor protein p62 promotes NF-κB activation in T cells. Forming the Right Clusters Engagement of T cells by antigen-presenting cells (APCs) through stimulation of the T cell receptor (TCR) leads to the formation of cytosolic multiprotein clusters (signalosomes), which enable activation of the transcription factor nuclear factor κB (NF-κB). Paul et al. showed the sequence of events in signalosome formation through confocal imaging of T cell–APC conjugates. Engagement of the TCR first led to formation of a protein complex containing the adaptor protein p62. Signalosomes containing p62 then recruited and activated the IKK complex, which is required for the activation of NF-κB. T cells from p62-deficient mice exhibited decreased signalosome formation and reduced NF-κB signaling, suggesting a central role for p62 in mediating TCR-dependent NF-κB activation. Antigen-mediated stimulation of the T cell receptor (TCR) triggers activation of nuclear factor κB (NF-κB), a key transcriptional regulator of T cell proliferation and effector cell differentiation. TCR signaling to NF-κB requires both the Carma1-Bcl10-Malt1 (CBM) complex and the inhibitor of κB (IκB) kinase (IKK) complex; however, the molecular mechanisms connecting the CBM complex to activation of IKK are incompletely defined. We found that the active IKK complex is a component of a TCR-dependent cytosolic Bcl10-Malt1 signalosome containing the adaptor protein p62, which forms in effector T cells. Phosphorylated IκBα and NF-κB were transiently recruited to this signalosome before NF-κB translocated to the nucleus. Inhibiting the activity of the kinase TAK1 or IKK blocked the phosphorylation of IKK, but not the formation of p62-Bcl10-Malt1 clusters, suggesting that activation of IKK occurs after signalosome assembly. Furthermore, analysis of T cells from p62-deficient mice demonstrated that the p62-dependent clustering of signaling components stimulated activation of NF-κB in effector T cells. Thus, TCR-stimulated activation of NF-κB requires the assembly of cytosolic p62-Bcl10-Malt1-IKK signalosomes, which may ensure highly regulated activation of NF-κB in response to TCR engagement.

Collaboration


Dive into the Brian C. Schaefer's collaboration.

Top Co-Authors

Avatar

Suman Paul

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Anuj K. Kashyap

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Clifton L. Dalgard

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

John W. Kappler

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Maria Traver

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Philippa Marrack

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Gauthaman Sukumar

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Lara M. Kingeter

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mouna Lagraoui

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Natalia G. Cartwright

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge