Network


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

Hotspot


Dive into the research topics where Paula Preston-Hurlburt is active.

Publication


Featured researches published by Paula Preston-Hurlburt.


Nature | 1997

A human homologue of the Drosophila Toll protein signals activation of adaptive immunity

Ruslan Medzhitov; Paula Preston-Hurlburt; Charles A. Janeway

Induction of the adaptive immune response depends on the expression of co-stimulatory molecules and cytokines by antigen-presenting cells. The mechanisms that control the initial induction of these signals upon infection are poorly understood. It has been proposed that their expression is controlled by the non-clonal, or innate, component of immunity that preceded in evolution the development of an adaptive immune system in vertebrates. We report here the cloning and characterization of a human homologue of the Drosophila toll protein (Toll) which has been shown to induce the innate immune response in adult Drosophila. Like Drosophila Toll, human Toll is a type I transmembrane protein with an extracellular domain consisting of a leucine-rich repeat (LRR) domain, and a cytoplasmic domain homologous to the cytoplasmic domain of the human interleukin (IL)-1 receptor. Both Drosophila Toll and the IL-1 receptor are known to signal through the NF-κB pathway. We show that a constitutively active mutant of human Toll transfected into human cell lines can induce the activation of NF-κB and the expression of NF-κB-controlled genes for the inflammatory cytokines IL-1, IL-6 and IL-8, as well as the expression of the co-stimulatory molecule B7.1, which is required for the activation of naive T cells.


Molecular Cell | 1998

MyD88 Is an Adaptor Protein in the hToll/IL-1 Receptor Family Signaling Pathways

Ruslan Medzhitov; Paula Preston-Hurlburt; Elizabeth Kopp; Andrew Stadlen; Chaoqun Chen; Sankar Ghosh; Charles A. Janeway

The Toll-mediated signaling cascade using the NF-kappaB pathway has been shown to be essential for immune responses in adult Drosophila, and we recently reported that a human homolog of the Drosophila Toll protein induces various immune response genes via this pathway. We now demonstrate that signaling by the human Toll receptor employs an adaptor protein, MyD88, and induces activation of NF-kappaB via the Pelle-like kinase IRAK and the TRAF6 protein, similar to IL-1R-mediated NF-kappaB activation. However, we find that Toll and IL-1R signaling pathways are not identical with respect to AP-1 activation. Finally, our findings implicate MyD88 as a general adaptor/regulator molecule for the Toll/IL-1R family of receptors for innate immunity.


Immunity | 1996

The Specificity and Orientation of a TCR to its Peptide–MHC Class II Ligands

Derek B. Sant'Angelo; Greg Waterbury; Paula Preston-Hurlburt; Sangwook Tim Yoon; Ruslan Medzhitov; Soon-cheol Hong; Charles A. Janeway

A T cell-mediated immune response is mainly determined by the 3-5 aa residues that protrude upwards from a peptide bound to an MHC molecule. Alterations of these peptide residues can diminish, eliminate or radically alter the signal that the T cell receives through its T cell receptor (TCR). We have used peptide immunizations of normal mice and mice carrying alpha or beta chain TCR transgenes to identify three distinct peptide contact points. One, near the carboxyl terminus of the peptide, involves the beta chain CDR3 region; the second was centrally located and interacted with both the alpha and beta chain CDR3 loops; the third was near the amino terminus of the peptide, and affected V alpha gene usage, but not the structure of CDR3 of either TCR chain. Based on these results, we propose an orientation for the TCR of this cloned line and argue for its generality.


Nature Immunology | 2002

Dual receptor T cells extend the immune repertoire for foreign antigens

Xin He; Charles A. Janeway; Matthew Levine; Eve Robinson; Paula Preston-Hurlburt; Christophe Viret; Kim Bottomly

Since the discovery of T cells that express two T cell receptors (TCRs), termed dual TCR cells, most studies have focused on their autoimmune potential, while their beneficial roles remained elusive. We identified, in normal mice, dual TCR cells that participated in the immune response to a foreign antigen. Unlike single TCR cells, dual TCR cells used the nonselected TCR to respond in the periphery, but relied on coexpression of a second TCR for intrathymic selection. We found that they were selected at low frequency in the naïve repertoire, but dominated the response to antigen through clonal expansion. Thus, dual TCR cells can extend the TCR repertoire for foreign antigens by rescuing functional TCRs that cannot be selected on single TCR cells; they can, therefore, benefit the immune system.


Diabetes | 2012

Synergistic Reversal of Type 1 Diabetes in NOD Mice With Anti-CD3 and Interleukin-1 Blockade: Evidence of Improved Immune Regulation

Vitaly Ablamunits; Octavian Henegariu; Jakob Bondo Hansen; Lynn Opare-Addo; Paula Preston-Hurlburt; Pere Santamaria; Thomas Mandrup-Poulsen; Kevan C. Herold

Inflammatory cytokines are involved in autoimmune diabetes: among the most prominent is interleukin (IL)-1β. We postulated that blockade of IL-1β would modulate the effects of anti-CD3 monoclonal antibody (mAb) in treating diabetes in NOD mice. To test this, we treated hyperglycemic NOD mice with F(ab′)2 fragments of anti-CD3 mAb with or without IL-1 receptor antagonist (IL-1RA), or anti–IL-1β mAb. We studied the reversal of diabetes and effects of treatment on the immune system. Mice that received a combination of anti-CD3 mAb with IL-1RA showed a more rapid rate of remission of diabetes than mice treated with anti-CD3 mAb or IL-1RA alone. Combination-treated mice had increased IL-5, IL-4, and interferon (IFN)-γ levels in circulation. There were reduced pathogenic NOD-relevant V7 peptide-V7+ T cells in the pancreatic lymph nodes. Their splenocytes secreted more IL-10, had increased arginase expression in macrophages and dendritic cells, and had delayed adoptive transfer of diabetes. After 1 month, there were increased concentrations of IgG1 isotype antibodies and reduced intrapancreatic expression of IFN-γ, IL-6, and IL-17 despite normal splenocyte cytokine secretion. These studies indicate that the combination of anti-CD3 mAb with IL-1RA is synergistic in reversal of diabetes through a combination of mechanisms. The combination causes persistent remission from islet inflammation.


Journal of Clinical Investigation | 2013

Specific peripheral B cell tolerance defects in patients with multiple sclerosis

Tuure Kinnunen; Nicolas Chamberlain; Henner Morbach; Tineke Cantaert; Megan Lynch; Paula Preston-Hurlburt; Kevan C. Herold; David A. Hafler; Kevin C. O’Connor; Eric Meffre

Multiple sclerosis (MS) is a genetically mediated autoimmune disease of the central nervous system. B cells have recently emerged as major contributors to disease pathogenesis, but the mechanisms responsible for the loss of B cell tolerance in patients with MS are largely unknown. In healthy individuals, developing autoreactive B cells are removed from the repertoire at 2 tolerance checkpoints during early B cell development. Both of these central and peripheral B cell tolerance checkpoints are defective in patients with rheumatoid arthritis (RA) and type 1 diabetes (T1D). Here, we found that only the peripheral, but not the central, B cell tolerance checkpoint is defective in patients with MS. We show that this specific defect is accompanied by increased activation and homeostatic proliferation of mature naive B cells. Interestingly, all of these MS features parallel defects observed in FOXP3-deficient IPEX patients, who harbor nonfunctional Tregs. We demonstrate that in contrast to patients with RA or T1D, bone marrow central B cell selection in MS appears normal in most patients. In contrast, patients with MS suffer from a specific peripheral B cell tolerance defect that is potentially attributable to impaired Treg function and that leads to the accumulation of autoreactive B cell clones in their blood.


Science Translational Medicine | 2012

Teplizumab Induces Human Gut-Tropic Regulatory Cells in Humanized Mice and Patients

Frank Waldron-Lynch; Octavian Henegariu; Songyan Deng; Paula Preston-Hurlburt; James E. Tooley; Richard A. Flavell; Kevan C. Herold

Humanized mice can be successfully used as a preclinical testing platform to identify the mechanism of action of immunotherapies in humans. Gut Reaction The immune system is tasked with protecting the body from foreign invaders; however, sometimes the body itself is perceived as foreign. One such case is type 1 diabetes, where the immune system attacks and destroys the insulin-producing β cells of the pancreas. An immunomodulatory drug, teplizumab, is currently being tested in clinical trials for new-onset type 1 diabetes on the basis of encouraging preclinical and phase 1 studies. However, data from the early trials did not confirm the proposed mechanism of action from the preclinical studies. Using a humanized mouse model, Waldron-Lynch et al. now identify a mechanism that is consistent with type 1 diabetes patients treated with teplizumab. Teplizumab is a humanized non–Fc-binding antibody to CD3—a molecule critical for T cell activation. Waldron-Lynch et al. found that in humanized mice, teplizumab causes certain T cells that express the gut-homing receptor CCR6 to leave the circulation and migrate to the gut. While there, these cells begin producing a regulatory cytokine, IL-10; these cells then return to circulation. These IL-10–producing CCR6+ cells were also found in the blood of type 1 diabetes patients after teplizumab treatment. By allowing examination of human immune cells without the restrictions of looking in human subjects, observations of humanized mice should improve the design—and hopefully increase the chances of success—of future clinical trials. The development and optimization of immune therapies in patients has been hampered by the lack of preclinical models in which their effects on human immune cells can be studied. As a result, observations that have been made in preclinical studies have suggested mechanisms of drug action in murine models that have not been confirmed in clinical studies. Here, we used a humanized mouse reconstituted with human hematopoietic stem cells to study the mechanism of action of teplizumab, an Fc receptor nonbinding humanized monoclonal antibody to CD3 being tested in clinical trials for the treatment of patients with type 1 diabetes mellitus. In this model, human gut-tropic CCR6+ T cells exited the circulation and secondary lymph organs and migrated to the small intestine. These cells then produced interleukin-10 (IL-10), a regulatory cytokine, in quantities that could be detected in the peripheral circulation. Blocking T cell migration to the small intestine with natalizumab, which prevents cellular adhesion by inhibiting α4 integrin binding, abolished the treatment effects of teplizumab. Moreover, IL-10 expression by CD4+CD25highCCR6+FoxP3 cells returning to the peripheral circulation was increased in patients with type 1 diabetes treated with teplizumab. These findings demonstrate that humanized mice may be used to identify novel immunologic mechanisms that occur in patients treated with immunomodulators.


PLOS ONE | 2012

RAGE Expression in Human T Cells: A Link between Environmental Factors and Adaptive Immune Responses

Eitan M. Akirav; Paula Preston-Hurlburt; Justin Garyu; Octavian Henegariu; Raphael Clynes; Ann Marie Schmidt; Kevan C. Herold

The Receptor for Advanced Glycation Endproducts (RAGE) is a scavenger ligand that binds glycated endproducts as well as molecules released during cell death such as S100b and HMGB1. RAGE is expressed on antigen presenting cells where it may participate in activation of innate immune responses but its role in adaptive human immune responses has not been described. We have found that RAGE is expressed intracellularly in human T cells following TCR activation but constitutively on T cells from patients with diabetes. The levels of RAGE on T cells from patients with diabetes are not related to the level of glucose control. It co-localizes to the endosomes. Its expression increases in activated T cells from healthy control subjects but bystander cells also express RAGE after stimulation of the antigen specific T cells. RAGE ligands enhance RAGE expression. In patients with T1D, the level of RAGE expression decreases with T cell activation. RAGE+ T cells express higher levels of IL-17A, CD107a, and IL-5 than RAGE- cells from the same individual with T1D. Our studies have identified the expression of RAGE on adaptive immune cells and a role for this receptor and its ligands in modulating human immune responses.


Journal of Immunology | 2014

Humanized Mice as a Model for Aberrant Responses in Human T Cell Immunotherapy

Nalini Vudattu; Frank Waldron-Lynch; Lucy A. Truman; Songyan Deng; Paula Preston-Hurlburt; Richard Torres; Maurice T. Raycroft; Mark J. Mamula; Kevan C. Herold

Immune-deficient mice, reconstituted with human stem cells, have been used to analyze human immune responses in vivo. Although they have been used to study immune responses to xenografts, allografts, and pathogens, there have not been models of autoimmune disease in which the mechanisms of the pathologic process can be analyzed. We have found that reconstituted “humanized” mice treated with anti–CTLA-4 Ab (ipilimumab) develop autoimmune disease characterized by hepatitis, adrenalitis, sialitis, anti-nuclear Abs, and weight loss. Induction of autoimmunity involved activation of T cells and cytokine production, and increased infiltration of APCs. When anti–CTLA-4 mAb–treated mice were cotreated with anti-CD3 mAb (teplizumab), hepatitis and anti-nuclear Abs were no longer seen and weight loss did not occur. The anti-CD3 blocked proliferation and activation of T cells, release of IFN-γ and TNF, macrophage infiltration, and release of IP-10 that was induced with anti–CTLA-4 mAb. We also found increased levels of T regulatory cells (CD25+CD127−) in the spleen and mesenteric lymph nodes in the mice treated with both Abs and greater constitutive phosphorylation of STAT5 in T regulatory cells in spleen cells compared with mice treated with anti–CTLA-4 mAb alone. We describe a model of human autoimmune disease in vivo. Humanized mice may be useful for understanding the mechanisms of biologics that are used in patients. Hepatitis, lymphadenopathy, and other inflammatory sequelae are adverse effects of ipilimumab treatment in humans, and this study may provide insights into this pathogenesis and the effects of immunologics on autoimmunity.


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

Induction of TLR4-target genes entails calcium/calmodulin-dependent regulation of chromatin remodeling

Dazhi Lai; Mimi Wan; Jie Wu; Paula Preston-Hurlburt; Ritu Kushwaha; Thomas Grundström; Anthony N. Imbalzano; Tian Chi

Upon toll-like receptor 4 (TLR4) signaling in macrophages, the mammalian Swi/Snf-like BAF chromatin remodeling complex is recruited to many TLR4 target genes where it remodels their chromatin to promote transcription. Here, we show that, surprisingly, recruitment is not sufficient for chromatin remodeling; a second event, dependent on calcium/calmodulin (CaM), is additionally required. Calcium/CaM directly binds the HMG domain of the BAF57 subunit within the BAF complex. Calcium/CaM antagonists, including a CaM-binding peptide derived from BAF57, abolish BAF-dependent remodeling and gene expression without compromising BAF recruitment. BAF57 RNAi and BAF57 dominant negative mutants defective in CaM binding similarly impair the induction of BAF target genes. Our data implicate calcium/CaM in TLR4 signaling, and reveal a previously undescribed, recruitment-independent mode of regulation of the BAF complex that is probably achieved through a direct CaM-BAF interaction.

Collaboration


Dive into the Paula Preston-Hurlburt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander Y. Rudensky

Memorial Sloan Kettering Cancer Center

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
Researchain Logo
Decentralizing Knowledge