Network


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

Hotspot


Dive into the research topics where Patrick E. Fields is active.

Publication


Featured researches published by Patrick E. Fields.


Science | 1996

Blocked Ras Activation in Anergic CD4+ T Cells

Patrick E. Fields; Thomas F. Gajewski; Frank W. Fitch

T cell anergy is a state of functional unresponsiveness characterized by the inability to produce interleukin-2 (IL-2) upon T cell receptor stimulation. The mitogen-activated protein kinases ERK-1 and ERK-2 and the guanosine triphosphate-binding protein p21Ras were found to remain unactivated upon stimulation of anergic murine T helper cell 1 clones. The inability to activate the Ras pathway did not result from a defect in association among Shc, Grb-2, and murine Son of Sevenless, nor from a defect in their tyrosine phosphorylation. This block in Ras activation may lead to defective transactivation at activator protein 1 sites in anergic cells and may enable T cells to shut down IL-2 production selectively during anergy.


Journal of Immunology | 2000

Cutting Edge: TGF-β Inhibits Th Type 2 Development Through Inhibition of GATA-3 Expression

Leonid Gorelik; Patrick E. Fields; Richard A. Flavell

TGF-β is an important immunomodulatory cytokine that can inhibit differentiation of effector T cells. In this report, we address the molecular mechanisms through which TGF-β inhibits differentiation of CD4+ cells into Th type 2 cells. We demonstrate that TGF-β inhibits GATA-3 expression in developing Th cells. We also show that inhibition of GATA-3 expression by TGF-β is a major mechanism of inhibition of Th2 differentiation by TGF-β as ectopic expression of GATA-3 in developing T cells overcomes the ability of TGF-β to inhibit Th2 differentiation. TGF-β likely inhibits GATA-3 expression at the transcriptional level and does so without interfering with IL-4 signaling.


Journal of Immunology | 2002

Cutting Edge: Changes in Histone Acetylation at the IL-4 and IFN-γ Loci Accompany Th1/Th2 Differentiation

Patrick E. Fields; Sean T. Kim; Richard A. Flavell

Peripheral T cell differentiation is accompanied by chromatin changes at the signature cytokine loci. Using chromatin immunoprecipitation we demonstrate that profound increases in histone acetylation occur at the IFN-γ and IL-4 loci during Th1/Th2 differentiation. These changes in histone acetylation status are locus and lineage specific, and are maintained by the transcription factors Tbet and GATA3 in a STAT-dependent manner. Our results suggest a model of cytokine locus activation in which TCR signals initiate chromatin remodeling and locus opening in a cytokine-independent fashion. Subsequently, cytokine signaling reinforces polarization by expanding and maintaining the accessible state at the relevant cytokine locus (IL-4 or IFN-γ). In this model, GATA3 and Tbet serve as transcriptional maintenance factors, which keep the locus accessible to the transcriptional machinery.


Immunity | 2003

Regulation of the Th2 Cytokine Locus by a Locus Control Region

Gap Ryol Lee; Patrick E. Fields; Thomas J. Griffin; Richard A. Flavell

The Th2 cytokine genes IL4, IL5, and IL13 are clustered and expressed in a cell lineage-specific manner. We investigated the global locus-specific regulation of these genes using BAC transgenic mice containing the murine Th2 cytokine cluster carrying an IL4 promoter-luciferase reporter. IL4 promoter activity in effector CD4 T cells from these transgenic mice was strong, Th2 specific, and copy number dependent, suggesting the presence of an LCR in the locus. The production of IL4 and IL13, but not IL5, by these cells was also copy number dependent. Deletion analysis defined a 25 kb fragment in the RAD50 gene as the region containing the LCR activity. Expression of the IL4 promoter-luciferase reporter was transactivated by GATA-3 irrespective of position in the locus, suggesting the global nature of this regulation. The LCR itself, however, does not respond directly to GATA-3.


Blood | 2010

Early mammalian erythropoiesis requires the Dot1L methyltransferase

Yi Feng; Yanping Yang; Manoela M. Ortega; Jessica N. Copeland; Mingcai Zhang; Jennifer B. Jacob; Timothy A. Fields; Jay L. Vivian; Patrick E. Fields

Histone methylation is an important regulator of gene expression; its coordinated activity is critical in complex developmental processes such as hematopoiesis. Disruptor of telomere silencing 1-like (DOT1L) is a unique histone methyltransferase that specifically methylates histone H3 at lysine 79. We analyzed Dot1L-mutant mice to determine influence of this enzyme on embryonic hematopoiesis. Mutant mice developed more slowly than wild-type embryos and died between embryonic days 10.5 and 13.5, displaying a striking anemia, especially apparent in small vessels of the yolk sac. Further, a severe, selective defect in erythroid, but not myeloid, differentiation was observed. Erythroid progenitors failed to develop normally, showing retarded progression through the cell cycle, accumulation during G₀/G₁ stage, and marked increase in apoptosis in response to erythroid growth factors. GATA2, a factor essential for early erythropoiesis, was significantly reduced in Dot1L-deficient cells, whereas expression of PU.1, a transcription factor that inhibits erythropoiesis and promotes myelopoiesis, was increased. These data suggest a model whereby DOT1L-dependent lysine 79 of histone H3 methylation serves as a critical regulator of a differentiation switch during early hematopoiesis, regulating steady-state levels of GATA2 and PU.1 transcription, thus controlling numbers of circulating erythroid and myeloid cells.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Demethylation of a specific hypersensitive site in the Th2 locus control region

Sean T. Kim; Patrick E. Fields; Richard A. Flavell

A growing body of literature has examined and implicated DNA methylation as a critical epigenetic modification in T helper (Th) cell differentiation. The absence of DNA methyltransferases or methyl-binding proteins derepresses many cytokine loci, allowing their ectopic expression, while methylation of specific CpG residues is sufficient to prevent expression. Here, we characterize demethylation events of the Th2 cytokine locus control region (LCR). rad50 hypersensitive site 7 (RHS7), a hypersensitive site within this LCR, becomes demethylated in a STAT6-dependent manner and only in cells stimulated under type 2 conditions. Robust demethylation appears to require signaling contributions from both IL-4 receptor, via STAT6, and CD28, but it cannot be effected by GATA3. Finally, RHS7 is demethylated independently of cell division, consistent with an “active,” rather than passive, mechanism. Taken together, these findings firmly connect RHS7 demethylation and Th2 LCR activation in the type 2 differentiation program.


Journal of Immunology | 2012

Th2 skewing by activation of Nrf2 in CD4+ T cells

Cheryl E. Rockwell; Mingcai Zhang; Patrick E. Fields; Curtis D. Klaassen

NF erythroid 2-related factor 2 (Nrf2) is a transcription factor that mediates the upregulation of a battery of cytoprotective genes in response to cell stress. Recent studies showed that Nrf2 also modulates immune responses and exhibits anti-inflammatory activity. In this article, we demonstrate that a common food preservative, tert-butylhydroquinone, can activate Nrf2 in T cells, as evidenced by Nrf2 binding to the antioxidant response element and the subsequent upregulation of Nrf2 target genes. The activation of Nrf2 suppresses IFN-γ production, while inducing the production of the Th2 cytokines IL-4, IL-5, and IL-13. Nrf2 activation also suppresses T-bet DNA binding and promotes GATA-binding protein 3 DNA binding. Collectively, the present studies suggested that Nrf2 activation skews CD4+ T cells toward Th2 differentiation and, thus, represents a novel regulatory mechanism in CD4+ T cells. Further studies are needed to determine whether the commercial use of Nrf2 activators as food preservatives promotes food allergies in humans.


Journal of Immunology | 2001

Absence of CTLA-4 Lowers the Activation Threshold of Primed CD8+ TCR-Transgenic T Cells: Lack of Correlation with Src Homology Domain 2-Containing Protein Tyrosine Phosphatase

Thomas F. Gajewski; Francesca Fallarino; Patrick E. Fields; Fabiola V. Rivas; Maria-Luisa Alegre

To examine the role of CTLA-4 in controlling Ag-specific CD8+ T cell activation, TCR-transgenic/CTLA-4 wild-type or -deficient mice were generated in a recombination-activating gene 2-deficient background. Naive T cells from these mice responded comparably whether or not CTLA-4 was expressed. In contrast, primed T cells responded more vigorously if they lacked CTLA-4 expression. We took advantage of the difference between naive and primed T cell responses to approach the mechanism of CTLA-4 function. Single-cell analyses demonstrated that a greater fraction of CTLA-4-deficient cells responded to a fixed dose of Ag compared with CTLA-4-expressing cells, whereas the magnitude of response per cell was comparable. A shift in the dose-response curve to APCs was also observed such that fewer APCs were required to activate CTLA-4-deficient T cells to produce intracellular IFN-γ and to proliferate. These results suggest that CTLA-4 controls the threshold of productive TCR signaling. Biochemical analysis comparing stimulated naive and primed TCR-transgenic cells revealed no obvious differences in expression of total CTLA-4, tyrosine-phosphorylated CTLA-4, and associated Src homology domain 2-containing protein tyrosine phosphatase. Thus, the biochemical mechanism explaining the differential inhibitory effect of CTLA-4 on naive and primed CD8+ T cells remains unclear.


Journal of Molecular Medicine | 1996

Control of T lymphocyte signal transduction through clonal anergy

Patrick E. Fields; Frank W. Fitch; Thomas F. Gajewski

Abstract Stimulation of interleukin-2 producing T lymphocytes via the T cell receptor (TCR) complex in the absence of other costimulatory factors results paradoxically not in activation but in an unresponsive state termed clonal anergy. T cell anergy appears to be a mechanism by which potentially autoreactive T lymphocytes are inactivated in the periphery, thus maintaining tolerance to self antigens. The breakdown of such tolerance may result in autoimmune diseases. In contrast, induction of peripheral tolerance is the ultimate goal in organ transplantation and is a potential mechanism by which a growing tumor evades immune destruction. The anergic state is characterized by an inability to secrete interleukin-2 and proliferate following restimulation via the TCR even in the presence of costimulatory factors. Recent studies have demonstrated a specific block in Ras activation in anergic T lymphocytes. This defect is correlated with a failure to activate the downstream effectors Erk and Jnk and a lack of activation of the AP-1 transcription factor complex, offering a plausible mechanism for the inability to initiate interleukin-2 gene transcription in the anergic state.


Cellular Signalling | 2009

RHO GTPASE ACTIVITY MODULATES WNT3A/β-CATENIN SIGNALING

Jessica Rossol-Allison; Laura N. Stemmle; Katherine I. Swenson-Fields; Patrick Kelly; Patrick E. Fields; Shannon McCall; Patrick J. Casey; Timothy A. Fields

Wnt proteins constitute a family of secreted signaling molecules that regulate highly conserved pathways essential for development and, when aberrantly activated, drive oncogenesis in a number of human cancers. A key feature of the most widely studied Wnt signaling cascade is the stabilization of cytosolic beta-catenin, resulting in beta-catenin nuclear translocation and transcriptional activation of multiple target genes. In addition to this canonical, beta-catenin-dependent pathway, Wnt3A has also been shown to stimulate RhoA GTPase. While the importance of activated Rho to non-canonical Wnt signaling is well appreciated, the potential contribution of Wnt3A-stimulated RhoA to canonical beta-catenin-dependent transcription has not been examined and is the focus of this study. We find that activated Rho is required for Wnt3A-stimulated osteoblastic differentiation in C3H10T1/2 mesenchymal stem cells, a biological phenomenon mediated by stabilized beta-catenin. Using expression microarrays and real-time RT-PCR analysis, we show that Wnt3A-stimulated transcription of a subset of target genes is Rho-dependent, indicating that full induction of these Wnt targets requires both beta-catenin and Rho activation. Significantly, neither beta-catenin stabilization nor nuclear translocation stimulated by Wnt3A is affected by inhibition or activation of RhoA. These findings identify Rho activation as a critical element of the canonical Wnt3A-stimulated, beta-catenin-dependent transcriptional program.

Collaboration


Dive into the Patrick E. Fields's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge