Network


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

Hotspot


Dive into the research topics where J. David Farrar is active.

Publication


Featured researches published by J. David Farrar.


Nature Immunology | 2000

Selective loss of type I interferon-induced STAT4 activation caused by a minisatellite insertion in mouse Stat2

J. David Farrar; Janice D. Smith; Theresa L. Murphy; Stewart Leung; George R. Stark; Kenneth M. Murphy

The use of murine systems to model pathogen-induced human diseases presumes that general immune mechanisms between these species are conserved. One important immunoregulatory mechanism involves linkage of innate and adaptive immunity to direct the development of T helper subsets, for example toward subset 1 (TH1) development through STAT4 activation. In analyzing type I interferon signaling, we uncovered a difference between murine and human cells which may affect how these two species control linkage between innate and adaptive immunity. We show that in humans, type I interferons induce TH1 development and can activate STAT4 by recruitment to the IFN-α receptor complex specifically via the carboxy-terminus of STAT2. However, the mouse Stat2 gene harbors a minisatellite insertion that has altered the carboxy-terminus and selectively disrupted its capacity to activate STAT4, but not other STATs. This defect in murine Stat2 suggests that the signals leading to STAT4 activation and TH1 development in CD4+ T cells are different between mice and humans.


Journal of Clinical Investigation | 2002

T helper subset development: roles of instruction, selection, and transcription

J. David Farrar; Hélène Asnagli; Kenneth M. Murphy

The past 15 years have seen the development of a field in immunology entirely devoted to understanding the divergence of CD4+ T cells into distinct cytokine-producing subsets. Generally speaking, the immune response to a pathogen can develop toward a more strongly cellular type (type 1), or toward a more allergic type of response (type 2). When well developed, these gear the immune response for the effective elimination of different types of pathogens, with a type 1 response being more effective against intracellular pathogens, for example Listeria monocytogenes or mycobacteria. Type 2 allergic responses, in contrast, favor the elimination of parasites such as helminths. The basis for this selective action resides in the cytokines made by the CD4+ T helper cell subsets in these responses. T helper type 1 (Th1) cells produce several characteristic cytokines, most notably IL-2 and IFN-γ, whereas Th2 cells produce a set of cytokines, most notably IL-4, IL-5, and IL-13. In turn, IL-2 and IFN-γ promote the development of strong cell-mediated immunity, whereas the type 2 cytokines promote allergic responses effective in eliminating parasites. When confronted with a pathogen, it is important that the immune system activate the appropriate type of response. Fortunately, it has developed reliable mechanisms that help naive CD4 T cells in this choice. These mechanisms have been fairly well worked out at many levels over the last decade, and several thorough reviews have described recent findings regarding the signaling pathways and transcription factors that contribute to peripheral CD4+ Th development (1–3). Despite the great progress in the molecular understanding of these processes, there are still issues in this area that are controversial and actively debated. Here, we will focus on some aspects that are unresolved within the various models of Th1 and Th2 development and will try to fit together some of the recent observations that have motivated these somewhat theoretical considerations. In particular, we will take this opportunity to focus on a difficult topic, that of whether Th1/Th2 development rests on selective or instructive mechanisms, a general issue that is also debated in regard to CD4/CD8 lineage commitment and is common to many developmental systems. Distinguishing between strictly selective and instructive models is difficult, as many experimental results can be compatible with both interpretations.


Immunology | 2011

Regulation of effector and memory T-cell functions by type I interferon.

Jonathan P. Huber; J. David Farrar

Type I interferon (IFN‐α/β) is comprised of a family of highly related molecules that exert potent antiviral activity by interfering with virus replication and spread. IFN‐α/β secretion is tightly regulated through pathogen sensing pathways that are operative in most somatic cells. However, specialized antigen‐presenting plasmacytoid dendritic cells are uniquely equipped with the capacity to secrete extremely high levels of IFN‐α/β, suggesting a key role for this cytokine in priming adaptive T‐cell responses. Recent studies in both mice and humans have demonstrated a role for IFN‐α/β in directly influencing the fate of both CD4+ and CD8+ T cells during the initial phases of antigen recognition. As such, IFN‐α/β, among other innate cytokines, is considered an important ‘third signal’ that shapes the effector and memory T‐cell pool. Moreover, IFN‐α/β also serves as a counter‐regulator of T helper type 2 and type 17 responses, which may be important in the treatment of atopy and autoimmunity, and in the development of novel vaccine adjuvants.


Immunology Today | 2000

Type I interferons and T helper development

J. David Farrar; Kenneth M. Murphy

Abstract Type I interferons (IFN-1s) activate Stat4 and promote Th1 development in human, but not mouse, CD4 + T cells. Recently, IFN-1-induced Stat4 activation in human cells was found to require interactions of Stat4 with Stat2. A remarkable genetic dissimilarity between murine and human Stat2 explains the ability of human, but not mouse, CD4 + T cells to undergo IFN-1-induced Stat4 activation and Th1 development.


Journal of Biological Chemistry | 2000

Recruitment of Stat4 to the Human Interferon-α/β Receptor Requires Activated Stat2

J. David Farrar; Janice D. Smith; Theresa L. Murphy; Kenneth M. Murphy

Stat4 activation is involved in differentiation of type 1 helper (Th1) T cells. Although Stat4 is activated by interleukin (IL)-12 in both human and murine T cells, Stat4 is activated by interferon (IFN)-α only in human, but not murine, CD4+ T cells. This species-specific difference in cytokine activation of Stat4 underlies critical differences in Th1 development in response to cytokines and is important to the interpretation of murine models of immunopathogenesis. Here, we sought to determine the mechanism of Stat4 recruitment and activation by the human IFN-α receptor. Analysis of phosphopeptide binding analysis suggests that Stat4 does not interact directly with tyrosine-phosphorylated amino acid residues within the cytoplasmic domains of either of the subunits of the IFN-α receptor complex. Expression of murine Stat4 in the Stat1-deficient U3A and the Stat2-deficient U6A cell lines shows that IFN-α-induced Stat4 phosphorylation requires the presence of activated Stat2 but not Stat1. Thus, in contrast to the direct recruitment of Stat4 by the IL-12 receptor, Stat4 activation by the human IFN-α receptor occurs through indirect recruitment by intermediates involving Stat2.


Journal of Immunology | 2010

Cutting Edge: Type I IFN Reverses Human Th2 Commitment and Stability by Suppressing GATA3

Jonathan P. Huber; Hilario J. Ramos; Michelle A. Gill; J. David Farrar

T helper 2 cells regulate inflammatory responses to helminth infections while also mediating pathological processes of asthma and allergy. IL-4 promotes Th2 development by inducing the expression of the GATA3 transcription factor, and the Th2 phenotype is stabilized by a GATA3-dependent autoregulatory loop. In this study, we found that type I IFN (IFN-α/β) blocked human Th2 development and inhibited cytokine secretion from committed Th2 cells. This negative regulatory pathway was operative in human but not mouse CD4+ T cells and was selective to type I IFN, as neither IFN-γ nor IL-12 mediated such inhibition. IFN-α/β blocked Th2 cytokine secretion through the inhibition of GATA3 during Th2 development and in fully committed Th2 cells. Ectopic expression of GATA3 via retrovirus did not overcome IFN-α/β–mediated inhibition of Th2 commitment. Thus, we demonstrate a novel role for IFN-α/β in blocking Th2 cells, suggesting its potential as a promising therapy for atopy and asthma.


PLOS ONE | 2013

SLE Peripheral Blood B Cell, T Cell and Myeloid Cell Transcriptomes Display Unique Profiles and Each Subset Contributes to the Interferon Signature

Amy M. Becker; Kathryn H. Dao; Bobby Kwanghoon Han; Roger Kornu; Shuchi Lakhanpal; Angela B. Mobley; Quan Zhen Li; Yun Lian; Andreas Reimold; Nancy J. Olsen; David R. Karp; Fatema Z. Chowdhury; J. David Farrar; Anne B. Satterthwaite; Chandra Mohan; Peter E. Lipsky; Edward K. Wakeland; Laurie S. Davis

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that is characterized by defective immune tolerance combined with immune cell hyperactivity resulting in the production of pathogenic autoantibodies. Previous gene expression studies employing whole blood or peripheral blood mononuclear cells (PBMC) have demonstrated that a majority of patients with active disease have increased expression of type I interferon (IFN) inducible transcripts known as the IFN signature. The goal of the current study was to assess the gene expression profiles of isolated leukocyte subsets obtained from SLE patients. Subsets including CD19+ B lymphocytes, CD3+CD4+ T lymphocytes and CD33+ myeloid cells were simultaneously sorted from PBMC. The SLE transcriptomes were assessed for differentially expressed genes as compared to healthy controls. SLE CD33+ myeloid cells exhibited the greatest number of differentially expressed genes at 208 transcripts, SLE B cells expressed 174 transcripts and SLE CD3+CD4+ T cells expressed 92 transcripts. Only 4.4% (21) of the 474 total transcripts, many associated with the IFN signature, were shared by all three subsets. Transcriptional profiles translated into increased protein expression for CD38, CD63, CD107a and CD169. Moreover, these studies demonstrated that both SLE lymphoid and myeloid subsets expressed elevated transcripts for cytosolic RNA and DNA sensors and downstream effectors mediating IFN and cytokine production. Prolonged upregulation of nucleic acid sensing pathways could modulate immune effector functions and initiate or contribute to the systemic inflammation observed in SLE.


Blood | 2009

Reciprocal responsiveness to interleukin-12 and interferon-α specifies human CD8+ effector versus central memory T-cell fates

Hilario J. Ramos; Ann M. Davis; Alexander G. Cole; John D. Schatzle; James Forman; J. David Farrar

Multiple innate signals regulate the genesis of effector and memory CD8+ T cells. In this study, we demonstrate that the innate cytokines interleukin (IL)-12 and interferon (IFN)-alpha/beta regulate distinct aspects of effector and memory human CD8+ T-cell differentiation. IL-12 exclusively promoted the development of IFN-gamma- and tumor necrosis factor (TNF)-alpha-secreting T effector memory (T(EM)) cells, whereas IFN-alpha drove the development of T central memory (T(CM)) cells. The development of T(EM) and T(CM) was linked to cell division. In rapidly dividing cells, IL-12 programmed T(EM) through induction of the IL-12 receptor beta2. In contrast, IFN-alpha regulated T(CM) development by slowing the progression of cell division in a subpopulation of cells that selectively expressed elevated IFN-alpha/beta receptor-2. The strength of signal delivered through T-cell receptor (TCR) engagement regulated the responsiveness of cells to IL-12 and IFN-alpha. In the presence of both IL-12 and IFN-alpha, these cytokine signals were amplified as the strength of the TCR signal was increased, promoting the simultaneous development of both T(CM) and T(EM). Together, our results support a novel model in which IL-12 and IFN-alpha act in a nonredundant manner to regulate the colinear generation of both effector and memory cells.


Blood | 2009

Reciprocal responsiveness to IL-12 and IFN-α specifies human CD8+ effector versus central memory T cell fates

Hilario J. Ramos; Ann M. Davis; Alexander G. Cole; John D. Schatzle; James Forman; J. David Farrar

Multiple innate signals regulate the genesis of effector and memory CD8+ T cells. In this study, we demonstrate that the innate cytokines interleukin (IL)-12 and interferon (IFN)-alpha/beta regulate distinct aspects of effector and memory human CD8+ T-cell differentiation. IL-12 exclusively promoted the development of IFN-gamma- and tumor necrosis factor (TNF)-alpha-secreting T effector memory (T(EM)) cells, whereas IFN-alpha drove the development of T central memory (T(CM)) cells. The development of T(EM) and T(CM) was linked to cell division. In rapidly dividing cells, IL-12 programmed T(EM) through induction of the IL-12 receptor beta2. In contrast, IFN-alpha regulated T(CM) development by slowing the progression of cell division in a subpopulation of cells that selectively expressed elevated IFN-alpha/beta receptor-2. The strength of signal delivered through T-cell receptor (TCR) engagement regulated the responsiveness of cells to IL-12 and IFN-alpha. In the presence of both IL-12 and IFN-alpha, these cytokine signals were amplified as the strength of the TCR signal was increased, promoting the simultaneous development of both T(CM) and T(EM). Together, our results support a novel model in which IL-12 and IFN-alpha act in a nonredundant manner to regulate the colinear generation of both effector and memory cells.


Journal of Immunology | 2007

IFN-α Is Not Sufficient to Drive Th1 Development Due to Lack of Stable T-bet Expression

Hilario J. Ramos; Ann M. Davis; Thaddeus C. George; J. David Farrar

During inflammatory immune responses, the innate cytokine IL-12 promotes CD4+ Th-1 development through the activation of the second messenger STAT4 and the subsequent expression of T-bet. In addition, type I IFN (IFN-αβ), secreted primarily during viral and intracellular bacterial infections, can promote STAT4 activation in human CD4+ T cells. However, the role of IFN-αβ in regulating Th1 development is controversial, and previous studies have suggested a species-specific pathway leading to Th1 development in human but not mouse CD4+ T cells. In this study, we found that although both IFN-α and IL-12 can promote STAT4 activation, IFN-α failed to promote Th1 commitment in human CD4+ T cells. The difference between these innate signaling pathways lies with the ability of IL-12 to promote sustained STAT4 tyrosine phosphorylation, which correlated with stable T-bet expression in committed Th1 cells. IFN-α did not promote Th1 development in human CD4+ T cells because of attenuated STAT4 phosphorylation, which was insufficient to induce stable expression of T-bet. Further, the defect in IFN-α-driven Th1 development was corrected by ectopic expression of T-bet within primary naive human CD4+ T cells. These results indicate that IL-12 remains unique in its ability to drive Th1 development in human CD4+ T cells and that IFN-α lacks this activity due to its inability to promote sustained T-bet expression.

Collaboration


Dive into the J. David Farrar's collaboration.

Top Co-Authors

Avatar

Kenneth M. Murphy

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ann M. Davis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan P. Huber

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michelle A. Gill

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Fatema Z. Chowdhury

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Theresa L. Murphy

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Leonardo D. Estrada

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

James Forman

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Loderick A. Matthews

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge