Network


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

Hotspot


Dive into the research topics where Creg J. Workman is active.

Publication


Featured researches published by Creg J. Workman.


Nature | 2007

The inhibitory cytokine IL-35 contributes to regulatory T-cell function

Lauren W. Collison; Creg J. Workman; Timothy T. Kuo; Kelli L. Boyd; Yao Wang; Kate M. Vignali; Richard Cross; David Sehy; Richard S. Blumberg; Dario A. A. Vignali

Regulatory T (Treg) cells are a critical sub-population of CD4+ T cells that are essential for maintaining self tolerance and preventing autoimmunity, for limiting chronic inflammatory diseases, such as asthma and inflammatory bowel disease, and for regulating homeostatic lymphocyte expansion. However, they also suppress natural immune responses to parasites and viruses as well as anti-tumour immunity induced by therapeutic vaccines. Although the manipulation of Treg function is an important goal of immunotherapy, the molecules that mediate their suppressive activity remain largely unknown. Here we demonstrate that Epstein-Barr-virus-induced gene 3 (Ebi3, which encodes IL-27β) and interleukin-12 alpha (Il12a, which encodes IL-12α/p35) are highly expressed by mouse Foxp3+ (forkhead box P3) Treg cells but not by resting or activated effector CD4+ T (Teff) cells, and that an Ebi3–IL-12α heterodimer is constitutively secreted by Treg but not Teff cells. Both Ebi3 and Il12a messenger RNA are markedly upregulated in Treg cells co-cultured with Teff cells, thereby boosting Ebi3 and IL-12α production in trans. Treg-cell restriction of this cytokine occurs because Ebi3 is a downstream target of Foxp3, a transcription factor that is required for Treg-cell development and function. Ebi3–/– and Il12a–/– Treg cells have significantly reduced regulatory activity in vitro and fail to control homeostatic proliferation and to cure inflammatory bowel disease in vivo. Because these phenotypic characteristics are distinct from those of other IL-12 family members, this novel Ebi3–IL-12α heterodimeric cytokine has been designated interleukin-35 (IL-35). Ectopic expression of IL-35 confers regulatory activity on naive T cells, whereas recombinant IL-35 suppresses T-cell proliferation. Taken together, these data identify IL-35 as a novel inhibitory cytokine that may be specifically produced by Treg cells and is required for maximal suppressive activity.


Nature Immunology | 2009

Coregulation of CD8 + T cell exhaustion by multiple inhibitory receptors during chronic viral infection

Shawn D. Blackburn; Haina Shin; W. Nicholas Haining; Tao Zou; Creg J. Workman; Antonio Polley; Michael R. Betts; Gordon J. Freeman; Dario A. A. Vignali; E. John Wherry

T cell exhaustion often occurs during chronic infection and prevents optimal viral control. The molecular pathways involved in T cell exhaustion remain poorly understood. Here we show that exhausted CD8+ T cells are subject to complex layers of negative regulation resulting from the coexpression of multiple inhibitory receptors. Exhausted CD8+ T cells expressed up to seven inhibitory receptors. Coexpression of multiple distinct inhibitory receptors was associated with greater T cell exhaustion and more severe infection. Regulation of T cell exhaustion by various inhibitory pathways was nonredundant, as blockade of the T cell inhibitory receptors PD-1 and LAG-3 simultaneously and synergistically improved T cell responses and diminished viral load in vivo. Thus, CD8+ T cell responses during chronic viral infections are regulated by complex patterns of coexpressed inhibitory receptors.T cell exhaustion often occurs during chronic infections and prevents optimal viral control. The molecular pathways involved in T cell exhaustion, however, remain poorly understood. We demonstrate that exhausted CD8+ T cells are subject to complex layers of negative regulation due to co-expression of multiple inhibitory receptors. Exhausted CD8+ T cells expressed up to 7 inhibitory receptors. Co-expression of multiple distinct inhibitory receptors correlated with greater T cell exhaustion and more severe infection. Regulation of T cell exhaustion by diverse inhibitory pathways was non-redundant since blockade of PD-1 and LAG-3 simultaneously in vivo synergistically improved T cell responses and reduced viral load. Thus, CD8+ T cell responses during chronic viral infections are regulated by complex patterns of co-expressed inhibitory receptors.


Cancer Research | 2012

Immune Inhibitory Molecules LAG-3 and PD-1 Synergistically Regulate T-cell Function to Promote Tumoral Immune Escape

Seng Ryong Woo; Meghan E. Turnis; Monica V. Goldberg; Jaishree Bankoti; Mark J. Selby; Christopher Nirschl; Matthew L. Bettini; David M. Gravano; Peter Vogel; Chih Long Liu; Stephanie Tangsombatvisit; Joseph F. Grosso; George J. Netto; Matthew P. Smeltzer; Alcides Chaux; Paul J. Utz; Creg J. Workman; Drew M. Pardoll; Alan J. Korman; Charles G. Drake; Dario A. A. Vignali

Inhibitory receptors on immune cells are pivotal regulators of immune escape in cancer. Among these inhibitory receptors, CTLA-4 (targeted clinically by ipilimumab) serves as a dominant off-switch while other receptors such as PD-1 and LAG-3 seem to serve more subtle rheostat functions. However, the extent of synergy and cooperative interactions between inhibitory pathways in cancer remain largely unexplored. Here, we reveal extensive coexpression of PD-1 and LAG-3 on tumor-infiltrating CD4(+) and CD8(+) T cells in three distinct transplantable tumors. Dual anti-LAG-3/anti-PD-1 antibody treatment cured most mice of established tumors that were largely resistant to single antibody treatment. Despite minimal immunopathologic sequelae in PD-1 and LAG-3 single knockout mice, dual knockout mice abrogated self-tolerance with resultant autoimmune infiltrates in multiple organs, leading to eventual lethality. However, Lag3(-/-)Pdcd1(-/-) mice showed markedly increased survival from and clearance of multiple transplantable tumors. Together, these results define a strong synergy between the PD-1 and LAG-3 inhibitory pathways in tolerance to both self and tumor antigens. In addition, they argue strongly that dual blockade of these molecules represents a promising combinatorial strategy for cancer.


Nature | 2013

Stability and function of regulatory T cells is maintained by a neuropilin-1–semaphorin-4a axis

Greg M. Delgoffe; Seng-Ryong Woo; Meghan E. Turnis; David M. Gravano; Cliff Guy; Abigail E. Overacre; Matthew L. Bettini; Peter Vogel; David Finkelstein; Jody Bonnevier; Creg J. Workman; Dario A. A. Vignali

Regulatory T cells (Treg cells) have a crucial role in the immune system by preventing autoimmunity, limiting immunopathology, and maintaining immune homeostasis. However, they also represent a major barrier to effective anti-tumour immunity and sterilizing immunity to chronic viral infections. The transcription factor Foxp3 has a major role in the development and programming of Treg cells. The relative stability of Treg cells at inflammatory disease sites has been a highly contentious subject. There is considerable interest in identifying pathways that control the stability of Treg cells as many immune-mediated diseases are characterized by either exacerbated or limited Treg-cell function. Here we show that the immune-cell-expressed ligand semaphorin-4a (Sema4a) and the Treg-cell-expressed receptor neuropilin-1 (Nrp1) interact both in vitro, to potentiate Treg-cell function and survival, and in vivo, at inflammatory sites. Using mice with a Treg-cell-restricted deletion of Nrp1, we show that Nrp1 is dispensable for suppression of autoimmunity and maintenance of immune homeostasis, but is required by Treg cells to limit anti-tumour immune responses and to cure established inflammatory colitis. Sema4a ligation of Nrp1 restrained Akt phosphorylation cellularly and at the immunologic synapse by phosphatase and tensin homologue (PTEN), which increased nuclear localization of the transcription factor Foxo3a. The Nrp1-induced transcriptome promoted Treg-cell stability by enhancing quiescence and survival factors while inhibiting programs that promote differentiation. Importantly, this Nrp1-dependent molecular program is evident in intra-tumoral Treg cells. Our data support a model in which Treg-cell stability can be subverted in certain inflammatory sites, but is maintained by a Sema4a–Nrp1 axis, highlighting this pathway as a potential therapeutic target that could limit Treg-cell-mediated tumour-induced tolerance without inducing autoimmunity.


Cellular and Molecular Life Sciences | 2009

The Development and Function of Regulatory T Cells

Creg J. Workman; Andrea L. Szymczak-Workman; Lauren W. Collison; Meenu R. Pillai; Dario A. A. Vignali

Regulatory T cells (Tregs) are a critical subset of T cells that mediate peripheral tolerance. There are two types of Tregs: natural Tregs, which develop in the thymus, and induced Tregs, which are derived from naive CD4+ T cells in the periphery. Tregs utilize a variety of mechanisms to suppress the immune response. While Tregs are critical for the peripheral maintenance of potential autoreactive T cells, they can also be detrimental by preventing effective anti-tumor responses and sterilizing immunity against pathogens. In this review, we will discuss the development of natural and induced Tregs as well as the role of Tregs in a variety of disease settings and the mechanisms they utilize for suppression.


Journal of Immunology | 2005

Negative Regulation of T Cell Homeostasis by Lymphocyte Activation Gene-3 (CD223)

Creg J. Workman; Dario A. A. Vignali

Lymphocyte homeostasis is a central biological process that is tightly regulated. However, its molecular and cellular control is poorly understood. We show that aged mice deficient in lymphocyte activation gene 3 (LAG-3), an MHC class II binding CD4 homologue, have twice as many T cells as wild-type controls. CD4+ and CD8+ LAG-3-deficient T cells showed enhanced homeostatic expansion in lymphopenic hosts, which was abrogated by ectopic expression of wild-type LAG-3, but not by a signaling-defective mutant. In addition, in vivo treatment with anti-LAG-3 mAb resulted in enhanced T cell expansion to a level comparable to that in LAG-3-deficient cells. This deregulation of T cell homeostasis also resulted in the expansion of multiple cell types, including B cells, macrophages, granulocytes, and dendritic cells. Lastly, regulatory T cells were dependent on LAG-3 for their optimal control of T cell homeostasis. Our data suggest that LAG-3 negatively regulates T cell homeostasis by regulatory T cell-dependent and independent mechanisms.


Journal of Immunology | 2011

Cutting Edge: Human Regulatory T Cells Require IL-35 To Mediate Suppression and Infectious Tolerance

Vandana Chaturvedi; Lauren W. Collison; Clifford S. Guy; Creg J. Workman; Dario A. A. Vignali

Human regulatory T cells (Treg) are essential for the maintenance of immune tolerance. However, the mechanisms they use to mediate suppression remain controversial. Although IL-35 has been shown to play an important role in Treg-mediated suppression in mice, recent studies have questioned its relevance in human Treg. In this study, we show that human Treg express and require IL-35 for maximal suppressive capacity. Substantial upregulation of EBI3 and IL12A, but not IL10 and TGFB, was observed in activated human Treg compared with conventional T cells (Tconv). Contact-independent Treg-mediated suppression was IL-35 dependent and did not require IL-10 or TGF-β. Lastly, human Treg-mediated suppression led to the conversion of the suppressed Tconv into iTr35 cells, an IL-35–induced Treg population, in an IL-35–dependent manner. Thus, IL-35 contributes to human Treg-mediated suppression, and its conversion of suppressed target Tconv into IL-35–induced Treg may contribute to infectious tolerance.


Journal of Immunology | 2004

Lymphocyte Activation Gene-3 (CD223) Regulates the Size of the Expanding T Cell Population Following Antigen Activation In Vivo

Creg J. Workman; Linda S. Cauley; In-Jeong Kim; Marcia A. Blackman; David L. Woodland; Dario A. A. Vignali

Lymphocyte activation gene-3 (LAG-3) is a CD4-related, activation-induced cell surface molecule that binds to MHC class II with high affinity. In this study, we used four experimental systems to reevaluate previous suggestions that LAG-3−/− mice had no T cell defect. First, LAG-3−/− T cells exhibited a delay in cell cycle arrest following in vivo stimulation with the superantigen staphylococcal enterotoxin B resulting in increased T cell expansion and splenomegaly. Second, increased T cell expansion was also observed in adoptive recipients of LAG-3−/− OT-II TCR transgenic T cells following in vivo Ag stimulation. Third, infection of LAG-3−/− mice with Sendai virus resulted in increased numbers of memory CD4+ and CD8+ T cells. Fourth, CD4+ T cells exhibited a delayed expansion in LAG-3−/− mice infected with murine gammaherpesvirus. In summary, these data suggest that LAG-3 negatively regulates T cell expansion and controls the size of the memory T cell pool.


European Journal of Immunology | 2003

The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells.

Creg J. Workman; Dario A. A. Vignali

The lymphocyte activation gene‐3 (LAG‐3, CD223) is a CD4‐related, activation‐induced cell surface molecule that binds to MHC class II with high affinity. The function of murine LAG‐3 on T cells is unclear. Here, we show that Vβ7/8+LAG‐3–/– T cells expand poorly following staphylococcal enterotoxin B (SEB) stimulation in vitro. LAG‐3–/– T cells proliferate at a normal rate, but exhibit increased cell death. Similar observations were made with LAG‐3–/–CD4+OT‐II TCR transgenic T cells following peptide stimulation. Despite reduced T cell expansion and increased cell death, LAG‐3–/–OT‐II+ T cells secrete more IL‐2 and IFN‐γ following stimulation. Antigen‐driven expansion of LAG‐3–/– T cells was restored by constitutive expression of LAG‐3 via retroviral‐mediated stem cell gene transfer. We further show that LAG‐3 function is mediated via its cytoplasmic domain, for which a conserved ‘KIEELE’ motif is essential. Our data support a role for LAG‐3 in regulating the expansion of activated T cells.


Journal of Immunology | 2002

Cutting Edge: Molecular Analysis of the Negative Regulatory Function of Lymphocyte Activation Gene-3

Creg J. Workman; Kari J. Dugger; Dario A. A. Vignali

Lymphocyte activation gene (LAG)-3 (CD223) is a CD4-related activation-induced cell surface molecule that binds to MHC class II molecules with high affinity and negatively regulates T cell expansion and homeostasis. In this study, we show that LAG-3 inhibits CD4-dependent, but not CD4-independent, T cell function via its cytoplasmic domain. Although high affinity interaction with MHC class II molecules is essential for LAG-3 function, tailless LAG-3 does not compete with CD4 for ligand binding. A single lysine residue (K468) within a conserved “KIEELE” motif is essential for interaction with downstream signaling molecules. These data provide insight into the mechanism of action of this important T cell regulatory molecule.

Collaboration


Dive into the Creg J. Workman's collaboration.

Top Co-Authors

Avatar

Dario A. A. Vignali

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Lauren W. Collison

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Kate M. Vignali

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea L. Szymczak-Workman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Charles Drake

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jonathan Powell

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yao Wang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Karen Forbes

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge