Network


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

Hotspot


Dive into the research topics where Rebecca S. McHugh is active.

Publication


Featured researches published by Rebecca S. McHugh.


Immunity | 2002

CD4+CD25+ Immunoregulatory T Cells: Gene Expression Analysis Reveals a Functional Role for the Glucocorticoid-Induced TNF Receptor

Rebecca S. McHugh; Matthew J. Whitters; Ciriaco A. Piccirillo; Deborah A. Young; Ethan M. Shevach; Mary Collins; Michael C. Byrne

CD4(+)CD25(+) immunoregulatory T cells represent a unique lineage of thymic-derived cells that potently suppress both in vitro and in vivo effector T cell function. We analyzed CD4(+)CD25(+) and CD4(+)CD25(-) T cells by DNA microarray, identifying 29 genes differentially expressed in the resting subpopulations, and 77 that were differentially expressed following activation. Most of these genes were elevated in the CD4(+)CD25(+) population, suggesting a previously activated phenotype. Among these were a number of genes that antagonize signaling, including members of the SOCS family, which may contribute to their anergic phenotype. Multiple cell surface receptors also had increased expression in CD4(+)CD25(+) cells, including GITR, a member of the TNF receptor superfamily. Importantly, antibodies to GITR abrogated suppression, demonstrating a functional role for this receptor in regulating the CD4(+)CD25(+) T cell subset.


Journal of Experimental Medicine | 2002

CD4+CD25+ Regulatory T Cells Can Mediate Suppressor Function in the Absence of Transforming Growth Factor β1 Production and Responsiveness

Ciriaco A. Piccirillo; John J. Letterio; Angela M. Thornton; Rebecca S. McHugh; Mizuko Mamura; Hidekazu Mizuhara; Ethan M. Shevach

CD4+CD25+ regulatory T cells inhibit organ-specific autoimmune diseases induced by CD4+CD25−T cells and are potent suppressors of T cell activation in vitro. Their mechanism of suppression remains unknown, but most in vitro studies suggest that it is cell contact–dependent and cytokine independent. The role of TGF-β1 in CD4+CD25+ suppressor function remains unclear. While most studies have failed to reverse suppression with anti–transforming growth factor (TGF)-β1 in vitro, one recent study has reported that CD4+CD25+ T cells express cell surface TGF-β1 and that suppression can be completely abrogated by high concentrations of anti–TGF-β suggesting that cell-associated TGF-β1 was the primary effector of CD4+CD25+-mediated suppression. Here, we have reevaluated the role of TGF-β1 in CD4+CD25+-mediated suppression. Neutralization of TGF-β1 with either monoclonal antibody (mAb) or soluble TGF-βRII-Fc did not reverse in vitro suppression mediated by resting or activated CD4+CD25+ T cells. Responder T cells from Smad3−/− or dominant-negative TGF-β type RII transgenic (DNRIITg) mice, that are both unresponsive to TGF-β1–induced growth arrest, were as susceptible to CD4+CD25+-mediated suppression as T cells from wild-type mice. Furthermore, CD4+CD25+ T cells from neonatal TGF-β1−/− mice were as suppressive as CD4+CD25+ from TGF-β1+/+ mice. Collectively, these results demonstrate that CD4+CD25+ suppressor function can occur independently of TGF-β1.


Immunological Reviews | 2001

Control of T‐cell activation by CD4+ CD25+ suppressor T cells

Ethan M. Shevach; Rebecca S. McHugh; Ciriaco A. Piccirillo; Angela M. Thornton

Summary: Depletion of the minor (∼10%) subpopulation of CD4+ T cells that co‐expresses CD25 (interleukin (IL)‐2 receptor α‐chain) by thymectomy of neonates on the third day of life or by treatment of adult CD4+ T cells with anti‐CD25 and complement results in the development of organ‐specific autoimmunity. Autoimmune disease can be prevented by reconstitution of the animals with CD4+ CD25+ cells. CD4+ CD25+‐mediated protection of autoimmune gastritis does not require the suppressor cytokines IL‐4, IL‐10, or transforming growth factor (TGF)‐β. Mice that express a transgenic T‐cell receptor (TCR) derived from a thymectomized newborn that recognizes the gastric parietal cell antigen H/K ATPase all develop severe autoimmune gastritis very early in life. CD4+ CD25+ T cells are also powerful suppressors of the activation of both CD4+ and CD8+ T cells in vitro. Suppression is mediated by a cell contact‐dependent, cytokine‐independent T–T interaction. Activation of CD4+ CD25+ via their TCR generates suppressor effector cells that are capable of non‐specifically suppressing the activation of any CD4+ or CD8+ T cell. Activation of suppressor effector function is independent of co‐stimulation mediated by CD28/CTLA‐4 interactions with CD80/CD86. We propose that CD4+ CD25+ T cells recognize organ‐specific antigens, are recruited to sites of autoimmune damage where they are activated by their target antigen, and then physically interact with autoreactive CD4+ or CD8+ effector cells to suppress the development of autoimmune disease.


Journal of Immunology | 2004

Engagement of Glucocorticoid-Induced TNFR Family-Related Receptor on Effector T Cells by its Ligand Mediates Resistance to Suppression by CD4+CD25+ T Cells

Geoffrey L. Stephens; Rebecca S. McHugh; Matthew J. Whitters; Deborah A. Young; Deborah Luxenberg; Beatriz M. Carreno; Mary Collins; Ethan M. Shevach

Nonactivated CD4+CD25+ regulatory T cells constitutively express glucocorticoid-induced TNFR family-related receptor (GITR), a TNFR family member whose engagement was presumed to abrogate regulatory T cell-mediated suppression. Using GITR−/− mice, we report that GITR engagement on CD25−, not CD25+ T cells abrogates T cell-mediated suppression. Mouse APCs constitutively express GITR ligand (GITR-L), which is down-regulated following TLR signaling in vivo. Although GITR−/−CD25− T cells were capable of mounting proliferative responses, they were incapable of proliferation in the presence of physiological numbers of CD25+ T cells. Thus, GITR-L provides an important signal for CD25− T cells, rendering them resistant to CD25+-mediated regulation at the initiation of the immune response. The down-regulation of GITR-L by inflammatory stimuli may enhance the susceptibility of effector T cells to suppressor activity during the course of an infectious insult.


Journal of Experimental Medicine | 2002

Constitutive Presentation of a Natural Tissue Autoantigen Exclusively by Dendritic Cells in the Draining Lymph Node

Clemens Scheinecker; Rebecca S. McHugh; Ethan M. Shevach; Ronald N. Germain

The major histocompatibility complex (MHC)-dependent presentation of processed tissue-specific self-antigens can contribute to either peripheral (extrathymic) tolerance or the differentiation of autoreactive T cells. Here, we have studied the MHC class II molecule presentation of gastric parietal cell (PC)-specific H+/K+-ATPase, which induces a destructive autoimmune gastritis in BALB/c mice lacking CD4+ CD25+ regulatory T cells. Immunofluorescence microscopy showed physical association of CD11c+ dendritic cells (DCs) with PCs in the gastric mucosa. H+/K+-ATPase protein was found within vesicular compartments of a few CD11c+ DCs only in the draining gastric lymph node (LN) and these antigen-containing DCs increased markedly in number with the onset of tissue destruction in autoimmune animals. Both CD8αhi and CD8αlo gastric DCs, but not peripheral or mesenteric DCs, showed evidence of constitutive in vivo processing and presentation of H+/K+-ATPase. These data provide direct support for a widely held model of local tissue antigen uptake and trafficking by DCs in normal animals and demonstrate that DCs in the draining LN can present a tissue-specific self-antigen under noninflammatory conditions without fully deleting autoreactive T cells or inducing active autoimmunity.


Journal of Immunology | 2002

Cutting Edge: Depletion of CD4+CD25+ Regulatory T Cells Is Necessary, But Not Sufficient, for Induction of Organ-Specific Autoimmune Disease

Rebecca S. McHugh; Ethan M. Shevach

Thymectomy of BALB/c mice on day 3 of life results in the development of autoimmune gastritis (AIG) due to the absence of CD4+CD25+ regulatory T cells. However, depletion of CD4+CD25+ T cells by treatment with anti-CD25 rarely resulted in AIG. Depletion was efficient, as transfer of splenocytes from depleted mice induced AIG in nu/nu mice. One explanation for this result is that CD4+CD25− T cells upon transfer to nude recipients undergo lymphopenia-induced proliferation, providing a signal for T cell activation. Cotransfer of CD25+ T cells did not inhibit initial proliferation but did suppress AIG. Surprisingly, immunization with the AIG target Ag, H/K ATPase, in IFA failed to induce disease in normal animals but induced severe AIG in CD25-depleted mice. These results demonstrate that second signals (nonspecific proliferation, TCR activation, or inflammation) are needed for induction of autoimmunity in the absence of CD25+ regulatory T cells.


Immunity | 2003

Neonates Support Lymphopenia-Induced Proliferation

Booki Min; Rebecca S. McHugh; Gregory D. Sempowski; Crystal L. Mackall; Gilles Foucras; William E. Paul

T cells expand without intentional antigen stimulation when transferred into adult lymphopenic environments. In this study, we show that the physiologic lymphopenic environment existing in neonatal mice also supports CD4 T cell proliferation. Strikingly, naive CD4 T cells that proliferate within neonates acquire the phenotypic and functional characteristics of memory cells. Such proliferation is inhibited by the presence of both memory and naive CD4 T cells, is enhanced by 3-day thymectomy, is independent of IL-7, and requires a class II MHC-TCR interaction and a CD28-mediated signal. CD44(bright) CD4 T cells in neonates have a wide repertoire as judged by the distribution of Vbeta expression. Thus, lymphopenia-induced T cell proliferation is a physiologic process that occurs during the early postnatal period.


European Journal of Immunology | 1999

Post‐thymectomy autoimmune gastritis: fine specificity and pathogenicity of anti‐H/K ATPase‐ reactive T cells

Elisabeth Suri-Payer; Anna Z. Amar; Rebecca S. McHugh; Kannan Natarajan; David H. Margulies; Ethan M. Shevach

Thymectomy at day 3 of life (d3Tx) results in the development of organ‐specific autoimmunity. We have recently shown that d3Tx BALB/c mice which develop autoimmune gastritis contain CD4+ T cells specific for the gastric parietal cell proton pump, H/K ATPase. Here, we demonstrate that freshly explanted gastric lymph node (LN) cells from d3Tx mice react significantly to the H/K ATPase α chain, but only marginally to the β chain. Two H/K ATPase‐reactive T cell lines were derived from the gastric LN of d3Tx mice. Both are CD4+ , TCR α / β+ , and I‐Ad restricted, and recognize distinct peptides from the H/K ATPase α chain. One cell line secretes Th1 and the other Th2 cytokines, but both are equally potent in inducing gastritis with distinct profiles of cellular infiltration in nu/nu recipient animals. Neither of the cell lines induced disease in normal BALB/c recipients and transfer of disease to nu/nu recipients was blocked by co‐transfer of normal BALB/c spleen cells containing CD4+ CD25+ cells. Although CD4+ CD25+ T cells are thought to emigrate from the thymus after day 3 of life, they could be identified in LN of 2‐day‐old animals. The capacity of CD4+ CD25+ T cells to abrogate the pathogenic activity in vivo of both activated Th1/Th2 lines strongly suggests that this suppressor T cell population may have a therapeutic role in other models of established autoimmunity. The availability of well‐characterized lines of autoantigen‐specific T cells should greatly facilitate the analysis of the mechanism of action and target of the CD4+ CD25+ immunoregulatory cells.


European Journal of Immunology | 2001

A T cell receptor transgenic model of severe, spontaneous organ-specific autoimmunity

Rebecca S. McHugh; Ethan M. Shevach; David H. Margulies; Kannan Natarajan

The development of mouse models of human organ‐specific autoimmune diseases has been hampered by the need to immunize mice with autoantigens in potent adjuvants. Even autoantigen‐specific T cell receptor transgenic models of autoimmunity have proven to be complex as the transgenic mice frequently fail to develop disease spontaneously. We have isolated a CD4+ T cell clone (TxA23)that recognizes the gastric parietal cell antigen, H/K ATPase α‐chain630–641, from a mouse with autoimmune gastritis that developed after thymectomy on day 3 of life. The T cell receptor α and β genes from this clone were used to generate A23 transgenic mice. All A23 transgenic animals spontaneously developed severe autoimmune gastritis, and evidence of disease was detected as early as day 10 of life. Gastritis could be transferred to immunocompromised mice with a limited number of transgenic thymocytes (103), but as many as 107 induced only mild disease in wild‐type animals. Due to the complete penetrance of spontaneous disease, identity of the auto‐antigen, susceptibility to immunoregulation, and close relation to autoimmune gastritis in man, A23 transgenic mice represent a unique CD4+ T cell‐mediated disease model for understanding the multiple factors regulating organ‐specific autoimmunity.


Novartis Foundation symposium | 2003

Control of T cell activation by CD4+CD25+ suppressor T cells.

Ethan M. Shevach; Ciriaco A. Piccirillo; Angela M. Thornton; Rebecca S. McHugh

Although the concept of a separate lineage of T cells specifically equipped to suppress immune responses was initially proposed more than 30 years ago, progress in this area of immunoregulation has been hampered by the lack of solid biochemical and molecular data to support the existence of the soluble products of these purported suppressor T cells. Studies over the past 5-10 years have identified a distinct lineage of CD4+CD25+ regulatory or suppressor T cells that control autoreactive effector cells and prevent autoimmunity. The mechanism by which CD4+CD25+ T cells inhibit T cell activation in vivo or in vitro is still poorly defined. While autoreactive effector T cells undergo massive proliferation and expansion following injection into immunocompromised recipients, CD4+CD25+ T cells do not inhibit this lymphopaenia-induced proliferation and act later in the activation process at the site of immune damage in the target organ. The development of in vitro models that partially mimic the in vivo properties of the CD4+CD25+ regulatory T cells has facilitated their characterization. A member of the tumour necrosis receptor family, the GITR is expressed on CD4+CD25+ T cells and after interaction with its ligand down-regulates suppressor activity. Multiple methods of manipulating both the numbers of CD4+CD25+ suppressor T cells and their activation status are now available and will rapidly be applied to therapy of autoimmune, infectious and malignant diseases.

Collaboration


Dive into the Rebecca S. McHugh's collaboration.

Top Co-Authors

Avatar

Ethan M. Shevach

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ciriaco A. Piccirillo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Angela M. Thornton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Beatriz M. Carreno

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David H. Margulies

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Geoffrey L. Stephens

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge