Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Cutting Edge: Anti-CD25 Monoclonal Antibody Injection Results in the Functional Inactivation, Not Depletion, of CD4+CD25+ T Regulatory Cells

Adam P. Kohm, Jeffrey S. McMahon, Joseph R. Podojil, Wendy Smith Begolka, Mathew DeGutes, Deborah J. Kasprowicz, Steven F. Ziegler and Stephen D. Miller
J Immunol March 15, 2006, 176 (6) 3301-3305; DOI: https://doi.org/10.4049/jimmunol.176.6.3301
Adam P. Kohm
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jeffrey S. McMahon
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joseph R. Podojil
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wendy Smith Begolka
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mathew DeGutes
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Deborah J. Kasprowicz
†Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Steven F. Ziegler
†Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephen D. Miller
*Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

CD4+CD25+ T regulatory (TR) cells are an important regulatory component of the adaptive immune system that limit autoreactive T cell responses in various models of autoimmunity. This knowledge was generated by previous studies from our lab and others using TR cell supplementation and depletion. Contrary to dogma, we report here that injection of anti-CD25 mAb results in the functional inactivation, not depletion, of TR cells, resulting in exacerbated autoimmune disease. Supporting this, mice receiving anti-CD25 mAb treatment display significantly lower numbers of CD4+CD25+ T cells but no change in the number of CD4+FoxP3+ TR cells. In addition, anti-CD25 mAb treatment fails to both reduce the number of Thy1.1+ congenic CD4+CD25+ TR cells or alter levels of CD25 mRNA expression in treatment recipients. Taken together, these findings have far-reaching implications for the interpretation of all previous studies forming conclusions about CD4+CD25+ TR cell depletion in vivo.

CD4+CD25+ T regulatory (TR)3cells are a member of the growing family of regulatory cell populations that serve to limit the activation, trafficking, and/or effector function of both CD4+ and CD8+ T cells (1, 2). Although the exact mechanism by which TR cells serve to limit T cell functionality remains unknown, IL-10 production, surface CTLA-4 expression, IL-2 sequestration, costimulatory molecule blockade, and surface/secreted TGF-β expression are all proposed mechanisms by which TR cells may down-regulate effector CD4+ and CD8+ T cell responses. Regardless of the exact mechanism of action, TR cells are believed to contribute to the protective processes that govern susceptibility to, progression of, and remission from various autoimmune diseases.

TR cells were described originally as CD4+ T cells that coexpress CD25 and high levels of CD62L in naive mice and are now more appropriately characterized as CD4+CD25+FOXP3+ cells (3). Recent studies have revealed a functional role for CD25 expression on TR cells such that interruption of the IL-2R/IL-2 signaling pathway blocks TR effector function potentially via alterations in the expression of the glucocorticoid-induced TNFR-family gene (GITR or TNFRSF18) (4, 5). Accordingly, a number of groups have targeted CD25 as a mechanism of depleting TR cells and studying resultant effects on T cell activation, trafficking, and/or effector function. It is widely believed that injection of anti-CD25 mAb results in the rapid and efficient depletion of CD4+CD25+ TR cells as determined by secondary staining with a mAb directed against a different CD25 epitope (6, 7, 8).

In the current study, we report that in vivo injection of anti-CD25 mAb fails to physically deplete CD4+CD25+ TR cells but, alternatively, down-regulates and/or induces shedding of CD25 from the surface of TR cells, resulting in exacerbated acute clinical experimental autoimmune encephalomyelitis (EAE). This conclusion is supported by our findings that anti-CD25 mAb treatment decreases the number of CD4+CD25+, but not CD4+FOXP3+ TR cells. These findings were confirmed using Thy1.1+ CD4+CD25+ congenic TR cells adoptively transferred into naive Thy 1.2+ recipients before anti-CD25 mAb treatment, which decreased the number of CD25+, but not Thy1.1+, CD4+ T cells. In light of the functional dependence of TR cells on CD25 expression, our data suggest that injection of anti-CD25 mAb induces functional inactivation, but not physical depletion, of CD4+CD25+ TR cells.

Materials and Methods

Mice and materials

SJL/J female mice, 5–6 wk old, were purchased from Harlan Sprague Dawley and SJL-Thy1a congenic mice were bred in the Northwestern University Center for Comparative Medicine (Chicago, IL).

Induction and clinical evaluation of proteolipid protein (PLP)139–151-induced EAE

Six- to 7-wk-old female mice were immunized s.c. with 200 μl of an emulsion containing 800 μg of Mycobacterium tuberculosis H37Ra (Difco) and a suboptimal dose (25 μg) of PLP139–151 distributed over three spots on the flanks. Individual animals were observed daily, and clinical scores were assessed in a blinded fashion on a 0–5 scale as follows: 0, no abnormality; 1, limp tail; 2, limp tail and hind limb weakness; 3, hind limb paralysis; 4, hind limb paralysis and forelimb weakness; and 5, moribund. The data are reported as the mean daily clinical score. In vitro ELISPOT assays were performed as described previously (9).

Immunohistochemistry and immunofluorescence

CNS immunohistochemistry was performed as described previously (9). For the detection of CD4+CD25+FOXP3+ T cells, single-cell suspensions were washed and first incubated with Abs directed against CD4 (L3T4; BD Biosciences) and CD25 (7D4 or PC61; BD Biosciences) for 60 min before cell permeabilization and incubation with anti-FOXP3 (FJK-16s; eBioscience) for 30 min per the manufacturer’s specifications. Fluorescent staining was analyzed using a LSRII flow cytometer and CellQuest Pro Analysis software (BD Biosciences).

Statistical analysis

Comparisons of clinical scores between the various treatment groups were analyzed by unpaired Student’s t test. Values of p < 0.01 were considered significant.

Results and Discussion

The capacity of CD4+CD25+ TR cells to efficiently influence effector T cell function has stimulated a significant level of interest in their potential to regulate immune responses during infection, autoimmune disease, and cancer. Using various experimental models, multiple groups have reached the common conclusion that CD4+CD25+ TR cells suppress autoreactive T cell effector function and autoimmune disease progression (10). More specifically, we have shown previously that supplementation of the TR cell population in naive mice confers protection from subsequent development/induction of EAE (11, 12).

To address the contribution of endogenous TR cells in regulating EAE onset and progression, we first examined the phenotype and distribution of TR cell populations at various times throughout the clinical disease course of EAE. Histological analysis revealed an influx of FOXP3+ cells into the CNS at times corresponding to the onset of clinical disease symptoms (Fig. 1⇓, A and B), and these cells appeared to be localized directly within disease lesions, as indicated by the paucity of PLP staining. This detection of TR cells within the CNS target organ suggested that endogenous TR cells may regulate the acute clinical disease phase. To test this, mice were depleted of CD4+CD25+ TR cells at various times either before or after disease induction and were followed for clinical disease progression. As seen in Fig. 1⇓C, anti-CD25 mAb injection at times either before, corresponding with, or after suboptimal disease induction (25 μg of PLP139–151) resulted in significant exacerbation of clinical disease incidence and severity, compared with the minimal disease noted in untreated mice. Accordingly, anti-CD25 mAb injection also enhanced the effector function of PLP139–151-specific T cells, as measured both by the level of IFN-γ produced (data not shown) and the number of IFN-γ-producing cells following in vitro restimulation with PLP139–151 (Fig. 1⇓D). These findings suggest that endogenous TR cells regulate normal EAE disease onset/progression, potentially via their presence in the CNS.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Regulatory role of TR cells during EAE. A and B, CNS TR cell infiltration during EAE. Naive SJL/J mice were immunized with 50 μg of PLP139–151/CFA s.c. on day 0 and then sacrificed at the onset of clinical disease for CNS histology. FOXP3+ T cells (red) were visualized in cerebellar tissue isolated from either naive (A) or diseased (B) mice. Tissue sections also were stained with PLP (green) and 4′,6′-diamidino-2-phenylindole (blue) as counterstains. Note the presence of FOXP3+ T cells within non-PLP staining areas of demyelinating lesions in diseased mice. All data are representative of three separate experiments. C, Effect of anti-CD25 mAb treatment on suboptimal EAE. Groups of five naive SJL/J mice were immunized with a suboptimal dose (25 μg) of the immunodominant epitope of PLP (PLP139–151) on day 0 and followed for clinical disease. Mice also received two injections of anti-CD25 mAb (7D4; 500 μg/injection i.p.) on either days −4 and −2 (pretreatment), 0 and 2 (induction), or 8 and 10 (postinduction) following disease induction. Data are presented as the mean clinical disease score of mice in each treatment group, and disease incidence is indicated by the numbers in parentheses. D, Effect of anti-CD25 mAb treatment on autoreactive CD4+ T cell effector function. Spleen and LN samples were isolated from mice at day 17 following disease induction and analyzed ex vivo for the number of IFN-γ-producing cells. Data are presented as the number of IFN-γ-secreting cells in response to PLP139–151.

The injection of anti-CD25 mAb treatment to physically “deplete” CD4+CD25+ TR cells in vivo is common practice. We have reported previously that injection of anti-CD25 mAb results in an apparent rapid, but short-lived, depletion of CD4+CD25+ TR cells, such that normal levels of CD4+CD25+ TR cells are observed within 10–14 days following treatment (13). However, the short-lived nature of this Ab-mediated depletion raised the question about the true mechanism of anti-CD25 mAb in vivo, because the de novo recovery of the CD4+CD25+ TR cell population should require >14 days to return to control levels. To address this, naive mice were injected with anti-CD25 mAb, followed by phenotypic analysis of the secondary lymphoid tissue to determine any effects of the treatment on the CD4+CD25+FOXP3+ T cell population. As expected, the number of CD4+CD25+ T cells was significantly decreased in vivo within 3 days of treatment but returned to control levels by day 10 following anti-CD25 mAb injection (Fig. 2⇓A). This relatively quick recovery of the TR cell population is in direct agreement with our previous findings (13). However, injection of anti-CD25 mAb failed to influence the number of CD4+FOXP3+ cells (Fig. 2⇓, B–D). In light of the rapid recovery of the CD4+CD25+ T cell population following anti-CD25 mAb treatment and the fact that the majority of CD4+CD25+ T cells also express FOXP3, it appeared that anti-CD25 mAb treatment was not physically depleting CD4+CD25+ TR cells. To validate these findings, we adoptively transferred sort-purified Thy1.2+ CD4+CD25+ TR cells into naive Thy1.1+ recipient mice before treatment with anti-CD25 mAb. In agreement with the findings above, anti-CD25 mAb injection decreased the percentage of CD4+CD25+ (Fig. 2⇓E), but not CD4+Thy1.2+ (Fig. 2⇓F), T cells in treated recipients. Thus, contrary to dogma, these data indicate that anti-CD25 mAb treatment enhances effector T cell function via mechanisms distinct from physical depletion of CD4+CD25+FOXP3+ TR cells in vivo.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

CD25 Expression on TR cells following anti-CD25 mAb treatment. A–D, Anti-CD25 mAb treatment exerts differential effects on the detection of CD4+CD25+ and CD4+FOXP3+ cell populations. Naive mice were injected with anti-CD25 mAb (7D4; 500 μg/injection i.p.) on days −2 and 0. Spleen and LN samples were then isolated 3, 6, and 10 days following treatment and then analyzed by flow cytometry (PC61 clone used for CD25 visualization) for the percentage of CD4+CD25+ (A) or CD4+FOXP3+ (B) cells. Data are presented as mean percent positive for each group. Spleen samples also were isolated from either control (C) or anti-CD25 mAb-injected (D) animals on day 2 following treatment to visualize CD25 and FOXP3 expression by immunohistochemistry. Isotype controls revealed no background staining, and data are representative of two separate experiments. E and F, Anti-CD25 mAb treatment does not deplete CD4+CD25+Thy1.2+ T cells. CD4+CD25+ TR cells were sort-purified from naive SJL-Thy1b mice and adoptively transferred into SJL-Thy1a congenic recipients before anti-CD25 mAb treatment (clone 7D4) as described above. Data are presented as the percentage of CD25+ (E) and Thy1.2+ (F) CD4+ T cells in treatment recipients and are representative of three separate experiments.

Because the primary technique for detecting TR cells involves the colocalization of CD4 and CD25 surface protein expression, it is possible that anti-CD25 mAb-mediated alterations in the level of CD25 expressed on the surface of TR cells may give the false impression of physical depletion. This is supported by the observation that both clones of anti-CD25 (7D4 and PC61) decreased the level of cell surface CD25 protein expression, but not CD25 or FOXP3 mRNA expression in the spleen and lymph nodes (LN)of treated recipients (Figs. 3⇓, A and B). In light of these findings, possible mechanisms to explain the decreased level of in vivo CD25 expression following anti-CD25 mAb treatment include the following: 1) exposure of TR cells to anti-CD25 mAb leads to internalization and/or shedding of the CD25 molecule from the cell surface in response to the anti-CD25 mAb treatment; or 2) the two clones of anti-CD25 mAb used for depletion and subsequent detection compete for the same epitope.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Effect of anti-CD25 mAb treatment on CD25 and FOXP3 mRNA expression. Naive SJL/J mice were injected with anti-CD25 mAb (7D4 or PC61; 500 μg/injection i.p.) on days −2 and 0. Spleen and LN samples were then isolated 5 days following treatment and analyzed by real-time PCR for the level of CD25 (A) and FOXP3 (B) mRNA expression. Data are presented as the amount of CD25 or FOXP3 mRNA (ag). Data are representative of two separate experiments.

To address these possibilities, we initially performed in vitro studies examining the kinetics of CD25 down-regulation, which revealed a significant decrease in the level of CD25 surface expression as early as 30 min following injection of anti-CD25 mAb (data not shown). To determine whether the Ab treatment induced internalization of CD25, spleen and LN cells were analyzed for both extracellular and extracellular/intracellular (total) CD25 expression at varying times following injection of anti-CD25 mAb in vivo. Importantly, CD4+FOXP3+ TR cells displayed significantly decreased levels of both surface (extracellular) and total (intracellular/extracellular) CD25 expression 24 h following anti-CD25 mAb injection (Fig. 4⇓A). Surprisingly, the level of CD25 expression was lower in both intact and anti-CD25 treated mice when we measured the combined intracellular/extracellular expression of the receptor in comparison to extracellular expression only. However, this decreased expression was most likely due to fixative-induced interference with the CD25 staining protocol. Intracellular FOXP3 expression was not altered by either injection of anti-CD25 mAb or by the technique of measuring extracellular vs intracellular/extracellular CD25 expression (Fig. 4⇓B). Intracellular/extracellular CD25 expression also was significantly decreased as early as 30 min following in vitro exposure to anti-CD25 mAb (data not shown), and importantly, the two clones of anti-CD25 mAb (PC61 and 7D4) used for depletion and subsequent detection were shown to not compete for a common binding epitope (data not shown). Finally, Western blot analysis of whole cellular protein lysates of CD4+CD25+ TR cells isolated 24 h following either injection of anti-CD25 mAb in vivo (Fig. 4⇓C) or the addition of anti-CD25 mAb in vitro (data not shown) revealed a decrease in total cellular CD25 expression as early as 24 h or 30 min, respectively.

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Mechanism of anti-CD25 mAb-induced regulation of CD25 expression. A and B, Intracellular/extracellular staining for CD25 following anti-CD25 mAb treatment. Naive mice were injected with anti-CD25 mAb (7D4; 500 μg/injection i.p.) on days −2 and 0. Spleen and LN samples were then isolated 24 h following treatment and analyzed by flow cytometry (PC61 clone used for CD25 visualization) for the percentage of CD4+CD25+ (A) or CD4+FOXP3+ (B) cells. In some samples, cells were permeabilized before CD25 detection to measure both intracellular and extracellular CD25 expression. Data are presented as mean percent positive for each phenotype. C, Anti-CD25 mAb treatment decreases total CD25 cellular protein expression in vivo. Naive mice were injected with anti-CD25 mAb (7D4; 500 μg/injection i.p.) on days −2 and 0, and then total cellular lysates were analyzed by Western blot for either CD25 or β-actin expression 24 h later. All data are representative of three separate experiments.

The findings that anti-CD25 mAb treatment decreases both the level of CD25 surface and total protein expression, in the absence of alterations in either CD25 mRNA expression or FOXP3 protein expression, supports the conclusion that injection of anti-CD25 mAb does not induce the physical depletion of CD4+CD25+ TR cells, but rather down-regulates CD25 cell surface expression. This observation appears to directly contradict earlier findings from our lab and others reporting that anti-CD25 mAb treatment results in both TR cell depletion and exacerbated immune responses (7). However, one important consideration is that IL-2 binding of CD25 is critical to the activation/function of TR cells (5). Thus, our current findings suggest that increased T cell effector function following anti-CD25 mAb treatment results from Ab-induced shedding of the CD25 molecule and the subsequent functional inactivation of CD4+CD25+ TR cells as opposed to their physical depletion.

One interesting observation is that anti-CD25 mAb treatment decreased the level of cell surface CD25 expression without altering either the level of intracellular CD25 protein or CD25 mRNA expression (Figs. 3⇑ and 4⇑). This finding suggests that CD25 may be shed from the surface of TR cells, rather than internalized, following anti-CD25 mAb treatment. In agreement with this, it is well established that IL-2 binding results in the shedding of CD25 (14, 15, 16). Consequently, additional studies are needed to investigate the mechanism by which anti-CD25 mAb binding of the IL-2R results in receptor shedding and the potential of long-term functional consequences of IL-2R shedding on both CD4+CD25+ TR and CD4+ effector cell function.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported in part by U.S. Public Health Service National Institutes of Health Research Grant NS-048411 and National Multiple Sclerosis Society Research Grant RG-A-3489.

  • ↵2 Address correspondence and reprint requests to Dr. Stephen D. Miller, Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611. E-mail address: s-d-miller{at}northwestern.edu

  • ↵3 Abbreviations used in this paper: TR, CD4+CD25+ T regulatory; EAE, experimental autoimmune encephalomyelitis; PLP, proteolipid protein; LN, lymph node.

  • Received December 14, 2005.
  • Accepted January 9, 2006.
  • Copyright © 2006 by The American Association of Immunologists

References

  1. ↵
    von Boehmer, H.. 2005. Mechanisms of suppression by suppressor T cells. Nat. Immunol. 6: 338-344.
    OpenUrlCrossRefPubMed
  2. ↵
    Shevach, E. M.. 2002. CD4+CD25+ suppressor T cells: More questions than answers. Nat. Rev. Immunol. 2: 389-400.
    OpenUrlPubMed
  3. ↵
    Fontenot, J. D., M. A. Gavin, A. Y. Rudensky. 2003. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 4: 330-336.
    OpenUrlCrossRefPubMed
  4. ↵
    Kohm, A. P., J. R. Podojil, J. S. Williams, J. S. McMahon, S. D. Miller. 2005. CD28 regulates glucocorticoid-induced TNF receptor family-related gene (GITR) expression on CD4+ T cells via IL-2 dependent mechanisms. Cell. Immunol. 235: 56-64.
    OpenUrlCrossRefPubMed
  5. ↵
    Thornton, A. M., E. E. Donovan, C. A. Piccirillo, E. M. Shevach. 2004. Cutting edge: IL-2 is critically required for the in vitro activation of CD4+CD25+ T cell suppressor function. J. Immunol. 172: 6519-6523.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    McHugh, R. S., E. M. Shevach. 2002. Cutting edge: depletion of CD4+CD25+ regulatory T cells is necessary, but not sufficient, for induction of organ-specific autoimmune disease. J. Immunol. 168: 5979-5983.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Onizuka, S., I. Tawara, J. Shimizu, S. Sakaguchi, T. Fujita, E. Nakayama. 1999. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor-α) monoclonal antibody. Cancer Res. 59: 3128-3133.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Kohm, A. P., J. S. Williams, S. D. Miller. 2004. Cutting Edge: Ligation of the glucocorticoid-induced TNF receptor enhances autoreactive CD4+ T cell activation and experimental autoimmune encephalomyelitis. J. Immunol. 172: 4686-4690.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Tompkins, S. M., J. Padilla, M. C. Dal Canto, J. P. Ting, L. Van Kaer, S. D. Miller. 2002. De novo central nervous system processing of myelin antigen is required for the initiation of experimental autoimmune encephalomyelitis. J. Immunol. 168: 4173-4183.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Lan, R. Y., A. A. Ansari, Z. X. Lian, M. E. Gershwin. 2005. Regulatory T cells: development, function and role in autoimmunity. Autoimmun. Rev. 4: 351-363.
    OpenUrlCrossRefPubMed
  11. ↵
    Kohm, A. P., P. A. Carpentier, H. A. Anger, S. D. Miller. 2002. Cutting Edge: CD4+CD25+ regulatory T cells suppress antigen-specific autoreactive immune responses and central nervous system inflammation during active experimental autoimmune encephalomyelitis. J. Immunol. 169: 4712-4716.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Kohm, A. P., P. A. Carpentier, S. D. Miller. 2003. Regulation of experimental autoimmune encephalomyelitis (EAE) by CD4+CD25+ regulatory T cells. Novartis Found. Symp. 252: 45-52.
    OpenUrlCrossRefPubMed
  13. ↵
    Kohm, A. P., J. S. Williams, A. L. Bickford, J. S. McMahon, L. Chatenoud, J. F. Bach, J. A. Bluestone, S. D. Miller. 2005. Treatment with nonmitogenic anti-CD3 monoclonal antibody induces CD4+ T cell unresponsiveness and functional reversal of established experimental autoimmune encephalomyelitis. J. Immunol. 174: 4525-4534.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Karlsson, H., L. Nassberger. 1995. Influence of compounds affecting synthesis, modification and transport of proteins on the expression and release of interleukin-2 receptor. Immunol. Cell Biol. 73: 81-88.
    OpenUrlCrossRefPubMed
  15. ↵
    Mullberg, J., C. T. Rauch, M. F. Wolfson, B. Castner, J. N. Fitzner, C. Otten-Evans, K. M. Mohler, D. Cosman, R. A. Black. 1997. Further evidence for a common mechanism for shedding of cell surface proteins. FEBS Lett. 401: 235-238.
    OpenUrlCrossRefPubMed
  16. ↵
    Raqib, R., A. A. Lindberg, L. Bjork, P. K. Bardhan, B. Wretlind, U. Andersson, J. Andersson. 1995. Down-regulation of γ-interferon, tumor necrosis factor type I, interleukin 1 (IL-1) type I, IL-3, IL-4, and transforming growth factor β type I receptors at the local site during the acute phase of Shigella infection. Infect. Immun. 63: 3079-3087.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 176 (6)
The Journal of Immunology
Vol. 176, Issue 6
15 Mar 2006
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Cutting Edge: Anti-CD25 Monoclonal Antibody Injection Results in the Functional Inactivation, Not Depletion, of CD4+CD25+ T Regulatory Cells
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Cutting Edge: Anti-CD25 Monoclonal Antibody Injection Results in the Functional Inactivation, Not Depletion, of CD4+CD25+ T Regulatory Cells
Adam P. Kohm, Jeffrey S. McMahon, Joseph R. Podojil, Wendy Smith Begolka, Mathew DeGutes, Deborah J. Kasprowicz, Steven F. Ziegler, Stephen D. Miller
The Journal of Immunology March 15, 2006, 176 (6) 3301-3305; DOI: 10.4049/jimmunol.176.6.3301

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Cutting Edge: Anti-CD25 Monoclonal Antibody Injection Results in the Functional Inactivation, Not Depletion, of CD4+CD25+ T Regulatory Cells
Adam P. Kohm, Jeffrey S. McMahon, Joseph R. Podojil, Wendy Smith Begolka, Mathew DeGutes, Deborah J. Kasprowicz, Steven F. Ziegler, Stephen D. Miller
The Journal of Immunology March 15, 2006, 176 (6) 3301-3305; DOI: 10.4049/jimmunol.176.6.3301
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results and Discussion
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Cutting Edge: Reduced Adenosine-to-Inosine Editing of Endogenous Alu RNAs in Severe COVID-19 Disease
  • Cutting Edge: EPHB2 Is a Coreceptor for Fungal Recognition and Phosphorylation of Syk in the Dectin-1 Signaling Pathway
  • Cutting Edge: Hypoxia-Induced Ubc9 Promoter Hypermethylation Regulates IL-17 Expression in Ulcerative Colitis
Show more CUTTING EDGE

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606