Skip to main content

Main menu

  • 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
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

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

Dual Function of CD70 in Viral Infection: Modulator of Early Cytokine Responses and Activator of Adaptive Responses

Atef Allam, Melissa Swiecki, William Vermi, Jonathan D. Ashwell and Marco Colonna
J Immunol July 15, 2014, 193 (2) 871-878; DOI: https://doi.org/10.4049/jimmunol.1302429
Atef Allam
*Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Melissa Swiecki
*Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
William Vermi
*Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
†Department of Pathology, University of Brescia, Brescia 25123, Italy; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jonathan D. Ashwell
‡Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marco Colonna
*Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
  • 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

This article has a correction. Please see:

  • Errata - February 15, 2015

Abstract

The role of the TNF family member CD70 in adaptive T cell responses has been intensively studied, but its function in innate responses is still under investigation. In this study, we show that CD70 inhibits the early innate response to murine CMV (MCMV) but is essential for the optimal generation of virus-specific CD8 T cells. CD70−/− mice reacted to MCMV infection with a robust type I IFN and proinflammatory cytokine response. This response was sufficient for initial control of MCMV, although at later time points, CD70−/− mice became more susceptible to MCMV infection. The heightened cytokine response during the early phase of MCMV infection in CD70−/− mice was paralleled by a reduction in regulatory T cells (Treg). Treg from naive CD70−/− mice were not as efficient at suppressing T cell proliferation compared with Treg from naive wild-type mice, and depletion of Treg during MCMV infection in Foxp3–diphtheria toxin receptor mice or in wild-type mice recapitulated the phenotype observed in CD70−/− mice. Our study demonstrates that although CD70 is required for the activation of the antiviral adaptive response, it has a regulatory role in early cytokine responses to viruses such as MCMV, possibly through maintenance of Treg survival and function.

This article is featured in In This Issue, p.465

Introduction

Members of the TNF and TNFR family provide costimulatory signals that facilitate optimal adaptive antiviral responses (1, 2). They promote the expansion of virus-specific T cells in primary responses and the generation of a memory T cell pool for secondary responses, mostly through induction of T cell survival pathways. Interactions between TNF and TNFR family members also induce signals that influence the functional polarization of CD4 Th cells (1, 2).

CD70 is a TNF family member that is constitutively expressed on certain APCs in the thymic medulla, possibly medullary thymic epithelial cells (3), and the intestinal mucosa (4, 5). Additionally, CD70 is transiently expressed on dendritic cells (DC), B, and T cells after activation (3, 6–8). CD70 binds and activates CD27, a TNFR family member constitutively expressed on the surface of naive CD8 T cells, CD4 T cells, and subsets of NK cells and B cells (6). Many studies have demonstrated that the CD70–CD27 costimulatory interaction supports clonal expansion of CD8 T cells during primary responses and/or the long-term survival of memory CD8 T cells, depending on the nature of the Ag (7–12). By interacting with DC, CD4 Th cells induce expression of CD70 on DC, which engages CD27 on naive CD8 T cells, providing the help that CD8 T cells require to generate memory responses (13). The CD70–CD27 interaction gives CD8 T cells survival signals (14, 15), which prevent TRAIL-mediated Ag-induced cell death (13).

CD70–CD27 interaction also facilitates CD4 T cell responses and directs CD4 Th cell functional polarization, promoting Th1 cell differentiation (16–18) and inhibiting Th17 effector functions (19). A recent study has shown that the CD70–CD27 interaction enables epithelial cells and DC in the thymic medulla to induce the development of CD4+Foxp3+ regulatory T cells (Treg) (20). Finally, CD27 is highly expressed on a subset of mature NK cells endowed with great capacity to release lytic granules and IFN-γ, as well as high homeostatic proliferation compared with NK cells with low amounts of CD27 (21). Whether CD70–CD27 interaction has a substantial impact on the expansion and effector functions of these NK cells is not known.

The function of CD70–CD27 interaction in viral infections has been extensively investigated in the context of adaptive responses, leading to the conclusion that this interaction impacts primary and/or memory antiviral CD8 T cell responses, depending on the type of virus (9–11, 22–25). However, the impact of CD70–CD27 interactions on antiviral innate responses remains largely unknown. To address this issue, we studied murine CMV (MCMV) infection in CD70−/− mice. We chose MCMV infection because the response to this virus in the C57BL/6 background is largely dependent on NK cells and cytokines (i.e., IFN-α and IL-12), which are produced by APCs such as plasmacytoid DC and conventional DC (26, 27). We found that CD70−/− mice responded to MCMV infection with an early burst of IFN-α, IL-12, and other proinflammatory cytokines. Although this response was sufficient to control MCMV in the first 36 h, at later time points, CD70−/− mice became more susceptible to MCMV infection compared with their wild-type (WT) counterparts because of a defect in the adaptive response. Furthermore, we found that the augmented cytokine response during the initial phase of MCMV infection in CD70−/− mice was a consequence of reduced numbers and impaired function of Treg. Depletion of Treg during MCMV infection in Foxp3–diphtheria toxin receptor (DTR) mice or in WT mice recapitulated the phenotype observed in CD70−/− mice. Thus, our study demonstrates that although CD70 is required for the activation of the antiviral adaptive response, it has a regulatory role in early cytokine responses to viruses such as MCMV.

Materials and Methods

Mice, viral infections, and treatments

All animal studies were approved by the Washington University Animal Studies Committee. Male and female C57BL/6 and CD70−/− mice on the C57BL/6 background (25) were bred in house and used between 8 and 12 wk of age. Foxp3-DTR mice were generated by Alexander Rudensky (Memorial Sloan-Kettering Cancer Center, New York, NY) (28) and kindly provided by Chyi-Song Hsieh (Washington University, St. Louis, MO). Foxp3-DTR mice were bred in house. Smith strains MCMV and AT1.5 (Δm157) were provided by W. Yokoyama and A. French (Washington University). Mice were infected with salivary gland MCMV or AT1.5 stocks prepared from BALB/c mice (doses specified in figure legends). To deplete Treg in Foxp3-DTR mice, DT (1 μg/mouse) was injected i.p. on days −3 and −1 of infection; control mice received PBS. To deplete Treg with Ab, C57BL/6 and CD70−/− mice were injected i.p. with 300–500 μg of anti-CD25 Ab (clone PC-61.5.3; BioXCell) or a rat IgG isotype control Ab on days −3 and −1 of infection. To block CD70–CD27 interactions in vivo, we injected C57BL/6 mice i.p. with 150 μg of anti-CD70 Ab (clone FR-70; BioXCell) or a rat IgG isotype control Ab on days − 1 and 0 of infection (29). The following primers were used to quantify MCMV in tissues using TaqMan PCR: for MCMV IE1, probe, 5′-/56-FAM/TCT CTT GCC/ZEN/CCGTCCTGAAAACC/3IABkFQ/-3′; primer 1, 5′-CCC TCT CCT AAC TCT CCC TTT-3′; and primer 2, 5′-TGG TGC TCT TTT CCC GTG-3′; and for actin, probe, 5′-/56-FAM/TTC AGG GTC/ZEN/AGGATACCTCT CTT GCT/3IABKFQ/-3′; primer 1, 5′-AGC TCA TTG TAG AAG GTG TGG-3′; and primer 2, 5′-GGT GGG AAT GGG TCA GAA G-3′.

Cell preparations

Single-cell suspensions of spleens were prepared by collagenase D digestion and passage through nylon mesh cell strainers (BD Biosciences). RBCs were lysed with RBC lysis buffer (Sigma-Aldrich). Whole blood was collected by cardiac puncture, and serum was stored at −20°C until analysis.

Abs and flow cytometry

The following reagents were from BD Biosciences, eBioscience, or BioLegend: fluorochrome-labeled anti-CD69 (H1.2F3), anti-CD11c (HL3), anti-CD8α (53-6.7), anti-CD4 (GK1.5), anti–I-Ab (AF6-120.1), anti-CD3 (145-2C11), anti-NK1.1 (PK136), anti-Ly49H (3D10), anti–IFN-γ (XMG1.2), anti–granzyme B (GB11), anti-CD11b (M1/70), anti-Ly6C (AL-21), anti-Ly6G (1A8), anti-CD25 (PK61.5), anti-Foxp3 (150D/E4), anti-CD262 (DR5, MD5-1), anti-TRAIL (N2B2), and streptavidin. The m45 tetramer used to visualize virus-specific CD8 T cells was a generous gift from M. Brown (University of Virginia, Charlottesville, VA). FcRs were blocked before surface staining with supernatant from HB-197 cells (American Type Culture Collection). Dead cells were excluded with propidium iodide. Intracellular staining for IFN-γ and granzyme B was performed on freshly isolated spleen cells or after 4-h incubation with brefeldin A using the BD Cytofix-Cytoperm Kit (BD Biosciences) according to the manufacturer's instructions. All flow cytometry was conducted on a dual-laser FACSCalibur or LSR II (BD Biosciences) and analyzed with FlowJo software (Tree Star). For NK cell purification, DX5+ cells were purified from spleens with anti-DX5 microbeads as recommended by the manufacturer (Miltenyi Biotec). Purities were always >85%.

ELISA and cytometric bead array

Serum samples from infected mice were collected at various time points postinfection (p.i.). IFN-α concentrations were measured by ELISA (PBL Assay Science). IL-12p70, IL-10, IL-6, MCP-1, TNF-α, and IFN-γ were measured by flow cytometry with the Mouse Inflammation CBA kit (BD Biosciences).

Cell lines and tissue culture

YAC-1 and primary cells were cultured in RPMI 1640 with 10% bovine calf serum, 1% glutamax, 1% HEPES, 1% nonessential amino acids, 1% sodium pyruvate, 1% penicillin/streptomycin, and 1% kanamycin sulfate (Life Technologies-Invitrogen).

Cytotoxicity assays

For NK cell cytotoxicity assays, splenic NK cells purified from MCMV-infected mice were resuspended in complete RPMI 1640 and serially diluted in 96-well round-bottom plates. YAC-1 cells were labeled with [51Cr] for 2 h and then incubated with effector cells at 37°C for 4 h. [51Cr] release in supernatants was measured with a γ-counter.

Treg suppression assays

CD4+CD25+ Treg were isolated from naive C57BL/6 and CD70−/− mice with the mouse CD4+CD25+ Regulatory T Cell Isolation Kit (Miltenyi Biotec). CD4+CD25− cells from C57BL/6 mice were used as conventional T cells (Tconv). Tconv were labeled with 5 μM CFSE and cultured with Treg according to Collison and Vignali (30) with modifications. Briefly, 96-well flat-bottom bottoms were coated overnight at 4°C with anti-CD3 and anti-CD28 Abs in PBS (2.5 μg/ml). Two-fold serially diluted Treg from C57BL/6 or CD70−/− mice were plated per well, and then CFSE-labeled Tconv cells were added to each well at a concentration of 1 × 105 cells/well. Cocultures were incubated at 37°C for 72 h and then analyzed by flow cytometry. Cells were stained with propidium iodide and anti-CD4 to visualize live, proliferating CFSE+ Tconv cells.

Tissue samples and immunohistochemistry

Formalin-fixed, paraffin-embedded tissue sections were used for immunohistochemical staining to visualize T cells and B cells. Sections were stained with anti-CD3 and anti-B220. Digital images were captured and processed using Analysis Image Processing software (Olympus).

Statistical analysis

The statistical significance of differences in mean values was analyzed with unpaired, two-tailed Student t test. The p values <0.05 were considered statistically significant.

Results

CD70 deficiency has opposing consequences on MCMV clearance at early and late time points of infection

We infected CD70−/− mice and WT C57BL/6 controls i.p. with MCMV and measured viral burden in tissues. At 1.5 d p.i., CD70−/− mice showed lower MCMV titers than WT mice. However, at 3 d p.i., MCMV titers were significantly higher in spleens and livers of CD70−/− mice compared with WT controls (Fig. 1A). Because MCMV has a tropism for salivary glands, where it remains detectable for a longer period of time postinfection (31), we also measured viral titers in salivary glands on day 8 p.i. Again, viral loads were higher in CD70−/− mice than in WT controls (Fig. 1A). To determine whether CD70−/− mice were more susceptible to infection, we injected a higher dose of MCMV (2 × 105 PFU). Both CD70−/− and WT mice lost a significant amount of body weight; however, whereas 90% of WT mice recovered 5 d p.i. and survived infection, only 30% of CD70−/− mice recovered and survived (Fig. 1B). Together, these results indicate that lack of CD70 delays viral replication during the early stage of infection but, ultimately, affects MCMV clearance and survival at later time points once the infection is established.

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

Effect of CD70 deficiency on viral load and survival during MCMV infection. (A) WT and CD70−/− mice were infected i.p. with 5 × 104 PFU of MCMV. Viral loads were determined in spleen on day 1.5, in spleen and liver on day 3, or in salivary gland (SG) on day 8. (B) Body weight and survival were monitored in WT and CD70−/− mice infected i.p. with 2 × 105 PFU of MCMV (n = 10). Data are from two to four independent experiments. Statistical significance is indicated by p values.

Lack of CD70 causes an early and exuberant cytokine response to MCMV infection

We next assessed the impact of CD70 deficiency on cytokine responses to MCMV. Remarkably, serum IFN-α levels were higher in CD70−/− mice than in WT mice at 36 and 48 h p.i. (Fig. 2A); at 72 h p.i., systemic IFN-α was undetectable in both strains (data not shown). Similarly, serum IL-12p70 was ∼3-fold higher in CD70−/− mice than WT controls at 36 h p.i., but then sharply declined (Fig. 2B). Serum TNF-α (Fig. 2C) and IL-6 (Fig. 2D) also peaked at 36 h p.i., and higher levels were detected in CD70−/− mice. Taken together, these data indicate that loss of CD70 impacts both the magnitude and timing of IFN-α and proinflammatory cytokine production, resulting in an early burst of cytokine secretion in response to MCMV. Most likely, the early wave of IFN-α accounts for the transient reduction of MCMV viral load during the first 36 h of infection in CD70−/− mice.

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

CD70 modulates the inflammatory response during MCMV infection. WT and CD70−/− mice were infected i.p. with 5 × 104 PFU of MCMV. (A) Serum IFN-α levels were measured by ELISA at 36 and 48 h p.i. Proinflammatory cytokine production in mice at various time points p.i. Serum levels of IL-12p70 (B), TNF-α (C), IL-6 (D), and MCP-1 (E). (F) Monocyte recruitment to spleens of WT and CD70−/− mice after MCMV infection. Data show numbers of CD11b+Ly6ChiLy6G− cells in spleens at various time points p.i. Data are from 2–10 independent experiments. Error bars represent the mean ± SEM. Statistical significance is indicated by p values.

In parallel with the enhanced inflammatory cytokine response, serum levels of CCL2 (MCP-1) were also elevated in CD70−/− mice compared with WT mice at 24 and 36 h p.i. (Fig. 2E). CCL2 promotes the recruitment of myeloid cells; accordingly, ∼3-fold more CD11b+Ly6ChiLy6G− monocytes were observed in the spleens of CD70−/− mice at 24 and 36 h p.i., but these numbers declined rapidly (Fig. 2F). Thus, lack of CD70 also causes the rapid influx of monocytes into the spleen in response to MCMV infection.

The robust cytokine response to MCMV in CD70−/− mice results in accelerated activation of NK cells

Because NK cells play an essential role in controlling MCMV infection through lysis of infected cells and IFN-γ production (26, 27), and CD70−/− mice have defective control of MCMV infection, we wanted to determine the impact of CD70 deficiency on NK cells. Prior to infection, NK cell numbers were similar in naive CD70−/− and WT mice (data not shown). CD70−/− mice had more NK cells in the spleen than WT mice 24 h p.i., but these numbers declined more rapidly in CD70−/− mice than in WT controls, such that at 48 h p.i., CD70−/− mice had fewer NK cells than WT mice (Fig. 3A, 3B). To evaluate NK cell activation, we monitored CD69 expression. Prior to infection, NK cells from both CD70−/− and WT mice expressed low and similar levels of CD69 (data not shown). A higher frequency of CD69+ NK cells was noted in CD70−/− mice at 24 and 36 h p.i., but by 48 h p.i., the majority of NK cells in both strains of mice expressed CD69 (Fig. 3C, 3D). Considering the changes in the absolute numbers of NK cells, more CD69+ NK cells were present in the spleens of CD70−/− mice at early time points p.i., but these numbers declined very rapidly in comparison with WT mice (Fig. 3C, 3D). These results indicate that CD70 deficiency results in accelerated activation of NK cells followed by a sharp decline in NK cell numbers during MCMV infection.

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

CD70 deficiency augments NK cell activation early during infection. (A–G) WT and CD70−/− mice were infected i.p. with 5 × 104 PFU of MCMV. (A) Frequencies of NK cells (NK1.1+CD3−) and CD3+ T cells in spleens at various time points p.i. (B) Total numbers of splenic NK cells at 24, 36, and 48 h p.i. Frequencies (C) and numbers (D) of splenic CD69-expressing NK cells. Serum levels of IFN-γ (E) and frequencies and mean fluorescence intensity (MF) of IFN-γ–producing NK cells (F) in spleens after MCMV infection. (G) Cytolytic activity of NK cells was assessed in standard 4-h [51Cr]release assays. Mice were infected for 36 or 48 h with 5 × 104 PFU MCMV. NK cells were purified from spleens and incubated with YAC-1 target cells. (H and I) Mice were infected i.p. with 1 × 104 PFU of Δm157 MCMV. Viral loads in spleen and liver were determined on day 3 p.i. (H), and serum IFN-α levels were measured at 36 h p.i. (I). Data are from two to three independent experiments. Error bars represent the mean ± SEM. Statistical significance is indicated by p values.

We next analyzed the effector functions of NK cells. Serum levels of IFN-γ were 3-fold higher in CD70−/− mice than in WT mice 36 h p.i. (Fig. 3E). At this time point p.i., serum IFN-γ most likely reflects NK cell secretion. Consistent with this, the frequency and mean fluorescence intensity of IFN-γ–producing NK cells were higher in CD70−/− than in control mice at 36 h p.i. (Fig. 3F). However, at 48 h p.i., NK cells from WT mice appeared to be making more IFN-γ on a per-cell basis even though the frequencies of IFN-γ–producing NK cells were similar in both groups of mice (Fig. 3F). At higher E:T ratios, NK cells from CD70−/− mice also showed a slight increase in cytolytic capacity (Fig. 3G) and expressed more granzyme B (data not shown) at 36 h p.i. than NK cells from WT mice. At 48 h p.i., specific lysis and granzyme B expression were similar in WT and CD70−/− mice (Fig. 3G and data not shown); however, we did observe a higher frequency of NK cells that expressed TRAIL in CD70−/− mice (data not shown). Taken together, these results indicate that CD70 deficiency elicits activation and function early on during MCMV infection followed by a reduction in NK cell numbers. Most likely, this initial activation of NK cells is due to the early boost of cytokines, such as IFN-α and IL-12p70, which activate NK cells (27).

The failure of CD70−/− mice to effectively control MCMV is independent of NK cell numbers

NK cells in C57BL/6 mice recognize MCMV-infected cells through the receptor Ly49H, which detects the MCMV-encoded MHC class I–like molecule m157 (26). To determine whether the increased MCMV burden in CD70−/− mice is due to defective NK cell numbers, we infected CD70−/− and WT mice with Δm157 MCMV. This virus eludes recognition by Ly49H; therefore, its clearance is NK cell independent (26). Similar to WT MCMV, viral loads were higher in CD70−/− mice than in WT mice on day 3 p.i. (Fig. 3H), indicating that the impaired control of MCMV infection in CD70−/− mice is not directly related to a defect in NK cell numbers. Likewise, the IFN-α response to Δm157 MCMV was ∼2.5-fold higher in CD70−/− mice compared with WT mice (Fig. 3I), suggesting that the enhanced cytokine response of CD70−/− mice to MCMV is independent of NK cell recognition of MCMV.

CD70 deficiency affects CD8α DC numbers and the MCMV-specific CD8 T cell response

Because an NK cell defect may not be the culprit of impaired MCMV control in CD70−/− mice, we investigated whether CD70 deficiency affected DC–T cell interaction and adaptive responses. We first examined whether DC express CD70 during MCMV infection. At 36 h p.i., ∼30% of splenic CD8α DC expressed CD70 and by 48 h p.i. CD70 was present on 60% of splenic CD8α DC (data not shown and Fig. 4A). When we evaluated DC numbers in CD70−/− and WT mice infected with MCMV, WT mice showed a progressive decline in total DC numbers and CD8α DC in the first 72 h p.i. and CD70−/− mice exhibited a more marked decrease (Fig. 4B, 4C). At 48 h p.i., CD8α DC in CD70−/− mice were greatly reduced compared with WT controls and almost undetectable at 72 h p.i. We also observed that during MCMV infection, higher frequencies of CD8α DC in CD70−/− mice expressed the TRAIL receptor DR5 compared with WT mice (Fig. 4D). Thus, it is plausible that the marked reduction of CD8α DC in CD70−/− mice is due at least in part to activation-induced cell death.

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

DC survival and the CD8 T cell response are impaired in CD70−/− mice following MCMV infection. WT and CD70−/− mice were infected i.p. with 5 × 104 PFU of MCMV. (A) Expression of CD70 on splenic CD11chiMHC class II (MHC II)+ DC and CD8α DC 48 h p.i. Numbers of splenic CD11chiMHC II+ DC (B) and CD8α DC (C) at various time points p.i. (D) DR5 expression on splenic CD11chiMHC II+ DC 48 h p.i. (E) Numbers and frequencies of m45-specific CD8 T cells in spleens on day 7 p.i. as measured by tetramer staining. (F) Numbers of Ly49H+ NK cells in spleens on day 7 p.i. Data are representative of two independent experiments. Statistical significance is indicated by p values.

Because CD8α DC prime naive CD8 T cells against MCMV through cross presentation, we next investigated the generation of MCMV-specific CD8 T cells by m45 tetramer staining. We found a significant reduction in total and MCMV-specific CD8 T cells in MCMV-infected CD70−/− mice compared with WT mice at day 7 p.i., suggesting that the CD8 T cell response was indeed affected by the loss of CD8α DC (data not shown and Fig. 4E). Immunohistochemical analysis of spleens from CD70−/− and WT mice at day 7 p.i. confirmed a reduction of CD3+ cells in CD70−/− mice (data not shown). We envision that the defect of the adaptive T cell response, and particularly of specific CD8 T cells, is a factor contributing to the impaired MCMV control in CD70−/− mice. Interestingly, CD70−/− mice showed a modest increase in Ly49H+ NK cells compared with WT mice on day 7 p.i. (Fig. 4F), suggesting that after the initial phase of infection, MCMV-specific NK cells expand to greater numbers in CD70−/− mice as a consequence of increased viral replication and an impaired adaptive response.

Treg modulate innate responses to MCMV infection

We sought to investigate the mechanism behind the exuberant cytokine response that occurs in CD70−/− mice very early after MCMV infection. We hypothesized that lack of CD70 may release the innate immune system, particularly APCs, from an inhibitory signal such that APC become hyperresponsive to microbial stimuli that engage TLRs. It has been shown that CD70−/− mice have reduced numbers of Treg (20) and that Treg regulate DC responses in vivo, such that in the absence of Treg, DC exhibit trafficking defects to sites of infection and can be more responsive to stimulation (32–36). Therefore, we examined numbers of Treg in WT and CD70−/− mice during MCMV infection. In agreement with a previous study, naive CD70−/− mice had a slight reduction of Treg compared with WT mice (Fig. 5A) (20). Following MCMV infection, Treg were consistently ∼2- to 3-fold less in CD70−/− mice at 24, 48, and 72 h p.i. (Fig. 5B). To determine whether Treg influenced the innate response to MCMV infection, we injected Foxp-DTR mice with DT or PBS to deplete Treg or not prior to infection with MCMV. We found that DT eliminated almost all CD3+CD4+Foxp3+ cells (Fig. 5C) and that depletion of Treg resulted in elevated serum IFN-α and TNF-α but reduced IL-12p70 levels (Fig. 5D). With respect to NK cell activation and function, frequencies of NK cells from Treg-depleted mice were similar to nondepleted mice but they expressed more CD69 (Fig. 5E), consistent with the elevated levels of IFN-α. In accordance with the lower levels of IL-12p70 in CD70−/− mice, fewer NK cells in CD70−/− mice produced IFN-γ by intracellular staining (Fig. 5F); however, systemic levels of IFN-γ were relatively similar to nondepleted mice (Fig. 5F). Thus, Treg appear to regulate innate responses during MCMV infection.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Treg are reduced in CD70−/− mice and control the innate response to MCMV. (A) Frequencies of Treg (CD3+CD4+Foxp3+) in spleens of naive WT and CD70−/− mice. (B) WT and CD70−/− mice were infected i.p. with 5 × 104 PFU of MCMV, and Treg frequencies in spleens were determined at various time points p.i. (C–F) Foxp3-DTR mice were injected with DT or PBS to deplete Treg or not and then infected with 5 × 104 PFU of MCMV. Mice were analyzed 36 h p.i. (C) Frequencies of CD3+CD4+Foxp3+ cells in spleens of infected mice. (D) Serum concentrations of IFN-α, TNF-α, and IL-12p70 36 h p.i. (E) Frequencies of NK cells in spleens of infected mice and NK cell expression of CD69. (F) Frequencies of IFN-γ–producing splenic NK cells and serum IFN-γ levels in control and Treg-depleted mice. Data are representative of (A and B) or combined from (C–F) two independent experiments. Statistical significance is indicated by p values. MFI, mean fluorescence intensity.

Treg are functionally impaired in CD70−/− mice

We next asked whether depletion of Treg in CD70−/− mice would have an impact on innate responses to MCMV infection. To test this, we depleted Treg in WT and CD70−/− mice using Ab-mediated depletion. Injection of anti-CD25 Ab eliminates the majority of CD25+ Treg. Anti-CD25 Ab treatment in WT mice resulted in a 50% reduction of CD3+CD4+Foxp3+ cells (Fig. 6A), and, consistent with data obtained using Foxp3-DTR mice, WT mice treated with anti-CD25 Ab had elevated levels of IFN-α and proinflammatory cytokines postinfection with MCMV (Fig. 6B). WT mice depleted of Treg with anti-CD25 Ab also had increased NK cell activation as measured by CD69 and IFN-γ production (Fig. 6C–E). In contrast to Treg depletion in WT mice, cytokine responses and NK cell activation in CD70−/− treated with anti-CD25 Ab were not altered in the absence of Treg (Fig. 6F–J). These data suggested that Treg might already be functionally impaired in CD70−/− mice. To address this, we performed in vitro Treg suppression assays (30). We found that Treg isolated from naive CD70−/− mice were not able to suppress proliferation of CD4+CD25− T cells (Tconv) as efficiently as Treg from naive WT mice (Fig. 6K). Also supporting the idea that Treg from CD70−/− might have a moderate, intrinsic defect in their suppressive capacity, transient blockade of CD70–CD27 interactions in WT mice had no impact on Treg numbers (Fig. 7A) or on cytokine responses and NK cell activation during MCMV infection (Fig. 7B-7C). Taken together, our findings indicate that Treg control innate responses to MCMV infection in WT mice and that reduced numbers and impaired function of Treg in CD70−/− mice contribute to hyperactivation of the innate response during MCMV infection.

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Treg are functionally impaired in CD70−/− mice. (A–J) WT and CD70−/− mice were injected with an isotype control Ab (Iso) or depleted of CD4+CD25+Foxp3+ cells with anti-CD25 Ab and then infected with MCMV. Mice were analyzed 36 h p.i. (A and F) Percentages of Treg in spleens. (B and G) Serum levels of IFN-α, IL-12p70, and TNF-α. (C and H) CD69 expression on NK cells and frequencies of IFN-γ–producing NK cells in spleens. Frequencies of splenic NK cells (D and I) and serum IFN-γ levels (E and J). Data are combined from two independent experiments. Statistical significance is indicated by p values. (K) Treg were isolated from naive WT and CD70−/− mice and cocultured with CFSE-labeled naive CD4+CD25− T cells (Tconv) from WT mice for 3 d. Proliferation was measured by flow cytometry. Numbers indicate percentages of Tconv that divided two times or more. MFI, mean fluorescence intensity.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Transient blockade of CD70–CD27 interactions does not impact innate responses to MCMV. (A–C) WT mice were injected with an isotype control Ab (Iso) or anti-CD70 Ab (FR70) prior to MCMV infection. Mice were analyzed at 36 h p.i. (A) Treg frequencies in spleens of infected mice. (B) Serum levels of IFN-α, IL-12p70, and TNF-α. (C) Frequencies of splenic NK cells, CD69 expression on splenic NK cells, frequencies of splenic IFN-γ–producing NK cells, and serum levels of IFN-γ. Data are combined from two independent experiments. Statistical significance is indicated by p values. MFI, mean fluorescence intensity.

Discussion

Our study shows that CD70 has two major functions in the antiviral immune response. On one hand, CD70 is required for an optimal CD8 T cell response and control of MCMV load. On the other hand, we found that CD70 is essential for regulating the innate inflammatory response during the initial phase of infection. The impairment of the adaptive T cell response was expected because activation of CD8 T cells through CD27 has been shown to provide survival signals that counter TRAIL-induced apoptosis (13–15). However, we found that lack of CD70 also resulted in reduced DC numbers early after MCMV infection, which may contribute to the reduction in the CD8 T cell response. CD70-deficient DC expressed more DR5 than their WT counterparts, which may increase their susceptibility to TRAIL-induced apoptosis. The remarkable finding of this study is that CD70 is required for the control of innate inflammatory response in the initial phase of infection. Accordingly, CD70−/− mice exhibited an early robust cytokine response to MCMV infection. The increased IFN-α response in CD70−/− mice facilitated the control of MCMV in the first 36 h of infection and, together with the burst of IL-12, probably promoted the nonspecific activation of NK cells and the enhanced secretion of IFN-γ. This elevated cytokine response appeared to be a consequence of a defect in Treg numbers and function. We found that CD70−/− mice have a modest reduction of Treg in steady state, as recently reported (20), which was intensified during viral infection, and that Treg from CD70−/− mice were not as efficient at suppressing responses by other cell types. Because Treg inhibit the activation and promote the trafficking of APC, it is likely that impaired survival and function of Treg in CD70−/− mice results in exuberant responsiveness of these cells to inflammatory stimuli and lessens their numbers at sites of infection (32–36). Corroborating this, WT but not CD70−/− mice depleted of CD25+ Treg displayed greater cytokine production postinfection with MCMV. However, transient blockade of CD70–CD27 interactions was not sufficient to cause changes in Treg numbers or the innate response, which is in agreement with a recent study (29). Because CD70 mediates reverse signaling (37) and translocates together with the invariant chain to the endosomal/lysosomal compartments (38), CD70 may also act by modulating TLR signaling and/or translocation of TLR into endosomal compartment where they interact with microbial ligands.

Surprisingly, although a substantial NK cell subset expresses CD27 (21, 39), and previous studies confirmed a role for DC–NK interaction in promoting control of viral infections (40, 41), NK cell effector functions were not reduced in CD70−/− mice. In fact, NK cell activation was transiently increased in CD70−/− mice at early time points after MCMV infection, which may reflect an increase of IFN-α and IL-12 as well as a decrease in Treg, which have been recently shown to modulate NK cell function and homeostasis (42–44). This early NK cell activation was followed by a reduction in NK cell numbers, but this minor change is unlikely to affect the control of MCMV infection, as CD70−/− mice showed defective control of Δm157 MCMV, which evades NK cell surveillance in C57BL/6 mice. Altogether, our results indicate the dual function of CD70 in viral infection as a modulator of early cytokine responses via maintenance of Treg function and survival and activator of adaptive responses through induction of virus-specific CD8 T cells.

Disclosures

The authors have no financial conflicts of interest.

Acknowledgments

We thank C. Hsieh, W. Yokoyama, A. French, and T. Geurs (Washington University) for mice, viruses, and advice; S. Gilfillan for advice on mouse genetics and breeding (Washington University); M. Bugatti for immunohistochemistry (University of Brescia); and the Rheumatic Diseases Core Center at Washington University for speed congenics.

Footnotes

  • A.A. was supported by Training Grant 5-T32CA09547 from the National Cancer Institute. M.S. was supported by Grant K01DK095972 from the National Institute of Diabetes and Digestive and Kidney Diseases. Research reported in this study was also supported by the National Institute of Arthritis and Musculoskeletal and Skin Diseases, part of the National Institutes of Health, under Award P30AR048335.

  • Abbreviations used in this article:

    DT
    diphtheria toxin
    DTR
    diphtheria toxin receptor
    MCMV
    murine CMV
    p.i.
    postinfection
    Tconv
    conventional T cell
    Treg
    regulatory T cell
    WT
    wild-type.

  • Received September 10, 2013.
  • Accepted May 6, 2014.

References

  1. ↵
    1. Croft M.
    2009. The role of TNF superfamily members in T-cell function and diseases. Nat. Rev. Immunol. 9: 271–285.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Watts T. H.
    2005. TNF/TNFR family members in costimulation of T cell responses. Annu. Rev. Immunol. 23: 23–68.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Tesselaar K.,
    2. Y. Xiao,
    3. R. Arens,
    4. G. M. van Schijndel,
    5. D. H. Schuurhuis,
    6. R. E. Mebius,
    7. J. Borst,
    8. R. A. van Lier
    . 2003. Expression of the murine CD27 ligand CD70 in vitro and in vivo. J. Immunol. 170: 33–40.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Laouar A.,
    2. V. Haridas,
    3. D. Vargas,
    4. X. Zhinan,
    5. D. Chaplin,
    6. R. A. van Lier,
    7. N. Manjunath
    . 2005. CD70+ antigen-presenting cells control the proliferation and differentiation of T cells in the intestinal mucosa. Nat. Immunol. 6: 698–706.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Atarashi K.,
    2. J. Nishimura,
    3. T. Shima,
    4. Y. Umesaki,
    5. M. Yamamoto,
    6. M. Onoue,
    7. H. Yagita,
    8. N. Ishii,
    9. R. Evans,
    10. K. Honda,
    11. K. Takeda
    . 2008. ATP drives lamina propria T(H)17 cell differentiation. Nature 455: 808–812.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Borst J.,
    2. J. Hendriks,
    3. Y. Xiao
    . 2005. CD27 and CD70 in T cell and B cell activation. Curr. Opin. Immunol. 17: 275–281.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Bullock T. N.,
    2. H. Yagita
    . 2005. Induction of CD70 on dendritic cells through CD40 or TLR stimulation contributes to the development of CD8+ T cell responses in the absence of CD4+ T cells. J. Immunol. 174: 710–717.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Sanchez P. J.,
    2. J. A. McWilliams,
    3. C. Haluszczak,
    4. H. Yagita,
    5. R. M. Kedl
    . 2007. Combined TLR/CD40 stimulation mediates potent cellular immunity by regulating dendritic cell expression of CD70 in vivo. J. Immunol. 178: 1564–1572.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Hendriks J.,
    2. L. A. Gravestein,
    3. K. Tesselaar,
    4. R. A. van Lier,
    5. T. N. Schumacher,
    6. J. Borst
    . 2000. CD27 is required for generation and long-term maintenance of T cell immunity. Nat. Immunol. 1: 433–440.
    OpenUrlCrossRefPubMed
    1. Hendriks J.,
    2. Y. Xiao,
    3. J. Borst
    . 2003. CD27 promotes survival of activated T cells and complements CD28 in generation and establishment of the effector T cell pool. J. Exp. Med. 198: 1369–1380.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Hendriks J.,
    2. Y. Xiao,
    3. J. W. Rossen,
    4. K. F. van der Sluijs,
    5. K. Sugamura,
    6. N. Ishii,
    7. J. Borst
    . 2005. During viral infection of the respiratory tract, CD27, 4-1BB, and OX40 collectively determine formation of CD8+ memory T cells and their capacity for secondary expansion. J. Immunol. 175: 1665–1676.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Taraban V. Y.,
    2. T. F. Rowley,
    3. D. F. Tough,
    4. A. Al-Shamkhani
    . 2006. Requirement for CD70 in CD4+ Th cell-dependent and innate receptor-mediated CD8+ T cell priming. J. Immunol. 177: 2969–2975.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Feau S.,
    2. Z. Garcia,
    3. R. Arens,
    4. H. Yagita,
    5. J. Borst,
    6. S. P. Schoenberger
    . 2012. The CD4⁺ T-cell help signal is transmitted from APC to CD8⁺ T-cells via CD27-CD70 interactions. Nat. Commun. 3: 948.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Peperzak V.,
    2. E. A. Veraar,
    3. A. M. Keller,
    4. Y. Xiao,
    5. J. Borst
    . 2010. The Pim kinase pathway contributes to survival signaling in primed CD8+ T cells upon CD27 costimulation. J. Immunol. 185: 6670–6678.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Peperzak V.,
    2. Y. Xiao,
    3. E. A. Veraar,
    4. J. Borst
    . 2010. CD27 sustains survival of CTLs in virus-infected nonlymphoid tissue in mice by inducing autocrine IL-2 production. J. Clin. Invest. 120: 168–178.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Soares H.,
    2. H. Waechter,
    3. N. Glaichenhaus,
    4. E. Mougneau,
    5. H. Yagita,
    6. O. Mizenina,
    7. D. Dudziak,
    8. M. C. Nussenzweig,
    9. R. M. Steinman
    . 2007. A subset of dendritic cells induces CD4+ T cells to produce IFN-gamma by an IL-12-independent but CD70-dependent mechanism in vivo. J. Exp. Med. 204: 1095–1106.
    OpenUrlAbstract/FREE Full Text
    1. van Oosterwijk M. F.,
    2. H. Juwana,
    3. R. Arens,
    4. K. Tesselaar,
    5. M. H. van Oers,
    6. E. Eldering,
    7. R. A. van Lier
    . 2007. CD27-CD70 interactions sensitise naive CD4+ T cells for IL-12-induced Th1 cell development. Int. Immunol. 19: 713–718.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Xiao Y.,
    2. V. Peperzak,
    3. A. M. Keller,
    4. J. Borst
    . 2008. CD27 instructs CD4+ T cells to provide help for the memory CD8+ T cell response after protein immunization. J. Immunol. 181: 1071–1082.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Coquet J. M.,
    2. S. Middendorp,
    3. G. van der Horst,
    4. J. Kind,
    5. E. A. Veraar,
    6. Y. Xiao,
    7. H. Jacobs,
    8. J. Borst
    . 2013. The CD27 and CD70 costimulatory pathway inhibits effector function of T helper 17 cells and attenuates associated autoimmunity. Immunity 38: 53–65.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Coquet J. M.,
    2. J. C. Ribot,
    3. N. Bąbała,
    4. S. Middendorp,
    5. G. van der Horst,
    6. Y. Xiao,
    7. J. F. Neves,
    8. D. Fonseca-Pereira,
    9. H. Jacobs,
    10. D. J. Pennington,
    11. et al
    . 2013. Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27-CD70 pathway. J. Exp. Med. 210: 715–728.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Hayakawa Y.,
    2. M. J. Smyth
    . 2006. CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity. J. Immunol. 176: 1517–1524.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Matter M.,
    2. S. Mumprecht,
    3. D. D. Pinschewer,
    4. V. Pavelic,
    5. H. Yagita,
    6. S. Krautwald,
    7. J. Borst,
    8. A. F. Ochsenbein
    . 2005. Virus-induced polyclonal B cell activation improves protective CTL memory via retained CD27 expression on memory CTL. Eur. J. Immunol. 35: 3229–3239.
    OpenUrlCrossRefPubMed
    1. Schildknecht A.,
    2. I. Miescher,
    3. H. Yagita,
    4. M. van den Broek
    . 2007. Priming of CD8+ T cell responses by pathogens typically depends on CD70-mediated interactions with dendritic cells. Eur. J. Immunol. 37: 716–728.
    OpenUrlCrossRefPubMed
    1. Penaloza-MacMaster P.,
    2. A. Ur Rasheed,
    3. S. S. Iyer,
    4. H. Yagita,
    5. B. R. Blazar,
    6. R. Ahmed
    . 2011. Opposing effects of CD70 costimulation during acute and chronic lymphocytic choriomeningitis virus infection of mice. J. Virol. 85: 6168–6174.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Munitic I.,
    2. M. Kuka,
    3. A. Allam,
    4. J. P. Scoville,
    5. J. D. Ashwell
    . 2013. CD70 deficiency impairs effector CD8 T cell generation and viral clearance but is dispensable for the recall response to lymphocytic choriomeningitis virus. J. Immunol. 190: 1169–1179.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Lanier L. L.
    2008. Evolutionary struggles between NK cells and viruses. Nat. Rev. Immunol. 8: 259–268.
    OpenUrlCrossRefPubMed
  23. ↵
    1. García-Sastre A.,
    2. C. A. Biron
    . 2006. Type 1 interferons and the virus-host relationship: a lesson in détente. Science 312: 879–882.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Kim J. M.,
    2. J. P. Rasmussen,
    3. A. Y. Rudensky
    . 2007. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8: 191–197.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Welten S. P.,
    2. A. Redeker,
    3. K. L. Franken,
    4. C. A. Benedict,
    5. H. Yagita,
    6. F. M. Wensveen,
    7. J. Borst,
    8. C. J. Melief,
    9. R. A. van Lier,
    10. K. P. van Gisbergen,
    11. R. Arens
    . 2013. CD27-CD70 costimulation controls T cell immunity during acute and persistent cytomegalovirus infection. J. Virol. 87: 6851–6865.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Collison L. W.,
    2. D. A. Vignali
    . 2011. In vitro Treg suppression assays. Methods Mol. Biol. 707: 21–37.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Krmpotic A.,
    2. I. Bubic,
    3. B. Polic,
    4. P. Lucin,
    5. S. Jonjic
    . 2003. Pathogenesis of murine cytomegalovirus infection. Microbes Infect. 5: 1263–1277.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Serra P.,
    2. A. Amrani,
    3. J. Yamanouchi,
    4. B. Han,
    5. S. Thiessen,
    6. T. Utsugi,
    7. J. Verdaguer,
    8. P. Santamaria
    . 2003. CD40 ligation releases immature dendritic cells from the control of regulatory CD4+CD25+ T cells. Immunity 19: 877–889.
    OpenUrlCrossRefPubMed
    1. Fallarino F.,
    2. U. Grohmann,
    3. K. W. Hwang,
    4. C. Orabona,
    5. C. Vacca,
    6. R. Bianchi,
    7. M. L. Belladonna,
    8. M. C. Fioretti,
    9. M. L. Alegre,
    10. P. Puccetti
    . 2003. Modulation of tryptophan catabolism by regulatory T cells. Nat. Immunol. 4: 1206–1212.
    OpenUrlCrossRefPubMed
    1. Misra N.,
    2. J. Bayry,
    3. S. Lacroix-Desmazes,
    4. M. D. Kazatchkine,
    5. S. V. Kaveri
    . 2004. Cutting edge: human CD4+CD25+ T cells restrain the maturation and antigen-presenting function of dendritic cells. J. Immunol. 172: 4676–4680.
    OpenUrlAbstract/FREE Full Text
    1. Godfrey W. R.,
    2. Y. G. Ge,
    3. D. J. Spoden,
    4. B. L. Levine,
    5. C. H. June,
    6. B. R. Blazar,
    7. S. B. Porter
    . 2004. In vitro-expanded human CD4(+)CD25(+) T-regulatory cells can markedly inhibit allogeneic dendritic cell-stimulated MLR cultures. Blood 104: 453–461.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Lund J. M.,
    2. L. Hsing,
    3. T. T. Pham,
    4. A. Y. Rudensky
    . 2008. Coordination of early protective immunity to viral infection by regulatory T cells. Science 320: 1220–1224.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Arens R.,
    2. M. A. Nolte,
    3. K. Tesselaar,
    4. B. Heemskerk,
    5. K. A. Reedquist,
    6. R. A. van Lier,
    7. M. H. van Oers
    . 2004. Signaling through CD70 regulates B cell activation and IgG production. J. Immunol. 173: 3901–3908.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Zwart W.,
    2. V. Peperzak,
    3. E. de Vries,
    4. A. M. Keller,
    5. G. van der Horst,
    6. E. A. Veraar,
    7. U. Geumann,
    8. H. Janssen,
    9. L. Janssen,
    10. S. H. Naik,
    11. et al
    . 2010. The invariant chain transports TNF family member CD70 to MHC class II compartments in dendritic cells. J. Cell Sci. 123: 3817–3827.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Hayakawa Y.,
    2. N. D. Huntington,
    3. S. L. Nutt,
    4. M. J. Smyth
    . 2006. Functional subsets of mouse natural killer cells. Immunol. Rev. 214: 47–55.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Andoniou C. E.,
    2. S. L. van Dommelen,
    3. V. Voigt,
    4. D. M. Andrews,
    5. G. Brizard,
    6. C. Asselin-Paturel,
    7. T. Delale,
    8. K. J. Stacey,
    9. G. Trinchieri,
    10. M. A. Degli-Esposti
    . 2005. Interaction between conventional dendritic cells and natural killer cells is integral to the activation of effective antiviral immunity. Nat. Immunol. 6: 1011–1019.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Andoniou C. E.,
    2. J. D. Coudert,
    3. M. A. Degli-Esposti
    . 2008. Killers and beyond: NK-cell-mediated control of immune responses. Eur. J. Immunol. 38: 2938–2942.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Gasteiger G.,
    2. S. Hemmers,
    3. P. D. Bos,
    4. J. C. Sun,
    5. A. Y. Rudensky
    . 2013. IL-2-dependent adaptive control of NK cell homeostasis. J. Exp. Med. 210: 1179–1187.
    OpenUrlAbstract/FREE Full Text
    1. Gasteiger G.,
    2. S. Hemmers,
    3. M. A. Firth,
    4. A. Le Floc’h,
    5. M. Huse,
    6. J. C. Sun,
    7. A. Y. Rudensky
    . 2013. IL-2-dependent tuning of NK cell sensitivity for target cells is controlled by regulatory T cells. J. Exp. Med. 210: 1167–1178.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Sitrin J.,
    2. A. Ring,
    3. K. C. Garcia,
    4. C. Benoist,
    5. D. Mathis
    . 2013. Regulatory T cells control NK cells in an insulitic lesion by depriving them of IL-2. J. Exp. Med. 210: 1153–1165.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 193 (2)
The Journal of Immunology
Vol. 193, Issue 2
15 Jul 2014
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • 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.
Dual Function of CD70 in Viral Infection: Modulator of Early Cytokine Responses and Activator of Adaptive Responses
(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
Dual Function of CD70 in Viral Infection: Modulator of Early Cytokine Responses and Activator of Adaptive Responses
Atef Allam, Melissa Swiecki, William Vermi, Jonathan D. Ashwell, Marco Colonna
The Journal of Immunology July 15, 2014, 193 (2) 871-878; DOI: 10.4049/jimmunol.1302429

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Dual Function of CD70 in Viral Infection: Modulator of Early Cytokine Responses and Activator of Adaptive Responses
Atef Allam, Melissa Swiecki, William Vermi, Jonathan D. Ashwell, Marco Colonna
The Journal of Immunology July 15, 2014, 193 (2) 871-878; DOI: 10.4049/jimmunol.1302429
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

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

Related Articles

Cited By...

More in this TOC Section

  • Genetic Deletion of LRP5 and LRP6 in Macrophages Exacerbates Colitis-Associated Systemic Inflammation and Kidney Injury in Response to Intestinal Commensal Microbiota
  • The Intracellular Interaction of Porcine β-Defensin 2 with VASH1 Alleviates Inflammation via Akt Signaling Pathway
  • Fungal Patterns Induce Cytokine Expression through Fluxes of Metabolic Intermediates That Support Glycolysis and Oxidative Phosphorylation
Show more INNATE IMMUNITY AND INFLAMMATION

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 © 2022 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606