Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2012 Jul;86(13):7060-71.
doi: 10.1128/JVI.00164-12. Epub 2012 May 2.

Therapeutic blockade of transforming growth factor beta fails to promote clearance of a persistent viral infection

Affiliations

Therapeutic blockade of transforming growth factor beta fails to promote clearance of a persistent viral infection

Lucile Garidou et al. J Virol. 2012 Jul.

Abstract

Persistent viral infections often overburden the immune system and are a major cause of disease in humans. During many persistent infections, antiviral T cells are maintained in a state of immune exhaustion characterized by diminished effector and helper functions. In mammalian systems, an extensive immune regulatory network exists to limit unwanted, potentially fatal immunopathology by inducing T cell exhaustion. However, this regulatory network at times overprotects the host and fosters viral persistence by severely dampening adaptive immune responsiveness. Importantly, recent studies have shown that T cell exhaustion is mediated in part by host immunoregulatory pathways (e.g., programmed death 1 [PD-1], interleukin 10 [IL-10]) and that therapeutic blockade of these pathways either before or during persistent infection can promote viral clearance. Transforming growth factor beta (TGF-β) is another immunosuppressive cytokine known to impede both self- and tumor-specific T cells, but its role in regulating antiviral immunity is not entirely understood. In this study, we inhibited TGF-β with three potent antagonists to determine whether neutralization of this regulatory molecule is a viable approach to control a persistent viral infection. Our results revealed that these inhibitors modestly elevate the number of antiviral T cells following infection with a persistent variant of lymphocytic choriomeningitis virus (LCMV) but have no impact on viral clearance. These data suggest that therapeutic neutralization of TGF-β is not an efficacious means to promote clearance of a persistent viral infection.

PubMed Disclaimer

Figures

Fig 1
Fig 1
dnTGF-β RII mice rapidly control a persistent LCMV infection. (A) TGF-β1, -2, and -3 expression was quantified by qPCR using splenic RNA extracted from naïve, day 7 Arm-infected, and day 7 CL13-infected mice (n = 4 mice per group). All TGF isoforms were significantly upregulated in LCMV-infected mice. Asterisks denote a statistically significant increase from the naïve control group as determined by one-way ANOVA (P < 0.05). (B) The geometric mean fluorescent intensities of pSmad2/3 (less the isotype control signal) was quantified in splenic DbGP33–41-specific Thy1.1+ CD8+ T cells (P14) and I-AbGP61–80 CD45.1+ CD4+ T cells (SMARTA) 7 days following infection with Arm or CL13 (n = 5 mice per group). No statistically significant differences between the groups were observed. (C, D) Frequencies of Db-GP33–41 tetramer+ CD8+ T cells (B) and I-Ab-GP61–80 tetramer+ CD4+ T cells (C) in the blood were determined in CL13-infected dnTGF-β RII+ and dnTGF-β RII mice at the denoted time points postinfection (n = 7 mice per group). Tetramer+ T cells were significantly increased (asterisks, P < 0.05) in dnTGF-β RII+ mice relative to in wild-type control mice. (E) At selected time points postinfection, serum viral titers were measured by plaque assay in CL13-infected dnTGF-β RII+ and dnTGF-β RII mice. dnTGF-β RII+ mice cleared virus significantly faster (asterisks, P < 0.05) than wild-type controls. All data in this figure are plotted as means ± standard deviations (SD) and are representative of at least two independent experiments.
Fig 2
Fig 2
LCMV CL13-infected dnTGF-β RII mice mount an increased antiviral T cell response. (A, B) The frequency (A) and absolute number (B) of MHC-I (DbGP33–41 and Db-NP396–404) and MHC-II (I-AbGP61–80) tetramer+ T cells were calculated in the spleen 10 days following CL13 infection of dnTGF-β RII+ and dnTGF-β RII mice (n = 5 mice per group). Representative dot plots are gated on CD8+ T cells for MHC-I tetramers and CD4+ T cells for MHC-II tetramers. Boxes denote the mean frequency of tetramer-positive cells in each group. Absolute numbers are plotted as means ± SD. (C) On day 10 postinfection, virus-specific CD8+ and CD4+ T cell responses were assessed by ex vivo peptide stimulation using splenocytes from wild-type and dnTGF-β RII+ mice (n = 5 mice per group). The absolute number of IFN-γ- and TNF-α-producing CD8+ (stimulated with GP33–41 or NP396–404) and CD4+ (stimulated with GP61–80) T cells is plotted as mean ± SD. (D) Geometric mean fluorescent intensities (GMFI) for IFN-γ and TNF-α production from panel C are plotted (n = 5 mice per group). Asterisks on all plots and graphs denote a statistically significant increase (P < 0.05) in dnTGF-β RII+ mice relative to wild-type controls. Data are representative of at least two independent experiments.
Fig 3
Fig 3
Therapeutic blockade of TGF-β increases antiviral T cells but does not impact viral clearance. To block TGF-β signaling, mice were injected with IN1233 (25 mg/kg/day), anti-TGF-β antibody (1 mg every other day), or TGF-β RII-Fc (50 μg every other day) (see Materials and Methods). (A, B) Graphs illustrate the frequency (A) and absolute number (B) of Db-GP33–41 tetramer+ CD8+ T cells and I-Ab-GP61–80 tetramer+ CD4+ T cells in the spleen of treated mice 10 days following infection (mean ± SD) (n = 5 mice per group). (C) The functionality of LCMV-specific CD4+ and CD8+ T cells in the spleen was evaluated by ex vivo peptide stimulation with GP61–80 and NP396–404, respectively. The graph shows the absolute number (mean ± SD) of IFN-γ- and TNF-α-producing T cells that respond to peptide (n = 5 mice per group). (D) Nine days postinfection, serum viral titers were determined by plaque assay (n = 5 mice per group). Data are expressed as PFU/ml of serum sampled (mean ± SD). All data are representative of at least two independent experiments. Asterisks denote a statistically significant increase (P < 0.05) relative to the PBS control group.
Fig 4
Fig 4
Administration of anti-TGF-β antibodies does not promote clearance of a persistent viral infection. To determine the efficiency of TGF-β neutralization, serum concentrations of the cytokine were quantified by ELISA in day 8 CL13-infected mice treated with anti-TGF-β or isotype control antibodies (1.5 mg every other day). Naïve untreated mice served as a control (n = 5 mice per group). Anti-TGF-β antibodies reduced the cytokine to an undetectable level (mean ± SD = 0 ± 0) in all CL13-infected mice. (B, C) MHC-I (DbGP33–41) and MHC-II (I-AbGP61–80) tetramers were used to calculate frequencies (A) and absolute numbers (B) of virus-specific T cells in the spleens of isotype versus anti-TGF-β antibody (1.5 mg every other day)-treated mice at day 9 postinfection. Representative dot plots (A) are gated on CD8+ T cells for MHC-I tetramers and CD4+ T cells for MHC-II tetramers. Boxes denote the mean frequency of tetramer-positive cells in each group. The absolute number of splenic tetramer-positive T cells was calculated from the frequency data shown in panel A (n = 5 mice per group). (D) On day 10 postinfection, virus-specific CD8+ and CD4+ T cell responses were assessed by ex vivo peptide stimulation of splenocytes from both isotype and anti-TGF-β antibody-treated mice. Boxes denote the absolute number (mean ± SD) of cytokine-producing cells for each group (n = 5 mice per group). (E) Average geometric mean fluorescent intensities (GMFI) from panel C were graphed for all cytokines. (F) At selected time points, serum viral titers were determined by plaque assay and plotted as means ± SD (n = 5 mice per group). Asterisks on all graphs denote a statistically significance increase (P < 0.05) relative to the control group, and data are representative of at least two independent experiments.
Fig 5
Fig 5
Adoptively transferred dnTGF-β RII memory T cells do not accelerate clearance of a persistent viral infection. A total of 104 wild-type or dnTGF-β RII Thy1.1+ CD8+ P14 cells was transferred into naïve C57BL/6J mice and then infected intravenously with LCMV CL13 (n = 5 mice per group). (A) Representative dot plots gated on CD8+ T cells show the mean frequency of Thy1.1+ CD8+ T cells (black boxes) in the blood of mice at day 8 and day 13 following LCMV infection. (B) The frequencies of Thy1.1+ CD8+ T cells in the blood are plotted over time (mean ± SD). Note that dnTGF-β RII Thy1.1+ CD8+ P14 cells are rejected from CL13-infected mice by day 13 postinfection. Asterisks denote a statistically significant difference between the groups (P < 0.05). (C) At 45 days following i.p. infection with LCMV Arm, memory splenocytes were extracted from wild-type and dnTGF-β RII mice and then adoptively transferred i.p. into wild-type and dnTGF-β RII LCMV carrier mice, respectively (n = 4 mice per group). The correction factor for dnTGF-β RII splenocytes was calculated by staining pooled memory cells with anti-CD8 antibody and a DbGP33–41 tetramer prior to injection. A correction factor was introduced into each experiment to ensure that comparable numbers of wild-type and dnTGF-β RII memory CD8+ T cells were transferred into LCMV carrier mice (see Materials & Methods). MHC-I (DbGP33–41) and MHC-II (I-AbGP61–80) tetramers were used to calculate the absolute number of transferred virus-specific T cells in the spleens of wild-type and dnTGF-β RII carrier mice at day 11 posttransfer. Data are plotted as means ± SD. (D) At selected time points following adoptive immunotherapy, serum viral titers in recipient carrier mice were determined by plaque assay (n = 4 mice per group). Data are expressed as PFU/ml of serum sampled (mean ± SD) and are representative of three independent experiments.
Fig 6
Fig 6
dnTGF-β RII mice contain a large repertoire of activated T cells prior to infection. (A) Heterozygous dnTGF-β RII mice were crossed with Thy1.1+ P14 mice. Splenic Thy1.1+ CD8+ T cells in the resultant F1 mice were screened at 8 weeks for expression of CD44 and CD122. Histograms and dot plots are gated on Thy1.1+ CD8+ P14 T cells. Note that CD44 and CD122 expressions are elevated on CD8+ T cells from dnTGF-β RII+ mice relative to those of wild-type littermate controls (n = 3 mice per group). Boxes denote the mean frequency of CD122+ Thy1.1+ CD8+ T cells in each group. (B) Naïve 8-week-old Thy1.2+ P14 mice were injected every other day i.p. for 3 weeks with 1 mg of anti-TGF-β or isotype control antibodies. CD44 and CD122 expression was then examined on splenic Thy1.2+ CD8+ T cells as described in panel A. No significant differences were observed between the groups (n = 3 mice per group). (C, D) dnTGF-β RII+ or dnTGF-β RII mice were seeded with 104 naïve Thy1.1+ CD8+ P14 cells and then infected i.v. with LCMV CL13. The frequency of Thy1.1+CD8+ P14 cells was determined in the blood at day 7 postinfection, and the absolute number of cells was calculated in the spleen at day 10 postinfection (n = 5 mice per group). Asterisks denote a statistically significance increase (P < 0.05) in dnTGF-β RII+ recipients relative to wild-type controls. All data are representative of at least two independent experiments.

Similar articles

Cited by

References

    1. Ahmed R, Salmi A, Butler LD, Chiller JM, Oldstone MB. 1984. Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160:521–540 - PMC - PubMed
    1. Ahmed R, et al. 1988. Genetic analysis of in vivo-selected viral variants causing chronic infection: importance of mutation in the L RNA segment of lymphocytic choriomeningitis virus. J. Virol. 62:3301–3308 - PMC - PubMed
    1. Barber DL, et al. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439:682–687 - PubMed
    1. Bellavance EC, et al. 2011. Development of tumor-infiltrating CD8+ T cell memory precursor effector cells and antimelanoma memory responses are the result of vaccination and TGF-beta blockade during the perioperative period of tumor resection. J. Immunol. 186:3309–3316 - PMC - PubMed
    1. Berger DP, Homann D, Oldstone MB. 2000. Defining parameters for successful immunocytotherapy of persistent viral infection. Virology 266:257–263 - PubMed

Publication types

MeSH terms

LinkOut - more resources