Advertisement

INTERFER(ON)ing Persistence

During persistent viral infections, a dysregulated immune response fails to control the infection. Wilson et al. (p. 202) and Teijaro et al. (p. 207; see the Perspective by Odorizzi and Wherry) show this occurs because type I interferons (IFN I), critical for early responses to viral infection, contribute to the altered immunity seen during persistent infection. Antibody blockade of IFN I signaling during chronic lymphocytic choriomeningitis virus (LCMV) in mice resulted in reduced viral titers at later stages of infection, reduced expression of inhibitory immune molecules and prevented the disruptions to secondary lymphoid organs typically observed during persistent infection with LCMV. Whether type I IFNs are also detrimental to persistent viral infection humans, such as HIV and hepatitis C virus, remains to be determined.

Abstract

During persistent viral infections, chronic immune activation, negative immune regulator expression, an elevated interferon signature, and lymphoid tissue destruction correlate with disease progression. We demonstrated that blockade of type I interferon (IFN-I) signaling using an IFN-I receptor neutralizing antibody reduced immune system activation, decreased expression of negative immune regulatory molecules, and restored lymphoid architecture in mice persistently infected with lymphocytic choriomeningitis virus. IFN-I blockade before and after establishment of persistent virus infection resulted in enhanced virus clearance and was CD4 T cell–dependent. Hence, we demonstrate a direct causal link between IFN-I signaling, immune activation, negative immune regulator expression, lymphoid tissue disorganization, and virus persistence. Our results suggest that therapies targeting IFN-I may help control persistent virus infections.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Figs. S1 to S6
Reference (37)

Resources

File (teijaro.sm.pdf)

References and Notes

1
Brooks D. G., et al., Interleukin-10 determines viral clearance or persistence in vivo. Nat. Med. 12, 1301 (2006).
2
Barber D. L., et al., Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682 (2006).
3
Yi J. S., Cox M. A., Zajac A. J., T-cell exhaustion: characteristics, causes and conversion. Immunology 129, 474 (2010).
4
Zajac A. J., et al., Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205 (1998).
5
Appay V., Sauce D., Immune activation and inflammation in HIV-1 infection: causes and consequences. J. Pathol. 214, 231 (2008).
6
Chang J. J., Altfeld M., Innate immune activation in primary HIV-1 infection. J. Infect. Dis. 202 (suppl. 2), S297 (2010).
7
d’Ettorre G., Paiardini M., Ceccarelli G., Silvestri G., Vullo V., HIV-associated immune activation: from bench to bedside. AIDS Res. Hum. Retroviruses 27, 355 (2011).
8
Sevilla N., et al., Immunosuppression and resultant viral persistence by specific viral targeting of dendritic cells. J. Exp. Med. 192, 1249 (2000).
9
Ahmed R., Salmi A., Butler L. D., Chiller J. M., Oldstone M. B., 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 (1984).
10
Salvato M., Borrow P., Shimomaye E., Oldstone M. B., Molecular basis of viral persistence: a single amino acid change in the glycoprotein of lymphocytic choriomeningitis virus is associated with suppression of the antiviral cytotoxic T-lymphocyte response and establishment of persistence. J. Virol. 65, 1863 (1991).
11
Evans C. F., Borrow P., de la Torre J. C., Oldstone M. B., Virus-induced immunosuppression: kinetic analysis of the selection of a mutation associated with viral persistence. J. Virol. 68, 7367 (1994).
12
Sullivan B. M., et al., Point mutation in the glycoprotein of lymphocytic choriomeningitis virus is necessary for receptor binding, dendritic cell infection, and long-term persistence. Proc. Natl. Acad. Sci. U.S.A. 108, 2969 (2011).
13
Bergthaler A., et al., Viral replicative capacity is the primary determinant of lymphocytic choriomeningitis virus persistence and immunosuppression. Proc. Natl. Acad. Sci. U.S.A. 107, 21641 (2010).
14
Scheu S., Dresing P., Locksley R. M., Visualization of IFNbeta production by plasmacytoid versus conventional dendritic cells under specific stimulation conditions in vivo. Proc. Natl. Acad. Sci. U.S.A. 105, 20416 (2008).
15
Macal M., et al., Plasmacytoid dendritic cells are productively infected and activated through TLR-7 early after arenavirus infection. Cell Host Microbe 11, 617 (2012).
16
Sheehan K. C., et al., Blocking monoclonal antibodies specific for mouse IFN-alpha/beta receptor subunit 1 (IFNAR-1) from mice immunized by in vivo hydrodynamic transfection. J. Interferon Cytokine Res. 26, 804 (2006).
17
Sevilla N., McGavern D. B., Teng C., Kunz S., Oldstone M. B., Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion. J. Clin. Invest. 113, 737 (2004).
18
Oldstone M. B., Campbell K. P., Decoding arenavirus pathogenesis: essential roles for alpha-dystroglycan-virus interactions and the immune response. Virology 411, 170 (2011).
19
Müller S., et al., Role of an intact splenic microarchitecture in early lymphocytic choriomeningitis virus production. J. Virol. 76, 2375 (2002).
20
Berger D. P., et al., Lymphotoxin-beta-deficient mice show defective antiviral immunity. Virology 260, 136 (1999).
21
Suresh M., et al., Role of lymphotoxin alpha in T-cell responses during an acute viral infection. J. Virol. 76, 3943 (2002).
22
Zeng M., et al., Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J. Clin. Invest. 121, 998 (2011).
23
Zeng M., et al., Lymphoid tissue damage in HIV-1 infection depletes naïve T cells and limits T cell reconstitution after antiretroviral therapy. PLoS Pathog. 8, e1002437 (2012).
24
Hahm B., Trifilo M. J., Zuniga E. I., Oldstone M. B., Viruses evade the immune system through type I interferon-mediated STAT2-dependent, but STAT1-independent, signaling. Immunity 22, 247 (2005).
25
Stetson D. B., Medzhitov R., Type I interferons in host defense. Immunity 25, 373 (2006).
26
Platanias L. C., Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 5, 375 (2005).
27
Isaacs A., Lindenmann J., Virus interference. I. The interferon. Proc. R. Soc. Lond. B Biol. Sci. 147, 258 (1957).
28
Schoggins J. W., Rice C. M., Interferon-stimulated genes and their antiviral effector functions. Curr. Opin. Virol. 1, 519 (2011).
29
Schoggins J. W., et al., A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 472, 481 (2011).
30
Manches O., Bhardwaj N., Resolution of immune activation defines nonpathogenic SIV infection. J. Clin. Invest. 119, 3512 (2009).
31
Jacquelin B., et al., Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. J. Clin. Invest. 119, 3544 (2009).
32
Bosinger S. E., et al., Global genomic analysis reveals rapid control of a robust innate response in SIV-infected sooty mangabeys. J. Clin. Invest. 119, 3556 (2009).
33
Deeks S. G., Walker B. D., Human immunodeficiency virus controllers: mechanisms of durable virus control in the absence of antiretroviral therapy. Immunity 27, 406 (2007).
34
Sáez-Cirión A., Pancino G., Sinet M., Venet A., Lambotte O., HIV controllers: how do they tame the virus? Trends Immunol. 28, 532 (2007).
35
Su A. I., et al., Genomic analysis of the host response to hepatitis C virus infection. Proc. Natl. Acad. Sci. U.S.A. 99, 15669 (2002).
36
Guidotti L. G., Chisari F. V., Immunobiology and pathogenesis of viral hepatitis. Annu. Rev. Pathol. 1, 23 (2006).
37
Borrow P., Evans C. F., Oldstone M. B., Virus-induced immunosuppression: immune system-mediated destruction of virus-infected dendritic cells results in generalized immune suppression. J. Virol. 69, 1059 (1995).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 340 | Issue 6129
12 April 2013

Submission history

Received: 15 January 2013
Accepted: 28 February 2013
Published in print: 12 April 2013

Permissions

Request permissions for this article.

Acknowledgments

The authors thank D. Fremgen, C. Cubitt, N. Ngo, and S. Rice for technical excellence. Data reported in the manuscript are tabulated in the main paper and in the supplementary materials. This research was supported by NIH grant AI09484 (M.B.A.O.); National Cancer Institute grants NCI CA43059 (R.D.S.) and U54AI057160 to the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research (MRCE) (R.D.S. and M.B.A.O.); grants AI077719 and AI047140 (J.C.d.l.T.), postdoctoral training grants AI007354; and American Heart fellowships 11POST7430106 (J.R.T.), HL007195 (C.N.), and NS041219 (B.M.S.).

Authors

Affiliations

John R. Teijaro*
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Cherie Ng*
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Andrew M. Lee
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Present address: Center for Genetics of Host Defense, Immunology, University of Texas Southwestern, Dallas, TX 75390–8505, USA.
Brian M. Sullivan
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Kathleen C. F. Sheehan
Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
Megan Welch
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Robert D. Schreiber
Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
Juan Carlos de la Torre
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
Michael B. A. Oldstone [email protected]
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.

Notes

*
These authors contributed equally to this work.
Corresponding author. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Emergence and fate of stem cell–like Tcf7+ CD8+ T cells during a primary immune response to viral infection, Science Immunology, 8, 89, (2023)./doi/10.1126/sciimmunol.adh3113
    Abstract
  2. Cellular and molecular waypoints along the path of T cell exhaustion, Science Immunology, 8, 87, (2023)./doi/10.1126/sciimmunol.adg3868
    Abstract
  3. Caspase-8 has dual roles in regulatory T cell homeostasis balancing immunity to infection and collateral inflammatory damage, Science Immunology, 7, 69, (2022)./doi/10.1126/sciimmunol.abn8041
    Abstract
  4. Metabolic regulation by PD-1 signaling promotes long-lived quiescent CD8 T cell memory in mice, Science Translational Medicine, 13, 615, (2021)./doi/10.1126/scitranslmed.aba6006
    Abstract
  5. HCV-Induced Immune Responses Influence the Development of Operational Tolerance After Liver Transplantation in Humans, Science Translational Medicine, 6, 242, (242ra81-242ra81), (2021)./doi/10.1126/scitranslmed.3008793
    Abstract
  6. Multifaceted Activities of Type I Interferon Are Revealed by a Receptor Antagonist, Science Signaling, 7, 327, (ra50-ra50), (2021)./doi/10.1126/scisignal.2004998
    Abstract
  7. In vivo genome-wide CRISPR screens identify SOCS1 as intrinsic checkpoint of CD4+ TH1 cell response, Science Immunology, 6, 66, (2021)./doi/10.1126/sciimmunol.abe8219
    Abstract
  8. Severely ill patients with COVID-19 display impaired exhaustion features in SARS-CoV-2–reactive CD8+ T cells, Science Immunology, 6, 55, (2021)./doi/10.1126/sciimmunol.abe4782
    Abstract
  9. The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness, Science Immunology, 1, 6, (2021)./doi/10.1126/sciimmunol.aai8593
    Abstract
  10. Interferon-driven deletion of antiviral B cells at the onset of chronic infection, Science Immunology, 1, 4, (2021)./doi/10.1126/sciimmunol.aah6817
    Abstract
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media