Chronic viral infections afflict more than 500 million people worldwide. Although altered stimulatory and inhibitory pathways contribute to T cell suppression during persistent viral infections in mice and humans, the underlying mechanisms are not fully understood. It should be noted that human persistent viruses, (including human immunodeficiency virus, hepatitis B and C viruses) are restricted to human and nonhuman primates and that the complexity of the immune system cannot be accurately recreated in vitro. Thus, the study of immune-regulation during chronic viral infections requires the use of appropriate whole organism viral systems. By using in vivo chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, we and others have significantly contributed to understanding the roles and dynamics of several immune-regulators during chronic viral infections. In particular, we have shown that transforming growth factor beta (TGF?) production and signaling are sustained during persistent compared to acute LCMV infection. More recently, we have used temporally and cell-specifically controlled mouse genetics and observed that TGF? signaling in activated CD4 T cells is essential for host survival early after chronic (but not acut) LCMV infection and promotes viral replication at later stages. We also found that TGF? suppresses proliferation, terminal differentiation and (unexpectedly) a cytotoxic program in CD4 T cells early after infection while inhibiting CD8 T cell functions and accumulation at later stages. Importantly, initial data suggest that TGF? suppression of virus-specific cytotoxic CD4 T cells extends to HIV-infected patients. Understanding the cellular and molecular factors underlying the complex but critical role of TGF? and its crosstalk with contextual cytokines during an in vivo persistent viral infection is essential for manipulating this pathway to boost ani-viral defenses without causing immunopathology. To this end, in Aim #1 we propose to fully discern the temporal role of TGF? in modulating CD8 and CD4 T cell responses, immunopathology and viral control early versus late after chronic LCMV infection. We will also evaluate the role of TGF?3 and the TGF? activating molecule, integrin-?8, in immune responses and viral persistence.
In Aim #2 we will determine the mechanisms underlying TGF? regulation of CD4 T cell responses by ablating SMAD-4 at different times after chronic LCMV infection and identifying SMAD-4 targeted gene promoters. We will also evaluate the role of transcription factors (TFs) that are putative SMAD-4 co-regulators and TFs that we found to be regulated by TGF? in CD4 T cells. Last, in Aim #3 we will examine whether interleukin-12 (IL-12) mediates the phenotypes induced by TGF? ablation during chronic LCMV infection, given that TGF? deficient CD4 T cells show enhanced IL-12 responsiveness. For that, we will block IL-12 in TGF?R sufficient and deficient mice and evaluate the consequences for T cell responses, immunopathology and viral control after persistent LCMV infection. These experiments will establish the foundation for future approaches aiming at enforcing IL-12 signaling to overcome TGF? suppression during chronic viral infection. In conclusion, our findings support the idea that TGF? plays vital roles during a chronic viral infection in vivo, influencing T cell responses, hos survival and viral control. While this suggests promising novel opportunities for immunotherapies, further understanding the dynamic activity of TGF? and the underlying molecular factors is necessary to effectively and safely manipulate this pathway and alleviate chronic viral diseases. Furthermore, given that LCMV is not only itself a pathogen of medical importance but it is also a prototypic member of the family arenaviridae, our work will help explaining how pathogenic arenaviruses suppress T cell responses to cause lethalhemorrhagic fevers in humans. Last, but not least, because TGF? is produced during several infections and T cells are central for anti-viral immunity, our work could have broad-reaching implications for multiple infectious diseases.

Public Health Relevance

Viruses that cause chronic infections, including Human Immunodeficiency virus (HIV), Hepatitis B virus (HBV) and Hepatitis C virus (HCV), infect more than 500 million people worldwide and represent a major health problem. Suppression of immune responses is a pre-requisite for a virus to persist. We have identified an inhibitory cytokine that counteract T cell responses and influences host survival and viral control during murine chronic viral infection. This proposal aims at further studying this cytokine operating mode and downstream signaling with the ultimate goal of rationally design alternative strategies to alleviate chronic viral diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI081923-10
Application #
9627894
Study Section
Virology - B Study Section (VIRB)
Program Officer
Jiang, Chao
Project Start
2009-07-01
Project End
2021-01-31
Budget Start
2019-02-01
Budget End
2021-01-31
Support Year
10
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of California, San Diego
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
804355790
City
La Jolla
State
CA
Country
United States
Zip Code
92093
Harker, James A; Wong, Kurt A; Dallari, Simone et al. (2018) Interleukin-27R Signaling Mediates Early Viral Containment and Impacts Innate and Adaptive Immunity after Chronic Lymphocytic Choriomeningitis Virus Infection. J Virol 92:
Wehrens, Ellen J; Wong, Kurt A; Gupta, Ankan et al. (2018) IL-27 regulates the number, function and cytotoxic program of antiviral CD4 T cells and promotes cytomegalovirus persistence. PLoS One 13:e0201249
Loureiro, MarĂ­a Eugenia; Zorzetto-Fernandes, Andre Luiz; Radoshitzky, Sheli et al. (2018) DDX3 suppresses type I interferons and favors viral replication during Arenavirus infection. PLoS Pathog 14:e1007125
Macal, Monica; Jo, Yeara; Dallari, Simone et al. (2018) Self-Renewal and Toll-like Receptor Signaling Sustain Exhausted Plasmacytoid Dendritic Cells during Chronic Viral Infection. Immunity 48:730-744.e5
Kakaradov, Boyko; Arsenio, Janilyn; Widjaja, Christella E et al. (2017) Early transcriptional and epigenetic regulation of CD8+ T cell differentiation revealed by single-cell RNA sequencing. Nat Immunol 18:422-432
Smelkinson, Margery G; Guichard, Annabel; Teijaro, John R et al. (2017) Influenza NS1 directly modulates Hedgehog signaling during infection. PLoS Pathog 13:e1006588
Macal, Monica; Tam, Miguel A; Hesser, Charles et al. (2016) CD28 Deficiency Enhances Type I IFN Production by Murine Plasmacytoid Dendritic Cells. J Immunol 196:1900-9
Lewis, Gavin M; Wehrens, Ellen J; Labarta-Bajo, Lara et al. (2016) TGF-? receptor maintains CD4 T helper cell identity during chronic viral infections. J Clin Invest 126:3799-3813
El Ghazal, Roland; Yin, Xin; Johns, Scott C et al. (2016) Glycan Sulfation Modulates Dendritic Cell Biology and Tumor Growth. Neoplasia 18:294-306
Taylor, Alison; Harker, James A; Chanthong, Kittiphat et al. (2016) Glycogen Synthase Kinase 3 Inactivation Drives T-bet-Mediated Downregulation of Co-receptor PD-1 to Enhance CD8(+) Cytolytic T Cell Responses. Immunity 44:274-86

Showing the most recent 10 out of 22 publications