Major racial and gender differences have been recognized in the natural history of hepatitis C virus (HCV) infection; however, the underlying mechanism for this phenomenon remain poorly understood. In studying the natural history of HCV infection, it has been well-recognized that i) white females have an 8-fold greater chance of spontaneous viral clearance of acute HCV infection; ii) African American (AA) males with genotype 1 HCV infection have higher rates of viral persistence or chronic infection than Caucasian Americans (CA); and iii) these differences are not explained by the baseline viral loads, genotypes, or disease characteristics, but related to the host immune status, in particular T cell responses. In studying the effect of viral infection on T cell functions, we and others have recently found that chronic HCV infection leads to T cell dysfunction mediated through up- regulation of aging markers, including killer cell lectin-like receptor subfamily G member 1 (KLRG1) and dual specific phosphatase 6 (DUSP6), concomitant with a decline of microRNA-181a (miR181) levels in CD4 T cells. These observations suggest that the inability of host to clear virus during chronic infection may be a result of microRNA-mediated impairment of host immunity, and that race/gender influences on the natural history of HCV infection may be due to a difference in virus-induced, miRNA-mediated signaling. Indeed, with increasing age, KLRG1 is up-regulated and leads to inhibition of T cell receptor (TCR) signaling, whereas DUSP6 is increased and leads to recalibration of the TCR activation threshold; miR181 declines to permit translation of a set of genes related to T cell inhibition. However, the mechanisms underlying miR181/KLRG1/DUSP6 expression and regulation of premature T cell aging during HCV infection and their relationships to the gender difference in the natural history of HCV infection remain unclear. We thus hypothesize that virus-induced microRNAs may exhibit a gender difference, which may affect T cell responses that contribute to the natural history of HCV infection. To test this hypothesis, we will carry out the following specific aims: 1) Does HCV induce a different level of miR181, resulting in different T cell responses from male and female HCV-infected or HCV- resolved patients? 2) Are there any differences in miR181 expression in CD4 and CD8 T cells from male and female healthy subjects in response to stimulation by TCR antibodies (anti-CD3/CD28) or HCV antigens? 3) What is the role of HCV-mediated miR181 on host immune responses by gain- or loss-of-function assays, including virus-specific T cell responses? The overall goal of this supplemental proposal is to employ a translational approach to obtain a unified overview on whether HCV may induce different levels of miR181 in T cells from male and female subjects, which may translate into different levels of host T cell responses, and thus contribute to different outcomes of HCV infection.

Public Health Relevance

HCV is characterized by clear race- and gender-related differences in the natural history and outcome of infection. Why this virus is able to persist in the majority of infected individuals, particularly in males versus females, remains unclear, but HCV-induced disruption of host immune responses appears to play a major role. Based on our preliminary studies, we suspect that gender-related difference in microRNA-mediated T cell regulation may explain the altered natural history and outcome of HCV infection. This project will examine whether there is a gender difference in microRNA (miR181 and/or miR155) induction by HCV, and will characterize its role in virus-specific T cell responses. This study will elucidate the gender-related variation of virus-induced microRNAs in the outcome of HCV infection and facilitate effective approaches to improve viral clearance, and thus is significant and timely.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
3R01AI114748-05S1
Application #
9896225
Study Section
Program Officer
Koshy, Rajen
Project Start
2019-06-17
Project End
2020-11-30
Budget Start
2019-06-17
Budget End
2020-11-30
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
East Tennessee State University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
051125037
City
Johnson City
State
TN
Country
United States
Zip Code
37614
Wang, Ling; Howell, Mary E A; McPeak, Brooke et al. (2018) LIMD1 is induced by and required for LMP1 signaling, and protects EBV-transformed cells from DNA damage-induced cell death. Oncotarget 9:6282-6297
Wang, L; Ren, J; Li, G et al. (2017) LMP1 signaling pathway activates IRF4 in latent EBV infection and a positive circuit between PI3K and Src is required. Oncogene 36:2265-2274
Wang, Ling; Wang, Yujia; Zhao, Juan et al. (2017) The Linear Ubiquitin Assembly Complex Modulates Latent Membrane Protein 1 Activation of NF-?B and Interferon Regulatory Factor 7. J Virol 91:
Ren, Jun P; Wang, Lin; Zhao, Juan et al. (2017) Decline of miR-124 in myeloid cells promotes regulatory T-cell development in hepatitis C virus infection. Immunology 150:213-220
Wang, Ling; Zhao, Juan; Ren, Junping et al. (2016) Protein phosphatase 1 abrogates IRF7-mediated type I IFN response in antiviral immunity. Eur J Immunol 46:2409-2419
Wang, Ling; Zhao, Juan; Ren, Jun P et al. (2016) Expansion of myeloid-derived suppressor cells promotes differentiation of regulatory T cells in HIV-1+ individuals. AIDS 30:1521-1531
Ren, Jun P; Zhao, Juan; Dai, Jun et al. (2016) Hepatitis C virus-induced myeloid-derived suppressor cells regulate T-cell differentiation and function via the signal transducer and activator of transcription 3 pathway. Immunology 148:377-86
Ren, J P; Ying, R S; Cheng, Y Q et al. (2016) HCV-induced miR146a controls SOCS1/STAT3 and cytokine expression in monocytes to promote regulatory T-cell development. J Viral Hepat 23:755-66
Zhou, Yun; Li, Guang Y; Ren, Jun P et al. (2016) Protection of CD4+ T cells from hepatitis C virus infection-associated senescence via ?Np63-miR-181a-Sirt1 pathway. J Leukoc Biol 100:1201-1211
Li, Guang Y; Zhou, Yun; Ying, Ruo S et al. (2015) Hepatitis C virus-induced reduction in miR-181a impairs CD4(+) T-cell responses through overexpression of DUSP6. Hepatology 61:1163-73

Showing the most recent 10 out of 13 publications