scholarly journals CD11c Controls Herpes Simplex Virus 1 Responses To Limit Virus Replication during Primary Infection

2011 ◽  
Vol 85 (19) ◽  
pp. 9945-9955 ◽  
Author(s):  
S. J. Allen ◽  
K. R. Mott ◽  
A. A. Chentoufi ◽  
L. BenMohamed ◽  
S. L. Wechsler ◽  
...  
2021 ◽  
Vol 17 (9) ◽  
pp. e1009950
Author(s):  
Nikhil Sharma ◽  
Chenyao Wang ◽  
Patricia Kessler ◽  
Ganes C. Sen

STING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replication. Here, we report that HSV1 evades this antiviral immune response by inducing a cellular microRNA, miR-24, which binds to the 3’ untranslated region of STING mRNA and inhibits its translation. Expression of the gene encoding miR-24 is induced by the transcription factor AP1 and activated by MAP kinases in HSV1-infected cells. Introduction of exogenous miR-24 or prior activation of MAPKs, causes further enhancement of HSV1 replication in STING-expressing cells. Conversely, transfection of antimiR-24 inhibits virus replication in those cells. HSV1 infection of mice causes neuropathy and death; using two routes of infection, we demonstrated that intracranial injection of antimiR-24 alleviates both morbidity and mortality of the infected mice. Our studies reveal a new immune evasion strategy adopted by HSV1 through the regulation of STING and demonstrates that it can be exploited to enhance STING’s antiviral action.


Microbiology ◽  
2000 ◽  
Vol 81 (7) ◽  
pp. 1763-1771 ◽  
Author(s):  
Tatsuo Suzutani ◽  
Masayoshi Nagamine ◽  
Taiichiro Shibaki ◽  
Masahiro Ogasawara ◽  
Itsuro Yoshida ◽  
...  

The UL41 gene product (vhs) of herpes simplex virus (HSV) is packaged in the virion, and mediates host protein synthesis shutoff at the early stage of the virus replication cycle. In order to clarify the role of vhs in virus replication and virulence, we isolated a completely UL41-deficient mutant (the VRΔ41 strain) and its revertant (the VRΔ41R strain). In the mouse encephalitis model, the replication of strain VRΔ41 was inhibited after 2 days post-infection, resulting in low virulence, by γ-ray-sensitive cells such as lymphocytes and/or neutrophils. The result suggested that some cytokines, produced in VRΔ41-inoculated brains, activate and induce the migration of γ-ray-sensitive cells to the infection site. Therefore, cytokines produced by HSV-1-infected human cells were screened, and potent inductions of interleukin (IL)-1β, IL-8 and macrophage inflammatory protein-1α by VRΔ41 infection were observed. Moreover, the VRΔ41 strain showed 20- and 5-fold higher sensitivity to interferon-α and -β compared to the wild-type strain, respectively. These results indicate that one important role of vhs in vivo is evasion from non-specific host defence mechanisms during primary infection through suppression of cytokine production in HSV-infected cells and reduction of the anti-HSV activity of interferon-α and -β.


2018 ◽  
Vol 4 (4) ◽  
pp. 36 ◽  
Author(s):  
Maja Cokarić Brdovčak ◽  
Andreja Zubković ◽  
Igor Jurak

Viruses utilize microRNAs (miRNAs) in a vast variety of possible interactions and mechanisms, apparently far beyond the classical understanding of gene repression in humans. Likewise, herpes simplex virus 1 (HSV-1) expresses numerous miRNAs and deregulates the expression of host miRNAs. Several HSV-1 miRNAs are abundantly expressed in latency, some of which are encoded antisense to transcripts of important productive infection genes, indicating their roles in repressing the productive cycle and/or in maintenance/reactivation from latency. In addition, HSV-1 also exploits host miRNAs to advance its replication or repress its genes to facilitate latency. Here, we discuss what is known about the functional interplay between HSV-1 and the host miRNA machinery, potential targets, and the molecular mechanisms leading to an efficient virus replication and spread.


2019 ◽  
Vol 34 (4) ◽  
pp. 386-396 ◽  
Author(s):  
Rongquan Huang ◽  
Xusha Zhou ◽  
Shuqi Ren ◽  
Xianjie Liu ◽  
Zhiyuan Han ◽  
...  

2017 ◽  
Vol 91 (15) ◽  
Author(s):  
Dhong Hyun Lee ◽  
Homayon Ghiasi

ABSTRACT Macrophages are the predominant infiltrate in the corneas of mice that have been ocularly infected with herpes simplex virus 1 (HSV-1). However, very little is known about the relative roles of M1 (classically activated or polarized) and M2 (alternatively activated or polarized) macrophages in ocular HSV-1 infection. To better understand these relationships, we assessed the impact of directed M1 or M2 activation of RAW264.7 macrophages and peritoneal macrophages (PM) on subsequent HSV-1 infection. In both the RAW264.7 macrophage and PM in vitro models, HSV-1 replication in M1 macrophages was markedly lower than in M2 macrophages and unstimulated controls. The M1 macrophages expressed significantly higher levels of 28 of the 32 tested cytokines and chemokines than M2 macrophages, with HSV-1 infection significantly increasing the levels of proinflammatory cytokines and chemokines in the M1 versus the M2 macrophages. To examine the effects of shifting the immune response toward either M1 or M2 macrophages in vivo, wild-type mice were injected with gamma interferon (IFN-γ) DNA or colony-stimulating factor 1 (CSF-1) DNA prior to ocular infection with HSV-1. Virus replication in the eye, latency in trigeminal ganglia (TG), and markers of T cell exhaustion in the TG were determined. We found that injection of mice with IFN-γ DNA, which enhances the development of M1 macrophages, increased virus replication in the eye; increased latency; and also increased CD4, CD8, IFN-γ, and PD-1 transcripts in the TG of latently infected mice. Conversely, injection of mice with CSF-1 DNA, which enhances the development of M2 macrophages, was associated with reduced virus replication in the eye and reduced latency and reduced the levels of CD4, CD8, IFN-γ,and PD-1 transcripts in the TG. Collectively, these results suggest that M2 macrophages directly reduce the levels of HSV-1 latency and, thus, T-cell exhaustion in the TG of ocularly infected mice. IMPORTANCE Our findings demonstrate a novel approach to further reducing HSV-1 replication in the eye and latency in the TG by modulating immune components, specifically, by altering the phenotype of macrophages. We suggest that inclusion of CSF-1 as part of any vaccination regimen against HSV infection to coax responses of macrophages toward an M2, rather than an M1, response may further improve vaccine efficacy against ocular HSV-1 replication and latency.


2011 ◽  
Vol 85 (14) ◽  
pp. 7203-7215 ◽  
Author(s):  
A. C. Haugo ◽  
M. L. Szpara ◽  
L. Parsons ◽  
L. W. Enquist ◽  
R. J. Roller

Sign in / Sign up

Export Citation Format

Share Document