scholarly journals Barrier-to-Autointegration Factor 1 Protects against a Basal cGAS-STING Response

mBio ◽  
2020 ◽  
Vol 11 (2) ◽  
Author(s):  
Hongming Ma ◽  
Wei Qian ◽  
Monika Bambouskova ◽  
Patrick L. Collins ◽  
Sofia I. Porter ◽  
...  

ABSTRACT Although the pathogen recognition receptor pathways that activate cell-intrinsic antiviral responses are well delineated, less is known about how the host regulates this response to prevent sustained signaling and possible immune-mediated damage. Using a genome-wide CRISPR-Cas9 screening approach to identify host factors that modulate interferon-stimulated gene (ISG) expression, we identified the DNA binding protein Barrier-to-autointegration factor 1 (Banf1), a previously described inhibitor of retrovirus integration, as a modulator of basal cell-intrinsic immunity. Ablation of Banf1 by gene editing resulted in chromatin activation near host defense genes with associated increased expression of ISGs, including Oas2, Rsad2 (viperin), Ifit1, and ISG15. The phenotype in Banf1-deficient cells occurred through a cGAS-, STING-, and IRF3-dependent signaling axis, was associated with reduced infection of RNA and DNA viruses, and was reversed in Banf1 complemented cells. Confocal microscopy and biochemical studies revealed that a loss of Banf1 expression resulted in higher level of cytosolic double-stranded DNA at baseline. Our study identifies an undescribed role for Banf1 in regulating the levels of cytoplasmic DNA and cGAS-dependent ISG homeostasis and suggests possible therapeutic directions for promoting or inhibiting cell-intrinsic innate immune responses. IMPORTANCE Although the interferon (IFN) signaling pathway is a key host mechanism to restrict infection of a diverse range of viral pathogens, its unrestrained activity either at baseline or in the context of an immune response can result in host cell damage and injury. Here, we used a genome-wide CRISPR-Cas9 screen and identified the DNA binding protein Barrier-to-autointegration factor 1 (Banf1) as a modulator of basal cell-intrinsic immunity. A loss of Banf1 expression resulted in higher level of cytosolic double-stranded DNA at baseline, which triggered IFN-stimulated gene expression via a cGAS-STING-IRF3 axis that did not require type I IFN or STAT1 signaling. Our experiments define a regulatory network in which Banf1 limits basal inflammation by preventing self DNA accumulation in the cytosol.

2005 ◽  
Vol 5 (8) ◽  
pp. 1216-1219 ◽  
Author(s):  
Yunfei Bai ◽  
Qinyu Ge ◽  
Quanjun Liu ◽  
Tongxiang Li ◽  
Jinke Wang ◽  
...  

1992 ◽  
Vol 225 (4) ◽  
pp. 999-1011 ◽  
Author(s):  
Maarten H. Stuiver ◽  
Wilma G. Bergsma ◽  
Annika C. Arnberg ◽  
Herbert van Amerongen ◽  
Rienk van Grondelle ◽  
...  

2001 ◽  
Vol 57 (12) ◽  
pp. 1893-1894 ◽  
Author(s):  
Ben N. Wardleworth ◽  
Rupert J. M. Russell ◽  
Malcolm F. White ◽  
Garry L. Taylor

2005 ◽  
Vol 79 (14) ◽  
pp. 9356-9358 ◽  
Author(s):  
Nina Bacher Reuven ◽  
Sandra K. Weller

ABSTRACT UL12 is a 5′- to 3′-exonuclease encoded by herpes simplex virus type 1 (HSV-1) which degrades single- and double-stranded DNA. UL12 and the single-strand DNA binding protein ICP8 mediate a strand exchange reaction. We found that ICP8 inhibited UL12 digestion of single-stranded DNA but stimulated digestion of double-stranded DNA threefold. The stimulatory effect of ICP8 was independent of a strand exchange reaction; furthermore, the effect was specific to ICP8, as it could not be reproduced by Escherichia coli single-stranded DNA binding protein. The effect of ICP8 on the rate of UL12 double-stranded DNA digestion is attributable to an increase in processivity in the presence of ICP8.


Author(s):  
Keith Wilson ◽  
Isao Tanaka ◽  
Krzysztof Appelt ◽  
Stephen White

1998 ◽  
Vol 17 (2) ◽  
pp. 169-175 ◽  
Author(s):  
PEILIN ZHANG ◽  
JEAN-LOUIS VIGNE ◽  
SYNTHIA H. MELLON

Sign in / Sign up

Export Citation Format

Share Document