bzlf1 promoter
Recently Published Documents


TOTAL DOCUMENTS

21
(FIVE YEARS 0)

H-INDEX

16
(FIVE YEARS 0)

2018 ◽  
Vol 14 (7) ◽  
pp. e1007179 ◽  
Author(s):  
Jillian A. Bristol ◽  
Reza Djavadian ◽  
Emily R. Albright ◽  
Carrie B. Coleman ◽  
Makoto Ohashi ◽  
...  

Virology ◽  
2017 ◽  
Vol 507 ◽  
pp. 220-230 ◽  
Author(s):  
Lena N. Lupey-Green ◽  
Stephanie A. Moquin ◽  
Kayla A. Martin ◽  
Shane M. McDevitt ◽  
Michael Hulse ◽  
...  

2012 ◽  
Vol 86 (9) ◽  
pp. 4752-4761 ◽  
Author(s):  
T. Murata ◽  
Y. Kondo ◽  
A. Sugimoto ◽  
D. Kawashima ◽  
S. Saito ◽  
...  

2011 ◽  
Vol 85 (10) ◽  
pp. 5081-5090 ◽  
Author(s):  
X. Yu ◽  
P. J. McCarthy ◽  
H.-J. Lim ◽  
T. Iempridee ◽  
R. J. Kraus ◽  
...  

2007 ◽  
Vol 81 (24) ◽  
pp. 13566-13577 ◽  
Author(s):  
Chia Chi Sun ◽  
David A. Thorley-Lawson

ABSTRACT Epstein-Barr virus (EBV) in vivo is known to establish persistent infection in resting, circulating memory B cells and to productively replicate in plasma cells. Until now, the molecular mechanism of how EBV switches from latency to lytic replication in vivo was not known. Here, we report that the plasma cell differentiation factor, XBP-1s, activates the expression of the master regulator of EBV lytic activation, BZLF1. Using reporter assays, we observed that XBP-1s was able to transactivate the BZLF1 promoter, Zp, in a plasma cell line and other lymphoid cell lines but, interestingly, not in epithelial cell lines. We have identified an XBP-1s binding site on the ZID/ZII region of Zp, which when abolished by site-directed mutagenesis led to abrogation of XBP-1s binding and promoter activation. Using the chromatin immunoprecipitation assay, we observed direct binding of XBP-1s to endogenous Zp in an EBV-infected plasma cell line. Finally, in the same cell line, we observed that overexpression of XBP-1s resulted in increased expression of BZLF1, while knockdown of XBP-1s with short hairpin RNA drastically reduces BZLF1 expression. We suggest that EBV harnesses the B-cell terminal differentiation pathway via XBP-1s as a physiological signal to reactivate and begin viral replication. We are currently investigating other signals, such as the endoplasmic reticulum stress response proteins, which act upstream of XBP-1s, to identify other interacting factors that initiate and/or amplify the lytic switch.


2003 ◽  
Vol 95 (13) ◽  
pp. 1009-1009
Author(s):  
M. I. Gutierrez ◽  
S. E. Straus ◽  
M. M. Ibrahim ◽  
J. K. Dale ◽  
K. Bhatia

2003 ◽  
Vol 95 (13) ◽  
pp. 1008-1009 ◽  
Author(s):  
J. H. M. Tong ◽  
K. W. Lo ◽  
F. W. L. Au ◽  
D. P. Huang ◽  
K.-F. To

2003 ◽  
Vol 84 (4) ◽  
pp. 959-964 ◽  
Author(s):  
Cornelia Thomas ◽  
Arnd Dankesreiter ◽  
Hans Wolf ◽  
Fritz Schwarzmann

2003 ◽  
Vol 77 (1) ◽  
pp. 199-207 ◽  
Author(s):  
Richard J. Kraus ◽  
Jacqueline G. Perrigoue ◽  
Janet E. Mertz

ABSTRACT Epstein-Barr virus (EBV) is a human herpesvirus capable of establishing a latent state in B lymphocytes. The product of the immediate-early BZLF1 gene, Zta, is a transcriptional transactivator essential for viral DNA amplification and virion production. Previously, we identified a negative cis-acting element within the BZLF1 promoter termed ZV. ZV contains the sequence 5′-CAGGTA-3′ located at nucleotides −17 to −12 relative to the transcription initiation site. It sequence specifically binds a cellular factor, ZVR. Based on sequence binding specificity, we postulated that ZVR may be zinc finger E-box binding factor (ZEB) or a related zinc finger/homeodomain family member. We show here by immunoshift assays that ZVR and human ZEB specifically cross-react with an antibody to δEF1, the chicken homolog of ZEB. Competition electrophoretic mobility shift assays confirmed that ZEB binds to the ZV element with the same binding specificity as ZVR. Overexpression of ZEB in either B-lymphocytic DG75 cells or mammary epithelial MCF-7 cells repressed Zta-induced activation of the BZLF1 promoter four- to fivefold via the ZV site. Thus, we conclude that the previously identified cellular repressor ZVR is, in fact, ZEB. We also present evidence that other cellular factors likely affect the transcriptional activity of ZEB. Lastly, we identify a ZEB-binding site within the promoter of the lytic BRLF1 gene of EBV. We postulate that ZEB likely plays an important role in regulating the life cycle of EBV.


Sign in / Sign up

Export Citation Format

Share Document