Biological Functions of Intracellular Hepatitis B e Antigen

dc.contributor.advisorGuo, Haitao
dc.contributor.authorMitra, Bidisha
dc.contributor.otherAndrophy, Elliot J.
dc.contributor.otherKaplan, Mark
dc.contributor.otherYu, Andy
dc.contributor.otherLu, Tao
dc.date.accessioned2019-10-10T14:08:36Z
dc.date.available2019-10-10T14:08:36Z
dc.date.issued2019-09
dc.degree.date2019en_US
dc.degree.discipline
dc.degree.grantorIndiana Universityen_US
dc.degree.levelPh.D.en_US
dc.descriptionIndiana University-Purdue University Indianapolis (IUPUI)en_US
dc.description.abstractThe function(s) of the intracellular form of HBeAg, previously reported as the preCore protein intermediate (p22) without the N-terminal signal peptide, remains elusive. Here, we propose to elucidate the translocation of p22 during its formation from endoplasmic reticulum (ER) to cytosol, how it differs from core in its inability to form a capsid and the biological functions of cytoplasmic and nuclear p22. Firstly, we have identified that a portion of p22, after the cleavage of its signal peptide in ER, is released back into the cytosol through an ERAD-independent mechanism, as neither wildtype nor dominant-negative p97 affected the ER-to-cytosol translocation of p22 or ER-Golgi secretion of HBeAg. Secondly, despite sharing the same sequence with core protein except for the extended 10 amino acid precore region at the N-terminus, we observed that p22 wildtype and C-7Q mutant are unable to form a capsid. Thirdly, we report that p22 but not the secreted HBeAg significantly reduced interferon stimulated response element (ISRE) activity and expression of interferon stimulated genes (ISGs) upon interferon-alpha (IFN- α) stimulation. Furthermore, in line with this, RNA-seq analysis of ISG induction profile from IFN-α treated patients showed that HBeAg(+) patients exhibited reduced and weak antiviral ISG upregulations compared to HBeAg(-) patients. Further, mechanistic study indicated that while p22 did not alter the total STAT1 or p-STAT1 levels in IFN-α treated cells, it blocked the nuclear translocation of p-STAT1 by interacting with karyopherin α1, indicating that the cytoplasmic p22 may impede JAK-STAT signaling to help the virus evade host innate immune response and cause resistance to IFN therapy in patients. Additionally, nuclear p22 and nuclear core were found to interact with the promoter regions (ISRE – containing) of ISGs, suggesting a new mechanism of inhibition of ISG expression upon stimulation. Finally, we found that the nuclear p22 can bind to cccDNA minichromosome and affects cccDNA maintenance and/or transcription. Thus, our results indicate that there is a novel ER sorting mechanism for the distribution of the intracellular and secretory HBeAg, and the intracellular HBeAg may contribute to HBV persistence by interfering with IFN-α elicited JAK-STAT signaling and regulating cccDNA metabolism.en_US
dc.identifier.urihttps://hdl.handle.net/1805/21091
dc.identifier.urihttp://dx.doi.org/10.7912/C2/1760
dc.language.isoen_USen_US
dc.subjectcccDNAen_US
dc.subjectcoreen_US
dc.subjectHBVen_US
dc.subjectIFN signalingen_US
dc.subjectp22en_US
dc.subjectprecoreen_US
dc.titleBiological Functions of Intracellular Hepatitis B e Antigenen_US
dc.typeDissertation
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
Mitra_iupui_0104D_10395.pdf
Size:
3.77 MB
Format:
Adobe Portable Document Format
Description:
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: