Abstract

Kaposi's sarcoma-associated herpesvirus (KSHV) has recently been found to generate circular RNAs (circRNAs) from several KSHV genes, most abundantly from K10 (viral interferon regulatory factor 4 [vIRF4]), K7.3, and polyadenylated nuclear (PAN) RNA. To define expression of these circRNAs, KSHV-infected cell lines, patient tissues, and purified virions were examined. KSHV circRNA expression was universally detected in tests of six primary effusion lymphoma (PEL) cell lines but ranged from low-level expression in BC-1 cells dually infected with tightly latent KSHV and Epstein-Barr virus to abundant expression in KSHV-only BCBL-1 cells with spontaneous virus production. Generally, the PAN/K7.3 locus broadly and bidirectionally generated circRNA levels that paralleled the corresponding linear RNA levels. However, RNA corresponding to a particular KSHV circularization site (circ-vIRF4) was minimally induced, despite linear vIRF4 RNA being activated by virus induction. In situ hybridization showed abundant circ-vIRF4 in noninduced PEL cells. All three KSHV circRNAs were isolated as nuclease-protected forms from gradient-purified virions collected from BrK.219 cells infected with a KSHV molecular clone. For circ-vIRF4, the fully processed form that is exported to the cytoplasm was incorporated into virus particles but the nuclear, intron-retaining form was not. The half-life of circ-vIRF4 was twice as long as that of its linear counterpart. The KSHV circRNAs could be detected at a higher rate than their corresponding linear counterparts by in situ hybridization in archival tissues and by reverse transcription-PCR (RT-PCR) in sera stored for over 25 years. In summary, KSHV circRNAs are expressed in infection-associated diseases, can be regulated depending on virus life cycle, and are incorporated into viral particles for preformed delivery, suggesting a potential function in early infection.IMPORTANCE KSHV has recently been found to encode circRNAs. circRNAs result from back-splicing of an upstream pre-mRNA splice donor exon-intron junction to an acceptor site, generating a covalently closed circle. This study revealed that for one KSHV region, the PAN/K7.3 locus, broadly and bidirectionally generated circRNA levels parallel corresponding linear RNA levels. Another KSHV circularization site (circ-vIRF4), however, showed expression that differed from that of the corresponding linear RNA. All KSHV circRNAs are incorporated into KSHV virions and are potentially expressed as immediate early products in newly infected cells.

Highlights

  • Kaposi’s sarcoma-associated herpesvirus (KSHV) has recently been found to generate circular RNAs from several KSHV genes, most abundantly from K10, K7.3, and polyadenylated nuclear (PAN) RNA

  • Expression profiles of KSHV-encoded circRNAs were assessed in primary effusion lymphoma (PEL) tumorderived cell lines infected with KSHV alone (BCP1, BC3, and BCBL1) or dually infected with KSHV and Epstein-Barr virus (EBV) (BC1, JSC1, and BBG1), as well as in BJABrKSHV.219, a BJAB cell line stably infected with a recombinant virus

  • We designed one divergent primer pair each for circPAN and circK7.3 that can detect the majority of the circRNA species

Read more

Summary

Introduction

Kaposi’s sarcoma-associated herpesvirus (KSHV) has recently been found to generate circular RNAs (circRNAs) from several KSHV genes, most abundantly from K10 (viral interferon regulatory factor 4 [vIRF4]), K7.3, and polyadenylated nuclear (PAN) RNA. This study revealed that for one KSHV region, the PAN/K7.3 locus, broadly and bidirectionally generated circRNA levels parallel corresponding linear RNA levels Another KSHV circularization site (circ-vIRF4), showed expression that differed from that of the corresponding linear RNA. In addition to Ͼ90 KSHV protein-coding mRNAs, most of which are predominantly expressed during full-virus lytic replication, noncoding viral RNAs (ncRNAs) are functionally important features of the KSHV transcriptome. These KSHV ncRNAs include 12 precursor microRNAs (pre-miRNAs) that give rise to a total of 25 mature miRNAs [12,13,14]. Notable among these are the antisense-to-latency transcript (ALT), which is transcribed antisense to the major viral latency locus; T3.0 and T1.2, which are oriented opposite to replication and transcription activator (RTA/ORF50); and K7.3, which runs antisense to PAN [12, 22, 23, 27, 30]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call