Abstract

Apoptosis of virus-infected cells is an effective antiviral mechanism in addition to interferon induction to establish antiviral state to restrict virus spread. The interferon-inducible 2′–5′ oligoadenylate synthetase/RNase L pathway results in activation of RNase L in response to double stranded RNA and cleaves diverse RNA substrates to amplify interferon induction and promote apoptosis. Here we show that RNase L induces expression of Death-associated protein kinase-Related Apoptosis-inducing protein Kinase 1 (DRAK1), a member of the death-associated protein kinase family and interferon-signaling pathway is required for induction. Overexpression of DRAK1 triggers apoptosis in the absence of RNase L activation by activating c-Jun N-terminal kinase (JNK), translocation of BCL2 Associated X (Bax) to the mitochondria accompanied by cytochrome C release and loss of mitochondrial membrane potential promoting cleavage of caspase 3 and Poly(ADP-Ribose) Polymerase 1 (PARP). Inhibitors of JNK and caspase 3 promote survival of DRAK1 overexpressing cells demonstrating an important role of JNK signaling pathway in DRAK1-mediated apoptosis. DRAK1 mutant proteins that lack kinase activity or nuclear localization fail to induce apoptosis highlighting the importance of cellular localization and kinase function in promoting cell death. Our studies identify DRAK1 as a mediator of RNase L-induced apoptosis.

Highlights

  • Apoptosis is a regulated process of cell death that plays a critical role in many physiological events, including the cellular response to virus infection

  • Microarray analysis of DU145 cells transfected with 2-5A to activate Ribonuclease L (RNase L) identified changes in mRNA levels of antiviral and antitumor genes including death-associated protein kinase (DAPK)-related apoptosis-inducing protein kinase 1 (DRAK1), a member of the death-associated protein (DAP) family of serine/threonine protein kinases

  • To determine if death-associated protein kinase-related apoptosis-inducing protein kinase 1 (DRAK1) is induced in prostate cancer cells in response to RNase L activation, PC3 and DU145 cells were transfected with –5 -oligoadenylate oligomers (2–5A) or synthetic dsRNA, Poly I:C, which activates oligoadenylate synthetase to produce 2–5A

Read more

Summary

Introduction

Apoptosis is a regulated process of cell death that plays a critical role in many physiological events, including the cellular response to virus infection. Type I IFN released by infected cells binds to interferon receptor (IFNAR) and triggers signaling pathways to induce expression of interferon-stimulated genes (ISGs) which are essential for the establishment of an antiviral state [2]. The 2 –5 -oligoadenylate synthetase (OAS)-RNase L pathway is an antiviral response that is induced by IFN and dsRNA produced during viral infections. Binding of RNase L inactive monomers to 2–5A, causes conformational change and dimerization to produce an active nuclease that cleaves single-stranded viral and cellular RNAs after UpUp or UpAp residues, producing small dsRNA products with roles in transcriptional activation of antiviral and antitumor genes, amplifying IFN production, activating inflammasomes and promoting switch from autophagy to apoptosis [12,13,14,15,16]. Among the genes that were transcriptionally induced by RNase L, death-associated protein kinase-related apoptosis-inducing protein kinase 1 (DRAK1) was a unique member of the DAP-kinase family, and its role in cell death by IFN signaling was unknown [12]

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.