Abstract

Autophagy-related proteins such as Beclin-1 are involved in an array of complex processes, including antiviral responses, and may also modulate the efficiency of gene therapy viral vectors. The Tat-Beclin-1 (TB1) peptide has been reported as an autophagy-inducing factor inhibiting the replication of pathogens such as HIV, type 1 (HIV-1). However, autophagy-related proteins are also essential for the early steps of HIV-1 infection. Therefore, we examined the effects of the Beclin-1 evolutionarily conserved domain in TB1 on viral transduction and autophagy in single-round HIV infection or with nonreplicative HIV-1-derived lentiviral vectors. TB1 enhanced transduction with various pseudotypes but without inducing the autophagy process. TB1 augmented the transduction of human CD34+ hematopoietic stem/progenitor cells while maintaining their capacity to engraft in vivo into humanized mice. TB1 was as effective as other transduction additives and functioned by enhancing the adhesion and fusion of viral particles with target cells but not their aggregation. We also found that the N-terminal L1 loop was critical for TB1 transduction-enhancing activity. Interestingly, the Tat-Beclin-2 (TB2) peptide, derived from the human Beclin-2 protein, was even more potent than TB1 in promoting viral transduction and infection. Taken together, our findings suggest that the TB1 and TB2 peptides enhance the viral entry step. Tat-Beclin peptides therefore represent a new family of viral transduction enhancers for potential use in gene therapy.

Highlights

  • The lysosomal degradation pathway of autophagy has a dual role in HIV, type 1 (HIV-1)3 replication and pathogenesis (1)

  • We evaluated the effect of TB1 on single-round infections with HIV-1 and on lentiviral vectors (LVs) pseudotyped with various envelope glycoproteins; namely, the Chikungunya virus glycoprotein (CHIKV-G), the modified gibbon ape leukemia virus glycoprotein (GALVTR), the modified RD114 feline endogenous retrovirus (RD114TR), and the vesicular stomatitis virus glycoprotein (VSV-G), the latter being broadly used in gene therapy

  • To evaluate the effect of the TB1 peptide on LV transduction, we used the human colon carcinoma cell line HCT116, which is routinely employed in our laboratory, to titer LV pseudotyped with the VSV-G envelope (VSV-G-LVs)

Read more

Summary

To whom correspondence may be addressed

Studies have shown that some autophagy-related proteins are essential for the early steps of HIV-1 infection (2– 4), but on the other hand, autophagy induction appears to be an antiviral strategy. Because the early steps of HIV-1 infection are highly dependent on various autophagy-related (Atg) proteins (3), the effect of TB1 on viral replication could be complex and may not be predicted for single-round infections or target cell transduction with non-replicative HIV-1– derived lentiviral vectors (LVs), currently used in various applications of gene therapy (11). We evaluated the effect of TB1 on single-round infections with HIV-1 and on LVs pseudotyped with various envelope glycoproteins; namely, the Chikungunya virus glycoprotein (CHIKV-G), the modified gibbon ape leukemia virus glycoprotein (GALVTR), the modified RD114 feline endogenous retrovirus (RD114TR), and the vesicular stomatitis virus glycoprotein (VSV-G), the latter being broadly used in gene therapy. We observed that the TB1 peptide was able to strongly promote viral infection or transduction with all LV pseudotypes tested, either on cell lines or hCD34ϩ hematopoietic stem/progenitor cells (HSPCs). This study was extended through the design of various TB1 variants and a new peptide called TatBeclin-2 (TB2), a fusion of the Tat (47–57) transduction peptide with the human Beclin-2 ECD249–266 (12)

Results
Discussion
Experimental procedures
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