학술논문

Transcriptional Modulation of Human Endogenous Retroviruses in Primary CD4+ T Cells Following Vorinostat Treatment
Document Type
article
Source
Subject
Medical Microbiology
Biomedical and Clinical Sciences
Immunology
HIV/AIDS
Human Genome
Genetics
6.1 Pharmaceuticals
Aetiology
Evaluation of treatments and therapeutic interventions
2.1 Biological and endogenous factors
Infection
Good Health and Well Being
Antirheumatic Agents
CD4-Positive T-Lymphocytes
Cells
Cultured
Endogenous Retroviruses
Gene Expression Regulation
HIV Infections
HIV-1
Histone Deacetylase Inhibitors
Humans
Immunity
Proviruses
Terminal Repeat Sequences
Virus Activation
Virus Latency
Vorinostat
human endogenous retroviruses
histone deacetylase inhibitor
primary CD4(+) T cells
total RNA-Seq
long terminal repeat
primary CD4+ T cells
Biochemistry and cell biology
Language
Abstract
The greatest obstacle to a cure for HIV is the provirus that integrates into the genome of the infected cell and persists despite antiretroviral therapy. A "shock and kill" approach has been proposed as a strategy for an HIV cure whereby drugs and compounds referred to as latency-reversing agents (LRAs) are used to "shock" the silent provirus into active replication to permit "killing" by virus-induced pathology or immune recognition. The LRA most utilized to date in clinical trials has been the histone deacetylase (HDAC) inhibitor-vorinostat. Potentially, pathological off-target effects of vorinostat may result from the activation of human endogenous retroviruses (HERVs), which share common ancestry with exogenous retroviruses including HIV. To explore the effects of HDAC inhibition on HERV transcription, an unbiased pharmacogenomics approach (total RNA-Seq) was used to evaluate HERV expression following the exposure of primary CD4+ T cells to a high dose of vorinostat. Over 2,000 individual HERV elements were found to be significantly modulated by vorinostat, whereby elements belonging to the ERVL family (e.g., LTR16C and LTR33) were predominantly downregulated, in contrast to LTR12 elements of the HERV-9 family, which exhibited the greatest signal, with the upregulation of 140 distinct elements. The modulation of three different LTR12 elements by vorinostat was confirmed by droplet digital PCR along a dose-response curve. The monitoring of LTR12 expression during clinical trials with vorinostat may be indicated to assess the impact of this HERV on the human genome and host immunity.