Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 9: 603, 2018.
Article in English | MEDLINE | ID: mdl-29706951

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.


Subject(s)
Antirheumatic Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Endogenous Retroviruses/physiology , HIV Infections/drug therapy , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Vorinostat/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Humans , Immunity/drug effects , Proviruses/genetics , Terminal Repeat Sequences/genetics , Virus Activation/drug effects , Virus Latency/drug effects , Vorinostat/therapeutic use
2.
Viruses ; 9(6)2017 05 31.
Article in English | MEDLINE | ID: mdl-28561791

ABSTRACT

Transposable elements, including endogenous retroviruses (ERVs), comprise almost 45% of the human genome. This could represent a significant pathogenic burden but it is becoming more evident that many of these elements have a positive contribution to make to normal human physiology. In particular, the contributions of human ERVs (HERVs) to gene regulation and the expression of noncoding RNAs has been revealed with the help of new and emerging genomic technologies. HERVs have the common provirus structure of coding open reading frames (ORFs) flanked by two long-terminal repeats (LTRs). However, over the course of evolution and as a consequence of host defence mechanisms, most of the sequences contain INDELs, mutations or have been reduced to single LTRs by recombination. These INDELs and mutations reduce HERV activity. However, there is a trade-off for the host cells in that HERVs can provide beneficial sources of genetic variation but with this benefit comes the risk of pathogenic activity and spread within the genome. For example, the LTRs are of critical importance as they contain promoter sequences and can regulate not only HERV expression but that of human genes. This is true even when the LTRs are located in intergenic regions or are in antisense orientation to the rest of the gene. Uncontrolled, this promoter activity could disrupt normal gene expression or transcript processing (e.g., splicing). Thus, control of HERVs and particularly their LTRs is essential for the cell to manage these elements and this control is achieved at multiple levels, including epigenetic regulations that permit HERV expression in the germline but silence it in most somatic tissues. We will discuss some of the common epigenetic mechanisms and how they affect HERV expression, providing detailed discussions of HERVs in stem cell, placenta and cancer biology.


Subject(s)
Endogenous Retroviruses/genetics , Epigenesis, Genetic , Gene Expression Regulation, Viral , Terminal Repeat Sequences , Humans
3.
J Gen Virol ; 96(Pt 6): 1207-1218, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26068187

ABSTRACT

The human genome comprises 8 % endogenous retroviruses (ERVs), the majority of which are defective due to deleterious mutations. Nonetheless, transcripts of ERVs are found in most tissues, and these transcripts could either be reverse transcribed to generate ssDNA or expressed to generate proteins. Thus, the expression of ERVs could produce nucleic acids or proteins with viral signatures, much like the pathogen-associated molecular patterns of exogenous viruses, which would enable them to be detected by the innate immune system. The activation of some pattern recognition receptors (PRRs) in response to ERVs has been described in mice and in the context of human autoimmune diseases. Here, we review the evidence for detection of ERVs by PRRs and the resultant activation of innate immune signalling. This is an emerging area of research within the field of innate antiviral immunity, showing how ERVs could initiate immune signalling pathways and might have implications for numerous inflammatory diseases.


Subject(s)
Endogenous Retroviruses/immunology , Immunity, Innate , Animals , Gene Expression , Humans , Mice , RNA, Viral/immunology , RNA, Viral/metabolism , Receptors, Pattern Recognition/metabolism , Transcription, Genetic , Viral Proteins/immunology , Viral Proteins/metabolism
5.
J Biol Chem ; 288(47): 33642-33653, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24114841

ABSTRACT

Vaccinia virus encodes a number of proteins that inhibit and manipulate innate immune signaling pathways that also have a role in virulence. These include A52, a protein shown to inhibit IL-1- and Toll-like receptor-stimulated NFκB activation, via interaction with interleukin-1 receptor-associated kinase 2 (IRAK2). Interestingly, A52 was also found to activate p38 MAPK and thus enhance Toll-like receptor-dependent IL-10 induction, which was TRAF6-dependent, but the manner in which A52 manipulates TRAF6 to stimulate p38 activation was unclear. Here, we show that A52 has a non-canonical TRAF6-binding motif that is essential for TRAF6 binding and p38 activation but dispensable for NFκB inhibition and IRAK2 interaction. Wild-type A52, but not a mutant defective in p38 activation and TRAF6 binding (F154A), caused TRAF6 oligomerization and subsequent TRAF6-TAK1 association. The crystal structure of A52 shows that it adopts a Bcl2-like fold and exists as a dimer in solution. Residue Met-65 was identified as being located in the A52 dimer interface, and consistent with that, A52-M65E was impaired in its ability to dimerize. A52-M65E although capable of interacting with TRAF6, was unable to cause either TRAF6 self-association, induce the TRAF6-TAK1 association, or activate p38 MAPK. The results suggest that an A52 dimer causes TRAF6 self-association, leading to TAK1 recruitment and p38 activation. This reveals a molecular mechanism whereby poxviruses manipulate TRAF6 to activate MAPKs (which can be proviral) without stimulating antiviral NFκB activation.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , TNF Receptor-Associated Factor 6/metabolism , Vaccinia virus/metabolism , Vaccinia/metabolism , Viral Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Substitution , Animals , Enzyme Activation , HEK293 Cells , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Mutation, Missense , Protein Binding , Protein Multimerization , TNF Receptor-Associated Factor 6/genetics , Vaccinia/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics
6.
AIDS Rev ; 15(3): 171-80, 2013.
Article in English | MEDLINE | ID: mdl-24002201

ABSTRACT

The treatment of HIV-1 infected patients with HAART has resulted in long-term suppression of viral replication and reduced progression to AIDS. However, the use of HAART has been associated with adverse effects, including metabolic dysregulation and changes in body fat deposition. This syndrome, known as HIV/HAART-associated lipodystrophy syndrome, is characterized by insulin resistance, dyslipidemia, lipodystrophy, and increased visceral adiposity, which contribute to an increased risk of cardiovascular disease amongst these patients. The thiazolidinediones are a class of agonists for the nuclear receptors, the peroxisome proliferator-activated receptor. Since peroxisome proliferator-activated receptor is critically involved in the regulation of insulin sensitivity and lipid metabolism, a number of clinical trials have analyzed whether thiazolidinediones could ameliorate the signs of HIV/HAART-associated lipodystrophy syndrome. Based on these trials, thiazolidinediones appear to up-regulate peroxisome proliferator-activated receptor-dependent genes such as adiponectin, an effect that could have important physiological benefits in the long-term for HIV/HAART-associated lipodystrophy syndrome patients. Critically, many of the studies were of short duration and thus the beneficial effects of thiazolidinediones might have been missed. In addition, the few studies on the thiazolidinedione pioglitazone showed a beneficial effect on limb fat mass that was not associated with a pro-atherogenic lipid profile. Based on these studies, a large-scale clinical trial of pioglitazone use in HIV/HAART-associated lipodystrophy syndrome patients is warranted.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , HIV Infections/drug therapy , HIV-Associated Lipodystrophy Syndrome/drug therapy , Hypoglycemic Agents/therapeutic use , PPAR gamma/drug effects , Thiazolidinediones/therapeutic use , Disease Progression , Female , HIV Infections/metabolism , HIV-Associated Lipodystrophy Syndrome/metabolism , Humans , Male , Pioglitazone , Treatment Outcome , Up-Regulation , Virus Replication/drug effects
7.
BMC Ophthalmol ; 12: 46, 2012 Sep 02.
Article in English | MEDLINE | ID: mdl-22937835

ABSTRACT

BACKGROUND: Systemic therapeutics targeting the peroxisome proliferator-activated receptors have been found to be beneficial in the treatment of diabetic retinopathy. In this paper, we provide a rationale for the use of these therapeutics as intraocular agents. In addition, we introduce the peroxisome proliferator-activated receptors and describe their functions in response to the drugs. DISCUSSION: Based on the evidence of large-scale clinical studies investigating the systemic administration of fenofibrate, this ligand for peroxisome proliferator-activated receptor-α is potentially a good candidate for intraocular delivery. Here, we describe the mechanisms by which it might be acting to improve diabetic retinopathy, its relative safety and we speculate on how it could be developed for intraocular delivery. SUMMARY: In this paper, we provide a rationale for the further investigation of peroxisome proliferator-activated receptor-α agonists as intraocular agents for the treatment of diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/drug therapy , Fenofibrate/administration & dosage , PPAR alpha/agonists , Eye , Humans , Hypolipidemic Agents/administration & dosage , Injections , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...