Next Article in Journal
Epstein–Barr Virus-Induced Post-Transplant Lymphoproliferative Disorder of the Central Nervous System Successfully Treated with Chemo-Immunotherapy
Next Article in Special Issue
Systems Biology Analysis of the Antagonizing Effects of HIV-1 Tat Expression in the Brain over Transcriptional Changes Caused by Methamphetamine Sensitization
Previous Article in Journal
Interaction between PHB2 and Enterovirus A71 VP1 Induces Autophagy and Affects EV-A71 Infection
Previous Article in Special Issue
Oxygen Levels Affect Macrophage HIV-1 Gene Expression and Delay Resolution of Inflammation in HIV-Tg Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure

1
Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
2
Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
*
Author to whom correspondence should be addressed.
Viruses 2020, 12(4), 415; https://doi.org/10.3390/v12040415
Submission received: 28 February 2020 / Revised: 31 March 2020 / Accepted: 4 April 2020 / Published: 8 April 2020
(This article belongs to the Special Issue HIV-1 Transcription Regulation)

Abstract

:
The human immunodeficiency virus type 1 (HIV) establishes a chronic infection that can be well controlled, but not cured, by combined antiretroviral therapy (cART). Interventions have been explored to accomplish a functional cure, meaning that a patient remains infected but HIV is undetectable in the blood, with the aim of allowing patients to live without cART. Tat, the viral transactivator of transcription protein, plays a critical role in controlling HIV transcription, latency, and viral rebound following the interruption of cART treatment. Therefore, a logical approach for controlling HIV would be to block Tat. Tackling Tat with inhibitors has been a difficult task, but some recent discoveries hold promise. Two anti-HIV proteins, Nullbasic (a mutant of Tat) and HT1 (a fusion of HEXIM1 and Tat functional domains) inhibit viral transcription by interfering with the interaction of Tat and cellular factors. Two small molecules, didehydro-cortistatin A (dCA) and triptolide, inhibit Tat by different mechanisms: dCA through direct binding and triptolide through enhanced proteasomal degradation. Finally, two Tat-based vaccines under development elicit Tat-neutralizing antibodies. These vaccines have increased the levels of CD4+ cells and reduced viral loads in HIV-infected people, suggesting that the new vaccines are therapeutic. This review summarizes recent developments of anti-Tat agents and how they could contribute to a functional cure for HIV.

Graphical Abstract

1. Introduction

The human immunodeficiency virus type 1 (HIV) was discovered in 1983 and is the cause of acquired immunodeficiency syndrome (AIDS). Antiretroviral therapy using two or more drugs, cART, can effectively control but not cure HIV infection. Most cART drugs inhibit one of the viral enzymes, reverse transcriptase, integrase or protease, although HIV capsid and the cellular receptors for HIV have also been successfully targeted to inhibit virus replication. cART suppresses viremia to undetectable levels, however, for most people living with HIV (HIV+ people), the infection is lifelong. This is because HIV persists in long-lived cell reservoirs in a wide variety of tissues [1,2,3], mainly in resting memory CD4+ T cells [4,5,6,7,8]. Recent evidence shows that macrophages in brain tissue and urethral mucosa are also HIV reservoirs [9,10], and that these reservoirs are difficult to treat with cART. Ongoing low levels of viral replication and/or homeostatic proliferation of latently infected cells are two proposed explanations for chronic HIV infection [11,12]. The cART regimens currently available do not inhibit viral transcription in HIV-infected cells. Therefore, when cART is discontinued or ineffective, viral rebound in HIV+ people is a reasonable certainty [13].
The precise mechanisms that establish HIV latency are not fully understood. Several factors impacting transcriptional silencing have been reviewed in detail elsewhere, including the site of HIV integration in host genome, the availability of host transcription factors including NF-κB and cyclin T1 (CycT1), and chromatin structure [14,15,16,17,18]. Dynamic interplay between the activation state of T cells infected by HIV and viral regulatory proteins [19,20,21], especially Tat [22,23,24], is an important regulator of HIV latency and reactivation. When CD4+ T cells undergo an effector-to-memory T cell transition, there can be a temporary upregulation in expression of CCR5, a viral co-receptor that along with CD4 facilitates the entry of HIV into the cell, and a rapid downregulation of cellular gene expression. HIV infection of these cells is characterized by suppressed HIV gene transcription, which establishes latency [25]. Cell models show polarized effector T cells infected by HIV enter into a quiescent stage, forcing HIV into latency [26]. In a study of populations of newly infected primary T cells, an analysis of viral RNA by single cell RNAseq indicated that the transcriptional programs of T cells with markers of central memory and naïve T cells are associated with viral latency [27], whereas Tat has been implicated as a master controller of HIV latency regardless of the cell state [22,23,24]. Another recent study demonstrated that HIV silencing and reactivation are heterogeneous and are affected by factors intrinsic to the virus. By using naturally occurring genetic variations that affect Tat expression, this study showed that viruses that poorly express Tat require a proportionally increased stimulus to achieve reactivation from latency. This result suggests that silencing and reactivation behaviors of HIV exist in a spectrum and are influenced by factors intrinsic to the virus, including Tat [28].
As Tat is essential for virus transcription and replication, antiviral agents that block Tat function might block virus production by infected cells. Here we provide a review of Tat function in HIV transcription, recent discoveries and developments of anti-Tat agents, and the possible contribution of anti-Tat agents to a functional cure for HIV infection.

2. Tat Transactivation of HIV Transcription

Tat is an essential viral protein for HIV replication. The functions of Tat, including its roles in HIV transcription, have been described in detail elsewhere [29]. Briefly, Tat is a small viral protein comprising 101 amino acids, in most HIV strains, encoded by two exons (Figure 1a) [30]. The first exon encodes 72 amino acids and is divided into 5 regions: a proline-rich acidic N-terminus (amino acids 1–21), a cysteine-rich region (amino acids 22–37), a hydrophobic core region (amino acids 38–48), an arginine-rich basic region (amino acids 49–57), and a glutamine-rich region (amino acids 58–72). Exon 2 encodes a C-terminal domain. The first 48 amino acids are the activation domain, followed by the basic domain (amino acids 49–57). The basic domain of Tat is required for Tat’s nuclear localization and the binding of Tat to the HIV RNA stem loop structure, called the transactivation response (TAR) RNA, as well as many other Tat functions [31]. Without Tat, HIV transcription by RNA polymerase II (RNAP II) is very inefficient and viral RNA transcripts corresponding to TAR RNA are less than 60 nucleotides long [32,33,34,35], as observed in cell line models of HIV latency and in HIV+ people [36]. Importantly, non-productively infected cells can be activated by cell stimuli to make infectious viruses [36].
A key point is that the HIV promoter is capable of a low basal level of transcription [37], that may stochastically produce a full length viral mRNA [22,38], which can be spliced into tat-encoding mRNA. The availability of Tat in an HIV-infected cell initiates a powerful feedback system that greatly increases HIV transcription (reviewed in [29,39]). Briefly, the activation domain of Tat can bind to the positive transcription elongation factor b (P-TEFb), a cellular complex composed of cyclin T1 (CycT1) and CDK9. In the cell, P-TEFb can be associated with the 7SK small nuclear ribonucleoprotein (snRNP), the Brd4 complex or the super elongation complex (SEC), and Tat can exploit P-TEFb from each of these resources [40]. Tat mediates binding of the Tat:P-TEFb complex at the HIV long terminal repeat (LTR) promoter (Figure 1b), which is achieved by specific binding of Tat:P-TEFb to TAR RNA. This leads to several important events that upregulate HIV transcription up to 100-fold over basal expression levels: 1) CDK9 phosphorylates the RNAP II associated transcriptional suppressors DRB sensitivity-inducing factor (DISF) and the negative transcription elongation factor (NELF), which dissociates from the RNAP II elongation complex; 2) CDK9 hyperphosphorylates the C-terminal repeat domain of RNAP II, which greatly increases productive transcription of full length viral mRNA, and; 3) cellular and viral proteins including Tat recruit chromosome remodeling histone acetyltransferases, so that nucleosomes flanking the LTR promoter are in an open euchromatin configuration of highly transcribed genes in cells.

3. A Tat-Based Block-and-Lock Functional Cure Strategy

Recently, a functional cure for HIV has been proposed that uses a block-and-lock strategy, in which anti-HIV agents that prevent transcription of HIV lock the cellular HIV reservoir into a “deep”-latent, transcriptionally silent state, preventing rebound after cessation of cART [41,42,43,44,45,46,47,48,49,50]. The block-and-lock strategy differs from the shock-and-kill strategy [51,52], where latency-reversing agents (LRAs) are used to stimulate and reactivate, or shock, latent HIV in the cellular reservoirs so that infected cells die as a direct result of virus production or are killed by the immune response to HIV. As Tat transactivation is a critical step in HIV transcription, agents that block Tat will inhibit HIV replication and viral transcription. Recent modeling suggests inhibitors that interact with Tat and disrupt the transactivation cycle, leading to epigenetic modifications of nucleosomes in the viral promoter, could cause prolonged inactivation, or deep latency, of HIV transcription [53]. The identification of Tat inhibitors is the result of more than two decades of research [54]. While none of the transactivation inhibitors discovered has advanced to clinical use, research on several Tat targeting agents continues.

4. Tat Trans-Dominant Negative Proteins and Nullbasic

4.1. Tat Trans-Dominant Negative Proteins

Tat mutants that inhibit HIV transactivation were first reported more than 30 years ago and were referred to as trans-dominant negative (TDN) proteins [55,56]. Early studies used a 72 amino acid TDN Tat encoded by the first exon where some but not all basic domain residues were mutated to glycine; this mutant Tat was found to inhibit HIV transcription if present at ~20-fold excess compared to wild type Tat. While the first TDN Tats were not powerful inhibitors of HIV transcription, a combined treatment of TDN Tat and TDN Rev (called M10) [57,58,59] showed greater inhibition of HIV replication than either as an individual treatment [60].
The basic domain of Tat is essential for TAR RNA interaction and has versatile roles in the interaction of Tat and the host cell factors, which have been reviewed by Kurnaeva et al. [31]. Tat interacts with HIV reverse transcriptase and stimulates reverse transcription [61,62]. Acetylation of Lys50 and Lys51 in the basic domain appears to regulate this activity [63]. Nullbasic is a 101 amino acid TDN Tat in which the entire basic domain is replaced by Gly and Ala residues [64]. Studies show that, when expressed in cell lines, Nullbasic inhibits Tat transactivation [64], reverse transcription [64,65] and Rev activity [64,66,67] (Table 1).

4.2. Nullbasic Inhibits HIV Transcription and Reactivation from Latency

When Jurkat cells expressing Nullbasic were infected with HIV, HIV replication was completely inhibited [69]. Although HIV proviral DNA could be detected in chromosomes of the infected cells, no measurable HIV RNA was identified in these cells. A co-culture experiment where infected Jurkat cells were mixed with uninfected Jurkat cells failed to rescue infectious virus, suggesting that HIV production was blocked in the infected cells. Transcription from proviral DNA in the cells could not be reactivated by LRAs including phorbol ester 12-myristate-13-acetate (PMA), which activates protein kinase C and reactivates NF-κB [88], and JQ-1, which binds to BRD4 and enables increased Tat binding to P-TEFb [89]. The histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA), which is a strong activator of latent cells [90,91,92,93], only modestly stimulated HIV transcription [69]. This suggests that Nullbasic forces HIV into a latent state in Jurkat cells, refractory to LRA reactivation.
Nullbasic also strongly inhibited HIV replication in human primary CD4+ cells [68,70], and against different HIV subtypes [94]. This is most likely because Nullbasic targets cellular factors such as P-TEFb [66] that are essential for replication by all HIV subtypes. Nullbasic was tested in two mouse models of acute HIV infection where mice were engrafted with primary human CD4+ cells. The cells were either transduced with Nullbasic first and then infected with HIV (pre-infection treatment), or infected with HIV first and then transduced with Nullbasic (post-infection treatment) [70]. In the pre-infection model, Nullbasic inhibited HIV transcription up to 2800 fold in CD4+ cells recovered from organs, and no HIV was detected in blood samples [70]. In the post-infection model, the inhibitory effect of Nullbasic was less robust, but suggested that Nullbasic provided CD4+ cells a survival benefit [70].
A gene therapeutic approach to cure HIV remains an important area of research because of the potential for durable antiviral effects. Nullbasic inhibits three different steps of HIV replication, which may explain, in part, why it is a potent inhibitor of HIV. Given the inhibition observed in this simple animal model of an acute and active HIV infection, Nullbasic could be a part of a block-and-lock functional cure strategy as a gene therapy candidate.

5. A HEXIM1-Tat Fusion Protein that Inhibits HIV Replication

P-TEFb activity is controlled by the 7SK snRNA complex [95]. In this complex, HEXIM1 binds to the 7SK snRNA through its arginine-rich RNA-binding domain (ARM) [96] and inhibits the kinase activity of CDK9 in P-TEFb complex with its central inhibitory domain (ID) [97,98]. HEXIM1 and its paralog HEXIM2 inhibits Tat activity by recruiting CycT1 to 7SK snRNA [71]. A fusion protein named HT1 was derived from HEXIM1 (amino acids 150–220, including ARM and ID domains) and Tat (amino acids 1–48; the domain binds to P-TEFb) [72,99] (Table 1). Cell based experiments demonstrated that HT1 inhibited HIV transcription by binding to TAR RNA, inactivated CDK9 activity of P-TEFb and competed with Tat for binding to P-TEFb. Expression of HT1 had no significant effect on host cell gene expression and growth. In cell line models of HIV latency, HT1 inhibited viral reactivation by PMA stimulation [72]. These results support the investigation of HT1 as a potential gene therapy agent in a block-and-lock functional cure strategy. However, HT1 has not been evaluated in animal models of HIV infection.

6. Two Compounds that Inhibit Tat Activity

6.1. Didehydro-Cortistatin A (dCA)

dCA is a synthetic analogue of the natural steroidal alkaloid Cortistatin A (CA) from the marine sponge Corticium simplex (Table 1) [100]. CA was reported to bind and inhibit mediator kinases including CDK8, CDK11, and CDK19 [101,102], but does not inhibit HIV transcription [103]. dCA potently inhibited Tat transactivation by specifically binding to Tat via the basic domain [73], but did not bind to CDK9 in the P-TEFb complex. dCA also specifically bound to the basic domain of HIV-2 Tat [73] and SIV Tat [76]. The binding of dCA to the basic domain of Tat occurred across HIV subtypes A to E in the main group M [104], but dCA did not bind to the basic domain of Rev and HEXIM1 [74]. dCA caused a redistribution of Tat in the nucleus from the nucleolus to the nucleoplasm and periphery of the nucleolus in a dose-dependent manner [73]. In vitro cell based experiments have demonstrated that dCA inhibited virus transactivation by Tat in different HIV subtypes [104] and SIV [76]. dCA did not inhibit the initiation of transcription but inhibited the interaction of RNAP II with the LTR promoter [46].
Histone modifications regulate the accessibility of chromatin DNA to transcription factors, and acetylation of histone proteins in nucleosomes is required for HIV transactivation [105]. Research using the latently infected OM-10.1 cell line showed that the inhibition of Tat by dCA resulted in an extremely repressive chromatin environment at the HIV promoter, characterized by decreased H3K27 acetylation levels at nucleosome 1 (located downstream of the HIV LTR transcription start site) and limited PBAF recruitment [75]. PBAF is a type of SWI/SNF chromatin remodeler recruited by acetylated Tat to the viral promoter [106]. The activity of dCA is directly correlated to the strength of the Tat-TAR feedback loop [75]. dCA only partially blocked HIV reactivation in U1 cells and produced no significant inhibition of HIV reactivation of ACH2 cells [75]. This may be because these cell lines harbor defective HIV proviruses that have either a defect in TAR RNA (U1 cells) or Tat (ACH2 cells) that incompletely impede transactivation, so that increased basal levels of transcription in these cell lines can be induced by activation of NF-κB. dCA prevents HIV reactivation from latency in primary CD4+ cells derived from infected individuals and in many cellular models of latency [46]. In a bone marrow-liver-thymus (BLT) HIV mouse model, adding dCA to ART reduced viral mRNA in tissues, and both delayed and reduced HIV rebound after an ART interruption [107]. These results suggest that dCA may be useful clinically.
HIV can rapidly become resistant to any single anti-viral drug, so it is not surprising that HIV strains resistant to dCA emerged from long-term cultures [108,109]. No mutations in Tat and TAR were identified in these strains. The mutations in the dCA resistant strains were identified in the LTR region, Nef and Vpr. Mutations in the LTR region increased basal HIV transcription by 10- to 30-fold compared the wild type LTR, while the Nef and Vpr mutants increased NF-κB nuclear translocation, thereby facilitating transcription from the HIV LTR promoter. In primary CD4+ cells, dCA resistant virus produced up to 150-fold more virus than WT in the presence of dCA. Whether dCA-resistant strains can sustain infection in vivo remains an important question.
Clearly dCA is a promising anti-Tat agent that will likely be tested in primate models of HIV infection. It remains to be seen if dCA will be a viable addition to traditional ART as part of a block-and-lock HIV cure strategy.

6.2. Triptolide

Triptolide is a natural product extracted from a traditional Chinese herbal medicine, Tripterygium wilfordii Hook F, which has been used for the treatment of rheumatoid arthritis [110,111]. Triptolide also exhibits anti-inflammatory and anti-cancer properties [112,113,114]. The anti-inflammatory mechanism involves inhibition of NF-κB activation and DNA binding of NF-AT to enhancer sites in the IL-2 promoter [115]. Triptolide is a global transcriptional inhibitor of RNAP I, II and III [116,117,118,119], which partly explains its anti-cancer properties. Triptolide inhibits both total RNA and mRNA de novo synthesis in A549 cells. Triptolide represses the expression of RPB1 (the main RNAP II subunit) and MYC proteins, inducing an accumulation of p53 in A549 cells [116]. Triptolide also binds covalently to XPB, a subunit of TFIIH (a component of the pre-initiation complex of RNAP II), and blocks transcriptional initiation [120]. Triptolide can alter RNAP II promoter occupancy [121]. A recent paper showed that triptolide reduced the formation of TFIIIB, a multi-subunit transcription factor for RNAP III at the promoters of tRNAs and 5S rRNA in colorectal cancer cells [119].
Triptolide has also been investigated as an anti-HIV reagent at concentrations that do not affect global cellular transcription [77] (Table 1). The anti-HIV activity of Triptolide has been tested in HeLa-derived TZM-bl cells [122,123,124], which have a Tat-responsive HIV LTR promoter expressing reporter genes encoding luciferase and beta-lactamase, as well as in Jurkat cells and human peripheral blood cells infected with different HIV strains (HIVNL 4-3, HIVLAI and HIVBaL). Triptolide inhibited HIV replication from about 89% to >99% in these cells. While triptolide did not affect the viability of TZM-bl and Jurkat cells, it reduced the viability of primary blood cells by about 25%. Time-of-addition experiments indicated that triptolide inhibited HIV transcription by targeting Tat. Triptolide was reported to reduce Tat steady-state levels by enhancing its proteasomal degradation, which could be blocked by the proteasomal inhibitor MG132. Protein arginine methyltransferase 6 (PRMT6) can methylate Tat, Rev and nucleocapsid protein and regulate Tat nuclear localization. Research has suggested that overexpression of PRMT6 can increase Tat steady-state levels in most cell lines [125]. A connection between PRMT6 and triptolide effects on Tat steady-state levels has not been investigated, but analysis of Tat mutants suggested an interaction between Tat with PRMT6 and the triptolide-mediated decrease of Tat steady-state levels both required the Tat activation domain.
However, due to its global inhibition of transcription, the clinical application of triptolide has been limited by its toxicity and side effects [126]. The utility of triptolide and Triptolide wilfordii as alternative ART await outcomes form clinical trials (NCT02219672 and NCT03403569).

7. Anti-Tat Vaccines that may Compliment cART

The prospect that a therapeutic anti-Tat vaccine could be an important approach to tackle HIV infection has been long argued [127]. As a powerful activator of HIV transcription, Tat enables rapid viremia and is hypothesized to regulate establishment of a cellular reservoir of infected cells [22,128]. The high mutability of HIV and subsequent generation of variant strains enables the virus to escape cellular and humoral immune responses that would normally control viremia. However HIV-infected cells are reported to secrete Tat in cell culture experiments [129] and in vivo [130,131]. Tat lacks a conventional signal sequence and is secreted through an unconventional secretion pathway utilized by fibroblast growth factor 2 [132], which requires the phospholipid component of the inner leaflet of the plasma membrane, phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) [133]. The secreted Tat can be passed into uninfected cells, where it can render the cells more susceptible to infection [134], disrupt immune response by inducing apoptosis of T lymphocytes [135], and down-modulate expression of MHC class I [136]. In addition, the secreted Tat can traffic to the central nervous system by crossing the blood–brain barrier [137], where it plays a role in HIV-associated neurocognitive disorders [138,139]. While antibodies against Tat can block cellular uptake in a cell based in vitro assay system [140], anti-Tat antibodies are relatively uncommon in HIV+ people and have been shown to decline with time [141]. Nevertheless, reports that the presence of anti-Tat antibodies in serum of HIV+ people correlates with decreased progression to AIDS [142,143,144,145] support the need for research into a safe and effective vaccine that elicits cellular and humoral immune responses to Tat.
HIV viruses are classified into four groups: M (major), O (outlier), N (non-M non-O) and P; a vast majority of HIV infections are induced by group M, which includes 9 subtypes and more than 70 circulating recombinant forms [146]. Vaccines undergoing animal and human clinical trials have used Tat protein derived from HIV strain BH10 [147] and Oyi [148], which are both group M subtype B viruses. There are differences in the primary amino acid sequences between Tat-BH10 and Tat-Oyi (Figure 2). Tat-BH10 is an 86 amino acid protein while Tat-Oyi, like a majority of Tat proteins in nature, has 101 amino acids. Tat-BH10 is a functional protein capable of transactivating the HIV LTR promoter, while Tat-Oyi is not. A Tat-Oyi C22S amino acid substitution, which is not present in Tat-BH10 and all other functional Tat isolates, can explain its defective phenotype in transactivation [149]. Finally, vaccination clinical trials have used Tat-BH10 purified from E. coli, while the Tat-Oyi used in vaccine trials was synthesized.
Both types of Tat vaccine were tested in non-human primate (SHIV) models, producing conflicting results regarding efficacy [80,81,87,151,152,153]. Here we recap recent small-scale phase l/lla vaccination clinical trials with Tat-BH10 or Tat-Oyi [78,79,82,86,154,155] (Table 1). In these trials, HIV-infected participants were treated with cART, which controlled viremia. The Tat-BH10 trial (see ISS T-002, also referred to as NCT0102455) did not include a double blinded, randomized controlled arm but did include a non-vaccinated HIV+ matched group for comparisons [84]. Vaccinees in the ISS T-002 study have been monitored for up to 8 years. A parallel Tat-BH10 trial (ISS T-003, yet to report) is randomized, blinded and includes a placebo arm. The Tat-Oyi trial was a randomized controlled study involving 48 people in four groups followed for 12 months [86]. Interestingly, optimal immune responses were reported when Tat-BH10 and Tat-Oyi were administered intradermally at 30 or 33 µg, respectively, and given 3 times, once monthly and without adjuvant. Both vaccines were found to be safe, and each vaccine produced humoral antibodies that recognize Tat. Tat-BH10 vaccination (ISS T-002) resulted in anti-Tat IgG, IgM and IgA antibodies being detected in ~90% of vaccinees in 6 months, which waned to ~37% after 8 years [83]. It is reported that only 12% to 20% of HIV+ people with chronic infection have antibodies to Tat [141,144,156,157,158,159]. At the start of the Tat-Oyi trial, >65% of subjects had no or low levels of antibodies to Tat. From months 5 to 12 post-inoculation with 33 µg or 96 µg of Tat-Oyi, ~70 to 80% of Tat-Oyi vaccinees developed anti-Tat IgG antibodies that recognize Tat from two to six of the most common HIV subtypes. However, these responders were not tested for levels of anti-Tat IgM and IgA. The number of responders with anti-Tat antibodies to at least three subtypes diminished by about 33% after 12 months post-inoculation compared to pre-inoculation levels. Likewise, Tat-BH10 vaccine responders produced antibodies that recognize Tat for HIV subtypes A, B, C and D. These antibodies may be therapeutic. Tat-BH10 responders had increased levels of CD4+ cells compared to baseline, as well as reduced viral loads in plasma and proviral DNA load in PBMCs [79,83]; and Tat-Oyi vaccine responders had reduced viral rebound following cessation of cART [86]. The evidence suggests that restoration of cell mediated immunity contributed to decreased proviral load. Clearly, these are encouraging outcomes that warrant further investigations.
Some questions remain regarding the overall efficacy of vaccines based on a single Tat protein. Although Tat functional domains from human and primate lentiviruses are relatively well conserved [160], numerous amino acid substitutions have been identified throughout the length of Tat [161]. As Tat requires functional domains to interact with cellular factors and TAR RNA for transactivation of HIV transcription, viral escape through Tat adaptation to an immune response could be limited. An early study using Tat peptides identified the Tat cysteine-rich and basic domains as epitopes for IgM responses to Tat [162], but there is evidence that Tat, although a poorly structured, unfolded protein [163], most likely presents conformational epitopes [85]. Interestingly an analysis of more than 300 Chinese HIV+ people with Tat antibodies showed neutralizing potential that correlated with a Tat N-terminal bias (amino acids 1–48) [157], although it is not known if anti-Tat responses in the Chinese cohort delayed disease progression. Finally, a new vaccine against Tat must be effective in a populations where there will be diverse HLA class-I and class-II variations, which may influence the overall efficacy of a Tat-based vaccine.

8. Conclusions

cART has greatly improved quality of life and decreased progression to AIDS for HIV+ people. The advent of pre-exposure prophylaxis (PrEP) [164], where uninfected people at high risk of contracting HIV infection take cART, has reduced new infections. While viral loads in HIV+ people can be strongly suppressed by cART, successful treatment for longer than 10 years does not appreciably decrease the size of the latent reservoirs [165], and for this reason interruption of cART results in a rapid viral rebound. HIV latency is established in resting T cells where little or no P-TEFb can be measured [166,167]. We suggest that the development of an agent that inhibits newly synthesized Tat and therefore the production of virus by infected cells would be an important step towards a functional cure [127]. Both Nullbasic and HT1 can strongly inhibit HIV transcription, and the former has been tested in mouse models of acute HIV infection. Their application would most likely require a gene therapy approach in hematopoietic stem cells. Whether immune cells protected from HIV infection by Nullbasic or HT1 would escape preexisting humoral and cellular immune responses to Tat is unknown. However, a phase I/II clinical trial of a TDN Rev called RevM10 showed stable expression for >100 weeks and preferential survival of transduced cells in one patient [168]. dCA has the remarkable ability to inactivate Tat and pre-clinical studies suggest that it can “lock” the HIV promoter so that transcription by RNAP II is inhibited durably even after dCA treatment is stopped. However, large-scale dCA production may be difficult [100], which could limit its availability. As the molecular interaction between dCA and Tat has been modeled [74], a future in silico screening project using pharmacophore based models of dCA interaction with Tat may identify additional agents with anti-Tat activity. Drug repurposing of the rheumatoid arthritis drug triptolide as an HIV inhibitor awaits safety and efficacy outcomes in clinical trials. Triptolide affects global transcription, which may limit its utility in safely controlling HIV infection. Anti-Tat vaccines have advanced to small clinical trials and 8-year follow-up results of a TatBH10 vaccine appear promising. Successive trials will be required to test the efficacy in a large cohort of HIV+ people. As no anti-Tat agents are available for clinical use, additional translational research of anti-Tat strategies should be a priority task.

Author Contributions

H.J., D.L., M.-H.L., L.L. and D.H. all edited and provided ideas for this article; D.H. and H.J. generated the figure and H.J. wrote the initial draft of the article. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Australian Centre for HIV and Hepatitis Research (ACH2), QIMR Berghofer Medical Research Institute and Project Grant APP1085359 from the National Health and Medical Research Council to DH.

Acknowledgments

We thank Siall Waterbright for her professional editorial assistance during production of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alexaki, A.; Liu, Y.; Wigdahl, B. Cellular reservoirs of HIV-1 and their role in viral persistence. Curr. HIV Res. 2008, 6, 388–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Araínga, M.; Edagwa, B.; Mosley, R.L.; Poluektova, L.Y.; Gorantla, S.; Gendelman, H.E. A mature macrophage is a principal HIV-1 cellular reservoir in humanized mice after treatment with long acting antiretroviral therapy. Retrovirology 2017, 14, 17. [Google Scholar] [CrossRef] [Green Version]
  3. Wong, J.K.; Yukl, S.A. Tissue reservoirs of HIV. Curr. Opin. HIV AIDS 2016, 11, 362–370. [Google Scholar] [CrossRef] [PubMed]
  4. Siliciano, J.D.; Kajdas, J.; Finzi, D.; Quinn, T.C.; Chadwick, K.; Margolick, J.B.; Kovacs, C.; Gange, S.J.; Siliciano, R.F. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat. Med. 2003, 9, 727–728. [Google Scholar] [CrossRef] [PubMed]
  5. Crooks, A.M.; Bateson, R.; Cope, A.B.; Dahl, N.P.; Griggs, M.K.; Kuruc, J.D.; Gay, C.L.; Eron, J.J.; Margolis, D.M.; Bosch, R.J.; et al. Precise quantitation of the latent HIV-1 reservoir: Implications for eradication strategies. J. Infect. Dis. 2015, 212, 1361–1365. [Google Scholar] [CrossRef]
  6. Siliciano, J.M.; Siliciano, R.F. The remarkable stability of the latent reservoir for HIV-1 in resting memory CD4+ T cells. J. Infect. Dis. 2015, 212, 1345–1347. [Google Scholar] [CrossRef] [Green Version]
  7. Cantero-Pérez, J.; Grau-Expósito, J.; Serra-Peinado, C.; Rosero, D.A.; Luque-Ballesteros, L.; Astorga-Gamaza, A.; Castellví, J.; Sanhueza, T.; Tapia, G.; Lloveras, B.; et al. Resident memory T cells are a cellular reservoir for HIV in the cervical mucosa. Nat. Commun. 2019, 10, 4739. [Google Scholar]
  8. Baxter, A.E.; Niessl, J.; Fromentin, R.; Richard, J.; Porichis, F.; Charlebois, R.; Massanella, M.; Brassard, N.; Alsahafi, N.; Delgado, G.-G.; et al. Single-cell characterization of viral translation-competent reservoirs in HIV-infected individuals. Cell Host Microbe 2016, 20, 368–380. [Google Scholar] [CrossRef] [Green Version]
  9. Ganor, Y.; Real, F.; Sennepin, A.; Dutertre, C.-A.; Prevedel, L.; Xu, L.; Tudor, D.; Charmeteau, B.; Couedel-Courteille, A.; Marion, S.; et al. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat. Microbiol. 2019, 4, 633–644. [Google Scholar] [CrossRef] [PubMed]
  10. Ko, A.; Kang, G.; Hattler, J.B.; Galadima, H.I.; Zhang, J.; Li, Q.; Kim, W.-K. Macrophages but not astrocytes harbor HIV DNA in the brains of HIV-1-infected aviremic individuals on suppressive antiretroviral therapy. J. Neuroimmune Pharmacol. 2019, 14, 110–119. [Google Scholar] [CrossRef] [Green Version]
  11. Sigal, A.; Kim, J.T.; Balazs, A.B.; Dekel, E.; Mayo, A.; Milo, R.; Baltimore, D. Cell-to-cell spread of HIV permits ongoing replication despite antiretroviral therapy. Nature 2011, 477, 95–98. [Google Scholar] [CrossRef] [PubMed]
  12. Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; Procopio, F.A.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef]
  13. Colby, D.J.; Trautmann, L.; Pinyakorn, S.; Leyre, L.; Pagliuzza, A.; Kroon, E.; Rolland, M.; Takata, H.; Buranapraditkun, S.; Intasan, J.; et al. Rapid HIV RNA rebound after antiretroviral treatment interruption in persons durably suppressed in Fiebig I acute HIV infection. Nat. Med. 2018, 24, 923–926. [Google Scholar] [CrossRef] [PubMed]
  14. Van Lint, C.; Bouchat, S.; Marcello, A. HIV-1 transcription and latency: An update. Retrovirology 2013, 10, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hakre, S.; Chavez, L.; Shirakawa, K.; Verdin, E. Epigenetic regulation of HIV latency. Curr. Opin. HIV AIDS 2011, 6, 19–24. [Google Scholar] [CrossRef] [PubMed]
  16. Trono, D.; Van Lint, C.; Rouzioux, C.; Verdin, E.; Barre-Sinoussi, F.; Chun, T.W.; Chomont, N. HIV persistence and the prospect of long-term drug-free remissions for HIV-infected individuals. Science 2010, 329, 174–180. [Google Scholar] [CrossRef]
  17. Cary, D.C.; Fujinaga, K.; Peterlin, B.M. Molecular mechanisms of HIV latency. J. Clin. Investig. 2016, 126, 448–454. [Google Scholar] [CrossRef]
  18. Elsheikh, M.M.; Tang, Y.; Li, D.; Jiang, G. Deep latency: A new insight into a functional HIV cure. EBioMedicine 2019, 45, 624–629. [Google Scholar] [CrossRef] [Green Version]
  19. Duverger, A.; Wolschendorf, F.; Zhang, M.; Wagner, F.; Hatcher, B.; Jones, J.; Cron, R.Q.; van der Sluis, R.M.; Jeeninga, R.E.; Berkhout, B.; et al. An AP-1 binding site in the enhancer/core element of the HIV-1 promoter controls the ability of HIV-1 to establish latent infection. J. Virol. 2013, 87, 2264–2277. [Google Scholar] [CrossRef] [Green Version]
  20. Verdin, E.; Paras, P.; Van Lint, C. Chromatin disruption in the promoter of human immunodeficiency virus type 1 during transcriptional activation activation [published erratum appears in EMBO J. 1993, 12, 4900]. EMBO J. 1993, 12, 3249–3259. [Google Scholar] [CrossRef]
  21. Van Lint, C.; Emiliani, S.; Ott, M.; Verdin, E. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J. 1996, 15, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
  22. Razooky, B.S.; Pai, A.; Aull, K.; Rouzine, I.M.; Weinberger, L.S. A hardwired HIV latency program. Cell 2015, 160, 990–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Cao, Y.; Lei, X.; Ribeiro, R.M.; Perelson, A.S.; Liang, J. Probabilistic control of HIV latency and transactivation by the Tat gene circuit. Proc. Natl. Acad. Sci. USA 2018, 115, 12453. [Google Scholar] [CrossRef] [PubMed]
  24. Chavez, L.; Calvanese, V.; Verdin, E. HIV latency is established directly and early in both resting and activated primary CD4 T cells. PLOS Pathog. 2015, 11, e1004955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Shan, L.; Deng, K.; Gao, H.; Xing, S.; Capoferri, A.A.; Durand, C.M.; Rabi, S.A.; Laird, G.M.; Kim, M.; Hosmane, N.N.; et al. Transcriptional reprogramming during effector-to-memory transition renders CD4+ T cells permissive for latent HIV-1 infection. Immunity 2017, 47, 766–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Dobrowolski, C.; Valadkhan, S.; Graham, A.C.; Shukla, M.; Ciuffi, A.; Telenti, A.; Karn, J. Entry of polarized effector cells into quiescence forces HIV latency. mBio 2019, 10, e00337-00319. [Google Scholar] [CrossRef] [Green Version]
  27. Bradley, T.; Ferrari, G.; Haynes, B.F.; Margolis, D.M.; Browne, E.P. Single-cell analysis of quiescent HIV infection reveals host transcriptional profiles that regulate proviral latency. Cell Rep. 2018, 25, 107–117. [Google Scholar] [CrossRef] [Green Version]
  28. Norton, N.J.; Mok, H.P.; Sharif, F.; Hirst, J.C.; Lever, A.M.L. HIV silencing and inducibility are heterogeneous and are affected by factors intrinsic to the virus. mBio 2019, 10, e00188-00119. [Google Scholar] [CrossRef] [Green Version]
  29. Clark, E.; Nava, B.; Caputi, M. Tat is a multifunctional viral protein that modulates cellular gene expression and functions. Oncotarget 2017, 8, 27569–27581. [Google Scholar] [CrossRef] [Green Version]
  30. Sivakumaran, H.; Cutillas, V.; Harrich, D. Revisiting transdominant-negative proteins in HIV gene therapy. Future Virol. 2013, 8, 757–768. [Google Scholar] [CrossRef]
  31. Kurnaeva, M.A.; Sheval, E.V.; Musinova, Y.R.; Vassetzky, Y.S. Tat basic domain: A “Swiss army knife” of HIV-1 Tat? Rev. Med. Virol. 2019, 29, e2031. [Google Scholar] [CrossRef]
  32. Feinberg, M.B.; Baltimore, D.; Frankel, A.D. The role of Tat in the human immunodeficiency virus life cycle indicates a primary effect on transcriptional elongation. Proc. Natl. Acad. Sci. USA 1991, 88, 4045–4049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kao, S.Y.; Calman, A.F.; Luciw, P.A.; Peterlin, B.M. Anti-termination of transcription within the long terminal repeat of HIV-1 by tat gene product. Nature 1987, 330, 489–493. [Google Scholar] [CrossRef] [PubMed]
  34. Laspia, M.F.; Rice, A.P.; Mathews, M.B. HIV-1 Tat protein increases transcriptional initiation and stabilizes elongation. Cell 1989, 59, 283–292. [Google Scholar] [CrossRef]
  35. Marciniak, R.A.; Sharp, P.A. HIV-1 Tat protein promotes formation of more-processive elongation complexes. EMBO J. 1991, 10, 4189–4196. [Google Scholar] [CrossRef] [PubMed]
  36. Adams, M.; Sharmeen, L.; Kimpton, J.; Romeo, J.M.; Garcia, J.V.; Peterlin, B.M.; Groudine, M.; Emerman, M. Cellular latency in human immunodeficiency virus-infected individuals with high CD4 levels can be detected by the presence of promoter-proximal transcripts. Proc. Natl. Acad. Sci. USA 1994, 91, 3862–3866. [Google Scholar] [CrossRef] [Green Version]
  37. Nabel, G.; Baltimore, D. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature 1987, 326, 711–713. [Google Scholar] [CrossRef]
  38. Weinberger, L.S.; Burnett, J.C.; Toettcher, J.E.; Arkin, A.P.; Schaffer, D.V. Stochastic gene expression in a lentiviral positive-feedback loop: HIV-1 Tat fluctuations drive phenotypic diversity. Cell 2005, 122, 169–182. [Google Scholar] [CrossRef] [Green Version]
  39. Li, Y.; Liu, M.; Chen, L.F.; Chen, R. P-tefb: Finding its ways to release promoter-proximally paused RNA polymerase II. Transcription 2018, 9, 88–94. [Google Scholar] [CrossRef]
  40. Rice, A.P. The HIV-1 Tat protein: Mechanism of action and target for HIV-1 cure strategies. Curr. Pharm. Des. 2017, 23, 4098–4102. [Google Scholar] [CrossRef] [Green Version]
  41. Dahabieh, M.S.; Battivelli, E.; Verdin, E. Understanding HIV latency: The road to an HIV cure. Annu. Rev. Med. 2015, 66, 407–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Planelles, V. An ounce of Tat prevention is worth a pound of functional cure. mBio 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Vranckx, L.S.; Demeulemeester, J.; Saleh, S.; Boll, A.; Vansant, G.; Schrijvers, R.; Weydert, C.; Battivelli, E.; Verdin, E.; Cereseto, A.; et al. Ledgin-mediated inhibition of integrase-ledgf/p75 interaction reduces reactivation of residual latent HIV. EBioMedicine 2016, 8, 248–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Darcis, G.; Van Driessche, B.; Van Lint, C. HIV latency: Should we shock or lock? Trends Immunol. 2017, 38, 217–228. [Google Scholar] [CrossRef]
  45. Mousseau, G.; Mediouni, S.; Valente, S.T. Targeting HIV transcription: The quest for a functional cure. Curr Top. Microbiol. Immunol. 2015, 389, 121–145. [Google Scholar]
  46. Mousseau, G.; Kessing, C.F.; Fromentin, R.; Trautmann, L.; Chomont, N.; Valente, S.T. The Tat inhibitor didehydro-cortistatin A prevents HIV-1 reactivation from latency. MBio 2015, 6, e00465. [Google Scholar] [CrossRef] [Green Version]
  47. Jean, M.J.; Hayashi, T.; Huang, H.; Brennan, J.; Simpson, S.; Purmal, A.; Gurova, K.; Keefer, M.C.; Kobie, J.J.; Santoso, N.G.; et al. Curaxin CBL0100 blocks HIV-1 replication and reactivation through inhibition of viral transcriptional elongation. Front. Microbiol. 2017, 8, 2007. [Google Scholar] [CrossRef] [Green Version]
  48. Kim, H.; Choi, M.S.; Inn, K.S.; Kim, B.J. Inhibition of HIV-1 reactivation by a telomerase-derived peptide in a HSP90-dependent manner. Sci. Rep. 2016, 6, 28896. [Google Scholar] [CrossRef] [Green Version]
  49. Anderson, I.; Low, J.S.; Weston, S.; Weinberger, M.; Zhyvoloup, A.; Labokha, A.A.; Corazza, G.; Kitson, R.A.; Moody, C.J.; Marcello, A.; et al. Heat shock protein 90 controls HIV-1 reactivation from latency. Proc. Natl. Acad. Sci. USA 2014, 111, E1528–E1537. [Google Scholar] [CrossRef] [Green Version]
  50. Besnard, E.; Hakre, S.; Kampmann, M.; Lim, H.W.; Hosmane, N.N.; Martin, A.; Bassik, M.C.; Verschueren, E.; Battivelli, E.; Chan, J.; et al. The mTOR complex controls HIV latency. Cell Host Microbe 2016, 20, 785–797. [Google Scholar] [CrossRef] [Green Version]
  51. Margolis, D.M.; Garcia, J.V.; Hazuda, D.J.; Haynes, B.F. Latency reversal and viral clearance to cure HIV-1. Science 2016, 353, aaf6517. [Google Scholar] [CrossRef] [Green Version]
  52. Kim, Y.; Anderson, J.L.; Lewin, S.R. Getting the “kill” into “shock and kill”: Strategies to eliminate latent HIV. Cell Host Microbe 2018, 23, 14–26. [Google Scholar] [CrossRef]
  53. Aguilera, L.U.; Rodriguez-Gonzalez, J. Modeling the effect of Tat inhibitors on HIV latency. J. Theor. Biol. 2019, 473, 20–27. [Google Scholar] [CrossRef]
  54. Tabarrini, O.; Desantis, J.; Massari, S. Recent advances in the identification of Tat-mediated transactivation inhibitors: Progressing toward a functional cure of HIV. Future Med. Chem. 2016, 8, 421–442. [Google Scholar] [CrossRef]
  55. Pearson, L.; Garcia, J.; Wu, F.; Modesti, N.; Nelson, J.; Gaynor, R. A transdominant Tat mutant that inhibits Tat-induced gene expression from the human immunodeficiency virus long terminal repeat. Proc. Natl. Acad. Sci. USA 1990, 87, 5079–5083. [Google Scholar] [CrossRef] [Green Version]
  56. Modesti, N.; Garcia, J.; Debouck, C.; Peterlin, M.; Gaynor, R. Trans-dominant Tat mutants with alterations in the basic domain inhibit HIV-1 gene expression. New Biol. 1991, 3, 759–768. [Google Scholar]
  57. Kishi, M.; Nishino, Y.; Sumiya, M.; Ohki, K.; Kimura, T.; Goto, T.; Nakai, M.; Kakinuma, M.; Ikuta, K. Cells surviving infection by human immunodeficiency virus type 1: Vif or Vpu mutants produce non-infectious or markedly less cytopathic viruses. J. Gen. Virol. 1992, 73, 77–87. [Google Scholar] [CrossRef]
  58. Bevec, D.; Dobrovnik, M.; Hauber, J.; Bohnlein, E. Inhibition of human immunodeficiency virus type 1 replication in human T cells by retroviral-mediated gene transfer of a dominant-negative Rev trans-activator. Proc. Natl. Acad. Sci. USA 1992, 89, 9870–9874. [Google Scholar] [CrossRef] [Green Version]
  59. Malim, M.H.; Freimuth, W.W.; Liu, J.; Boyle, T.J.; Lyerly, H.K.; Cullen, B.R.; Nabel, G.J. Stable expression of transdominant Rev protein in human T cells inhibits human immunodeficiency virus replication. J. Exp. Med. 1992, 176, 1197–1201. [Google Scholar] [CrossRef] [Green Version]
  60. Ulich, C.; Harrich, D.; Estes, P.; Gaynor, R.B. Inhibition of human immunodeficiency virus type 1 replication is enhanced by a combination of transdominant Tat and Rev proteins. J. Virol. 1996, 70, 4871–4876. [Google Scholar] [CrossRef] [Green Version]
  61. Harrich, D.; Ulich, C.; Garcia-Martinez, L.F.; Gaynor, R.B. Tat is required for efficient HIV-1 reverse transcription. EMBO J. 1997, 16, 1224–1235. [Google Scholar] [CrossRef] [PubMed]
  62. Apolloni, A.; Meredith, L.W.; Suhrbier, A.; Kiernan, R.; Harrich, D. The HIV-1 Tat protein stimulates reverse transcription in vitro. Curr. HIV Res. 2007, 5, 473–483. [Google Scholar] [CrossRef] [PubMed]
  63. Boudier, C.; Humbert, N.; Chaminade, F.; Chen, Y.; de Rocquigny, H.; Godet, J.; Mauffret, O.; Fossé, P.; Mély, Y. Dynamic interactions of the HIV-1 Tat with nucleic acids are critical for Tat activity in reverse transcription. Nucleic Acids Res. 2014, 42, 1065–1078. [Google Scholar] [CrossRef] [Green Version]
  64. Meredith, L.W.; Sivakumaran, H.; Major, L.; Suhrbier, A.; Harrich, D. Potent inhibition of HIV-1 replication by a Tat mutant. PLoS ONE 2009, 4, e7769. [Google Scholar] [CrossRef] [Green Version]
  65. Lin, M.H.; Apolloni, A.; Cutillas, V.; Sivakumaran, H.; Martin, S.; Li, D.; Wei, T.; Wang, R.; Jin, H.; Spann, K.; et al. A mutant Tat protein inhibits HIV-1 reverse transcription by targeting the reverse transcription complex. J. Virol. 2015, 89, 4827–4836. [Google Scholar] [CrossRef] [Green Version]
  66. Lin, M.H.; Sivakumaran, H.; Jones, A.; Li, D.; Harper, C.; Wei, T.; Jin, H.; Rustanti, L.; Meunier, F.A.; Spann, K.; et al. A HIV-1 Tat mutant protein disrupts HIV-1 Rev function by targeting the DEAD-box RNA helicase DDX1. Retrovirology 2014, 11, 121. [Google Scholar] [CrossRef]
  67. Lin, M.H.; Sivakumaran, H.; Apolloni, A.; Wei, T.; Jans, D.A.; Harrich, D. Nullbasic, a potent anti-HIV Tat mutant, induces CRM1-dependent disruption of HIV Rev trafficking. PLoS ONE 2012, 7, e51466. [Google Scholar] [CrossRef] [Green Version]
  68. Apolloni, A.; Lin, M.H.; Sivakumaran, H.; Li, D.; Kershaw, M.H.; Harrich, D. A mutant Tat protein provides strong protection from HIV-1 infection in human CD4+ T cells. Hum. Gene Ther. 2013, 24, 270–282. [Google Scholar] [CrossRef] [Green Version]
  69. Jin, H.; Li, D.; Sivakumaran, H.; Lor, M.; Rustanti, L.; Cloonan, N.; Wani, S.; Harrich, D. Shutdown of HIV-1 transcription in T cells by Nullbasic, a mutant Tat protein. MBio 2016, 7, e00518-16. [Google Scholar] [CrossRef] [Green Version]
  70. Jin, H.; Sun, Y.; Li, D.; Lin, M.-H.; Lor, M.; Rustanti, L.; Harrich, D. Strong in vivo inhibition of HIV-1 replication by Nullbasic, a Tat mutant. mBio 2019, 10, e01769-01719. [Google Scholar] [CrossRef] [Green Version]
  71. Fraldi, A.; Varrone, F.; Napolitano, G.; Michels, A.A.; Majello, B.; Bensaude, O.; Lania, L. Inhibition of Tat activity by the HEXIM1 protein. Retrovirology 2005, 2, 42. [Google Scholar] [CrossRef] [Green Version]
  72. Leoz, M.; Kukanja, P.; Luo, Z.; Huang, F.; Cary, D.C.; Peterlin, B.M.; Fujinaga, K. HEXIM1-Tat chimera inhibits HIV-1 replication. PLOS Pathogens 2018, 14, e1007402. [Google Scholar] [CrossRef]
  73. Mousseau, G.; Clementz, M.A.; Bakeman, W.N.; Nagarsheth, N.; Cameron, M.; Shi, J.; Baran, P.; Fromentin, R.; Chomont, N.; Valente, S.T. An analog of the natural steroidal alkaloid cortistatin A potently suppresses Tat-dependent HIV transcription. Cell Host Microbe 2012, 12, 97–108. [Google Scholar] [CrossRef] [Green Version]
  74. Mediouni, S.; Chinthalapudi, K.; Ekka, M.K.; Usui, I.; Jablonski, J.A.; Clementz, M.A.; Mousseau, G.; Nowak, J.; Macherla, V.R.; Beverage, J.N.; et al. Didehydro-cortistatin A inhibits HIV-1 by specifically binding to the unstructured basic region of Tat. MBio 2019, 10, e02662-18. [Google Scholar] [CrossRef] [Green Version]
  75. Li, C.; Mousseau, G.; Valente, S.T. Tat inhibition by didehydro-cortistatin A promotes heterochromatin formation at the HIV-1 long terminal repeat. Epigenetics Chromatin 2019, 12, 23. [Google Scholar] [CrossRef]
  76. Mediouni, S.; Kessing, C.F.; Jablonski, J.A.; Thenin-Houssier, S.; Clementz, M.; Kovach, M.D.; Mousseau, G.; de Vera, I.M.S.; Li, C.; Kojetin, D.J.; et al. The Tat inhibitor didehydro-cortistatin A suppresses SIV replication and reactivation. FASEB J. 2019, 33, 8280–8293. [Google Scholar] [CrossRef]
  77. Wan, Z.; Chen, X. Triptolide inhibits human immunodeficiency virus type 1 replication by promoting proteasomal degradation of Tat protein. Retrovirology 2014, 11, 88. [Google Scholar] [CrossRef]
  78. Ensoli, B.; Fiorelli, V.; Ensoli, F.; Lazzarin, A.; Visintini, R.; Narciso, P.; Di Carlo, A.; Tripiciano, A.; Longo, O.; Bellino, S.; et al. The preventive phase I trial with the HIV-1 Tat-based vaccine. Vaccine 2009, 28, 371–378. [Google Scholar] [CrossRef]
  79. Ensoli, B.; Nchabeleng, M.; Ensoli, F.; Tripiciano, A.; Bellino, S.; Picconi, O.; Sgadari, C.; Longo, O.; Tavoschi, L.; Joffe, D.; et al. HIV-Tat immunization induces cross-clade neutralizing antibodies and CD4+ T cell increases in antiretroviral-treated South African volunteers: A randomized phase II clinical trial. Retrovirology 2016, 13, 34. [Google Scholar] [CrossRef] [Green Version]
  80. Cafaro, A.; Caputo, A.; Fracasso, C.; Maggiorella, M.T.; Goletti, D.; Baroncelli, S.; Pace, M.; Sernicola, L.; Koanga-Mogtomo, M.L.; Betti, M.; et al. Control of SHIV-89.6p-infection of cynomolgus monkeys by HIV-1 Tat protein vaccine. Nat. Med. 1999, 5, 643–650. [Google Scholar] [CrossRef]
  81. Maggiorella, M.T.; Baroncelli, S.; Michelini, Z.; Fanales-Belasio, E.; Moretti, S.; Sernicola, L.; Cara, A.; Negri, D.R.; Butto, S.; Fiorelli, V.; et al. Long-term protection against SHIV89.6p replication in HIV-1 Tat vaccinated cynomolgus monkeys. Vaccine 2004, 22, 3258–3269. [Google Scholar] [CrossRef]
  82. Ensoli, F.; Cafaro, A.; Casabianca, A.; Tripiciano, A.; Bellino, S.; Longo, O.; Francavilla, V.; Picconi, O.; Sgadari, C.; Moretti, S.; et al. HIV-1 Tat immunization restores immune homeostasis and attacks the HAART-resistant blood HIV DNA: Results of a randomized phase II exploratory clinical trial. Retrovirology 2015, 12, 33. [Google Scholar] [CrossRef] [Green Version]
  83. Sgadari, C.; Monini, P.; Tripiciano, A.; Picconi, O.; Casabianca, A.; Orlandi, C.; Moretti, S.; Francavilla, V.; Arancio, A.; Paniccia, G.; et al. Continued decay of HIV proviral DNA upon vaccination with HIV-1 Tat of subjects on long-term ART: An 8-year follow-up study. Front. Immunol. 2019, 10, 233. [Google Scholar] [CrossRef]
  84. Ensoli, B.; Bellino, S.; Tripiciano, A.; Longo, O.; Francavilla, V.; Marcotullio, S.; Cafaro, A.; Picconi, O.; Paniccia, G.; Scoglio, A.; et al. Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART. PLoS ONE 2010, 5, e13540. [Google Scholar] [CrossRef] [Green Version]
  85. Opi, S.; Peloponese, J.M.; Esquieu, D.; Campbell, G.; de Mareuil, J.; Walburger, A.; Solomiac, M.; Gregoire, C.; Bouveret, E.; Yirrell, D.L.; et al. Tat HIV-1 primary and tertiary structures critical to immune response against non-homologous variants. J. Biol. Chem. 2002, 277, 35915–35919. [Google Scholar] [CrossRef] [Green Version]
  86. Loret, E.P.; Darque, A.; Jouve, E.; Loret, E.A.; Nicolino-Brunet, C.; Morange, S.; Castanier, E.; Casanova, J.; Caloustian, C.; Bornet, C.; et al. Intradermal injection of a Tat Oyi-based therapeutic HIV vaccine reduces of 1.5 log copies/mL the HIV RNA rebound median and no HIV DNA rebound following cart interruption in a phase I/II randomized controlled clinical trial. Retrovirology 2016, 13, 21. [Google Scholar] [CrossRef] [Green Version]
  87. Watkins, J.D.; Lancelot, S.; Campbell, G.R.; Esquieu, D.; de Mareuil, J.; Opi, S.; Annappa, S.; Salles, J.P.; Loret, E.P. Reservoir cells no longer detectable after a heterologous SHIV challenge with the synthetic HIV-1 Tat Oyi vaccine. Retrovirology 2006, 3, 8. [Google Scholar] [CrossRef] [Green Version]
  88. Spina, C.A.; Anderson, J.; Archin, N.M.; Bosque, A.; Chan, J.; Famiglietti, M.; Greene, W.C.; Kashuba, A.; Lewin, S.R.; Margolis, D.M.; et al. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog. 2013, 9, e1003834. [Google Scholar] [CrossRef] [Green Version]
  89. Banerjee, C.; Archin, N.; Michaels, D.; Belkina, A.C.; Denis, G.V.; Bradner, J.; Sebastiani, P.; Margolis, D.M.; Montano, M. BET bromodomain inhibition as a novel strategy for reactivation of HIV-1. J. Leukoc Biol. 2012, 92, 1147–1154. [Google Scholar] [CrossRef]
  90. Ylisastigui, L.; Archin, N.M.; Lehrman, G.; Bosch, R.J.; Margolis, D.M. Coaxing HIV-1 from resting CD4 T cells: Histone deacetylase inhibition allows latent viral expression. AIDS 2004, 18, 1101–1108. [Google Scholar] [CrossRef]
  91. Archin, N.M.; Espeseth, A.; Parker, D.; Cheema, M.; Hazuda, D.; Margolis, D.M. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res. Hum. Retroviruses 2009, 25, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Contreras, X.; Schweneker, M.; Chen, C.S.; McCune, J.M.; Deeks, S.G.; Martin, J.; Peterlin, B.M. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J. Biol. Chem. 2009, 284, 6782–6789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Archin, N.M.; Liberty, A.L.; Kashuba, A.D.; Choudhary, S.K.; Kuruc, J.D.; Crooks, A.M.; Parker, D.C.; Anderson, E.M.; Kearney, M.F.; Strain, M.C.; et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 2012, 487, 482–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rustanti, L.; Jin, H.; Lor, M.; Lin, M.H.; Rawle, D.J.; Harrich, D. A mutant Tat protein inhibits infection of human cells by strains from diverse HIV-1 subtypes. Virol. J. 2017, 14, 52. [Google Scholar] [CrossRef] [PubMed]
  95. AJ, C.Q.; Bugai, A.; Barboric, M. Cracking the control of RNA polymerase II elongation by 7SK snRNP and P-TEFb. Nucleic Acids Res. 2016, 44, 7527–7539. [Google Scholar]
  96. Yik, J.H.; Chen, R.; Pezda, A.C.; Samford, C.S.; Zhou, Q. A human immunodeficiency virus type 1 Tat-like arginine-rich RNA-binding domain is essential for HEXIM1 to inhibit RNA polymerase II transcription through 7SK snRNA-mediated inactivation of P-TEFb. Mol. Cell Biol. 2004, 24, 5094–5105. [Google Scholar] [CrossRef] [Green Version]
  97. Michels, A.A.; Fraldi, A.; Li, Q.; Adamson, T.E.; Bonnet, F.; Nguyen, V.T.; Sedore, S.C.; Price, J.P.; Price, D.H.; Lania, L.; et al. Binding of the 7SK snRNA turns the HEXIM1 protein into a P-TEFb (CDK9/cyclin T) inhibitor. EMBO J. 2004, 23, 2608–2619. [Google Scholar] [CrossRef] [Green Version]
  98. Yik, J.H.; Chen, R.; Nishimura, R.; Jennings, J.L.; Link, A.J.; Zhou, Q. Inhibition of P-TEFb (CDK9/cyclin T) kinase and RNA polymerase II transcription by the coordinated actions of HEXIM1 and 7SK snRNA. Mol. Cell 2003, 12, 971–982. [Google Scholar] [CrossRef]
  99. Tahirov, T.H.; Babayeva, N.D.; Varzavand, K.; Cooper, J.J.; Sedore, S.C.; Price, D.H. Crystal structure of HIV-1 Tat complexed with human P-TEFb. Nature 2010, 465, 747–751. [Google Scholar] [CrossRef]
  100. Shi, J.; Manolikakes, G.; Yeh, C.H.; Guerrero, C.A.; Shenvi, R.A.; Shigehisa, H.; Baran, P.S. Scalable synthesis of cortistatin A and related structures. J. Am. Chem. Soc. 2011, 133, 8014–8027. [Google Scholar] [CrossRef] [Green Version]
  101. Pelish, H.E.; Liau, B.B.; Nitulescu, I.I.; Tangpeerachaikul, A.; Poss, Z.C.; Da Silva, D.H.; Caruso, B.T.; Arefolov, A.; Fadeyi, O.; Christie, A.L.; et al. Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature 2015, 526, 273–276. [Google Scholar] [CrossRef] [PubMed]
  102. Cee, V.J.; Chen, D.Y.; Lee, M.R.; Nicolaou, K.C. Cortistatin A is a high-affinity ligand of protein kinases ROCK, CDK8, and CDK11. Angew. Chem. Int. Ed. Engl. 2009, 48, 8952–8957. [Google Scholar] [CrossRef] [PubMed]
  103. Cary, D.C.; Rheinberger, M.; Rojc, A.; Peterlin, B.M. HIV transcription is independent of mediator kinases. AIDS Res. Hum. Retroviruses 2019, 35, 710–717. [Google Scholar] [CrossRef] [PubMed]
  104. Mediouni, S.; Jablonski, J.; Paris, J.J.; Clementz, M.A.; Thenin-Houssier, S.; McLaughlin, J.P.; Valente, S.T. Didehydro-cortistatin A inhibits HIV-1 Tat mediated neuroinflammation and prevents potentiation of cocaine reward in Tat transgenic mice. Curr. HIV Res. 2015, 13, 64–79. [Google Scholar] [CrossRef] [Green Version]
  105. Turner, A.W.; Margolis, D.M. Chromatin regulation and the histone code in HIV latency. Yale J. Biol. Med. 2017, 90, 229–243. [Google Scholar]
  106. Easley, R.; Carpio, L.; Dannenberg, L.; Choi, S.; Alani, D.; Van Duyne, R.; Guendel, I.; Klase, Z.; Agbottah, E.; Kehn-Hall, K.; et al. Transcription through the HIV-1 nucleosomes: Effects of the PBAF complex in Tat activated transcription. Virology 2010, 405, 322–333. [Google Scholar] [CrossRef] [Green Version]
  107. Kessing, C.F.; Nixon, C.C.; Li, C.; Tsai, P.; Takata, H.; Mousseau, G.; Ho, P.T.; Honeycutt, J.B.; Fallahi, M.; Trautmann, L.; et al. In vivo suppression of HIV rebound by didehydro-cortistatin A, a “block-and-lock” strategy for HIV-1 treatment. Cell Rep. 2017, 21, 600–611. [Google Scholar] [CrossRef] [Green Version]
  108. Rice, A.P. Unexpected mutations in HIV-1 that confer resistance to the Tat inhibitor didehydro-cortistatin A. MBio 2019, 10, e01547-19. [Google Scholar] [CrossRef] [Green Version]
  109. Mousseau, G.; Aneja, R.; Clementz, M.A.; Mediouni, S.; Lima, N.S.; Haregot, A.; Kessing, C.F.; Jablonski, J.A.; Thenin-Houssier, S.; Nagarsheth, N.; et al. Resistance to the Tat inhibitor didehydro-cortistatin A is mediated by heightened basal HIV-1 transcription. MBio 2019, 10, e01750-18. [Google Scholar] [CrossRef] [Green Version]
  110. Bao, J.; Dai, S.-M. A chinese herb Tripterygium wilfordii hook F in the treatment of rheumatoid arthritis: Mechanism, efficacy, and safety. Rheumatol. Int. 2011, 31, 1123–1129. [Google Scholar] [CrossRef]
  111. Tao, X.; Younger, J.; Fan, F.Z.; Wang, B.; Lipsky, P.E. Benefit of an extract of Tripterygium wilfordii hook F in patients with rheumatoid arthritis: A double-blind, placebo-controlled study. Arthritis Rheum 2002, 46, 1735–1743. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, S.-R.; Dai, Y.; Zhao, J.; Lin, L.; Wang, Y.; Wang, Y. A mechanistic overview of triptolide and celastrol, natural products from Tripterygium wilfordii hook F. Front. Pharmacol. 2018, 9, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kim, S.T.; Kim, S.Y.; Lee, J.; Kim, K.; Park, S.H.; Park, Y.S.; Lim, H.Y.; Kang, W.K.; Park, J.O. Triptolide as a novel agent in pancreatic cancer: The validation using patient derived pancreatic tumor cell line. BMC Cancer 2018, 18, 1103. [Google Scholar] [CrossRef] [PubMed]
  114. Ziaei, S.; Halaby, R. Immunosuppressive, anti-inflammatory and anti-cancer properties of triptolide: A mini review. Avicenna J. Phytomed. 2016, 6, 149–164. [Google Scholar]
  115. Qiu, D.; Zhao, G.; Aoki, Y.; Shi, L.; Uyei, A.; Nazarian, S.; Ng, J.C.H.; Kao, P.N. Immunosuppressant PG490 (triptolide) inhibits T-cell interleukin-2 expression at the level of purine-box/nuclear factor of activated T-cells and NF-κB transcriptional activation. J. Biol. Chem. 1999, 274, 13443–13450. [Google Scholar] [CrossRef] [Green Version]
  116. Vispé, S.; DeVries, L.; Créancier, L.; Besse, J.; Bréand, S.; Hobson, D.J.; Svejstrup, J.Q.; Annereau, J.-P.; Cussac, D.; Dumontet, C.; et al. Triptolide is an inhibitor of RNA polymerase I and II–dependent transcription leading predominantly to down-regulation of short-lived mRNA. Mol. Cancer Ther. 2009, 8, 2780. [Google Scholar]
  117. Wang, Y.; Lu, J.-j.; He, L.; Yu, Q. Triptolide (TPL) inhibits global transcription by inducing proteasome-dependent degradation of RNA polymerase II (Pol II). PLoS ONE 2011, 6, e23993. [Google Scholar] [CrossRef] [Green Version]
  118. Chen, F.X.; Smith, E.R.; Shilatifard, A. Born to run: Control of transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 2018, 19, 464–478. [Google Scholar] [CrossRef]
  119. Liang, X.; Xie, R.; Su, J.; Ye, B.; Wei, S.; Liang, Z.; Bai, R.; Chen, Z.; Li, Z.; Gao, X. Inhibition of RNA polymerase III transcription by triptolide attenuates colorectal tumorigenesis. J. Exp. Clin. Cancer Res. 2019, 38, 217. [Google Scholar] [CrossRef]
  120. Titov, D.V.; Gilman, B.; He, Q.-L.; Bhat, S.; Low, W.-K.; Dang, Y.; Smeaton, M.; Demain, A.L.; Miller, P.S.; Kugel, J.F.; et al. XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nat. Chem. Biol. 2011, 7, 182–188. [Google Scholar] [CrossRef] [Green Version]
  121. Chen, F.; Gao, X.; Shilatifard, A. Stably paused genes revealed through inhibition of transcription initiation by the TFIIH inhibitor triptolide. Genes Dev. 2015, 29, 39–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Derdeyn, C.A.; Decker, J.M.; Sfakianos, J.N.; Wu, X.; O’Brien, W.A.; Ratner, L.; Kappes, J.C.; Shaw, G.M.; Hunter, E. Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is modulated by coreceptor specificity defined by the V3 loop of gp120. J. Virol. 2000, 74, 8358–8367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Platt, E.J.; Wehrly, K.; Kuhmann, S.E.; Chesebro, B.; Kabat, D. Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J. Virol. 1998, 72, 2855–2864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wei, X.; Decker, J.M.; Liu, H.; Zhang, Z.; Arani, R.B.; Kilby, J.M.; Saag, M.S.; Wu, X.; Shaw, G.M.; Kappes, J.C. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother. 2002, 46, 1896–1905. [Google Scholar] [CrossRef] [Green Version]
  125. Sivakumaran, H.; Lin, M.-H.; Apolloni, A.; Cutillas, V.; Jin, H.; Li, D.; Wei, T.; Harrich, D. Overexpression of PRMT6 does not suppress HIV-1 Tat transactivation in cells naturally lacking PRMT6. Virol. J. 2013, 10, 207. [Google Scholar] [CrossRef] [Green Version]
  126. Fan, D.; Guo, Q.; Shen, J.; Zheng, K.; Lu, C.; Zhang, G.; Lu, A.; He, X. The effect of triptolide in rheumatoid arthritis: From basic research towards clinical translation. Int. J. Mol. Sci. 2018, 19, 376. [Google Scholar] [CrossRef] [Green Version]
  127. Goldstein, G. HIV-1 Tat protein as a potential AIDS vaccine. Nat. Med. 1996, 2, 960–964. [Google Scholar] [CrossRef]
  128. Singh, A.; Weinberger, L.S. Stochastic gene expression as a molecular switch for viral latency. Curr. Opin. Microbiol. 2009, 12, 460–466. [Google Scholar] [CrossRef] [Green Version]
  129. Ensoli, B.; Barillari, G.; Salahuddin, S.Z.; Gallo, R.C.; Wong-Staal, F. Tat protein of HIV-1 stimulates growth of cells derived from Kaposi’s sarcoma lesions of AIDS patients. Nature 1990, 345, 84–86. [Google Scholar] [CrossRef]
  130. Westendorp, M.O.; Frank, R.; Ochsenbauer, C.; Stricker, K.; Dhein, J.; Walczak, H.; Debatin, K.M.; Krammer, P.H. Sensitization of T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature 1995, 375, 497–500. [Google Scholar] [CrossRef]
  131. Xiao, H.; Neuveut, C.; Tiffany, H.L.; Benkirane, M.; Rich, E.A.; Murphy, P.M.; Jeang, K.T. Selective CXCR4 antagonism by Tat: Implications for in vivo expansion of coreceptor use by HIV-1. Proc. Natl. Acad. Sci. USA 2000, 97, 11466–11471. [Google Scholar] [CrossRef] [Green Version]
  132. Rayne, F.; Debaisieux, S.; Bonhoure, A.; Beaumelle, B. HIV-1 Tat is unconventionally secreted through the plasma membrane. Cell Biol. Int. 2010, 34, 409–413. [Google Scholar] [CrossRef]
  133. Mele, A.R.; Marino, J.; Chen, K.; Pirrone, V.; Janetopoulos, C.; Wigdahl, B.; Klase, Z.; Nonnemacher, M.R. Defining the molecular mechanisms of HIV-1 Tat secretion: Ptdins(4,5)p2 at the epicenter. Traffic 2018, 19, 655–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Li, C.J.; Ueda, Y.; Shi, B.; Borodyansky, L.; Huang, L.; Li, Y.Z.; Pardee, A.B. Tat protein induces self-perpetuating permissivity for productive HIV-1 infection. Proc. Natl. Acad. Sci. USA 1997, 94, 8116–8120. [Google Scholar] [CrossRef] [Green Version]
  135. Li, C.J.; Friedman, D.J.; Wang, C.; Metelev, V.; Pardee, A.B. Induction of apoptosis in uninfected lymphocytes by HIV-1 Tat protein. Science 1995, 268, 429–431. [Google Scholar] [CrossRef] [PubMed]
  136. Howcroft, T.K.; Strebel, K.; Martin, M.A.; Singer, D.S. Repression of MHC class I gene promoter activity by two-exon tat of HIV. Science 1993, 260, 1320–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Banks, W.A.; Robinson, S.M.; Nath, A. Permeability of the blood-brain barrier to HIV-1 Tat. Exp. Neurol. 2005, 193, 218–227. [Google Scholar] [CrossRef]
  138. Price, T.O.; Ercal, N.; Nakaoke, R.; Banks, W.A. HIV-1 viral proteins gp120 and Tat induce oxidative stress in brain endothelial cells. Brain Res. 2005, 1045, 57–63. [Google Scholar] [CrossRef]
  139. Rojas-Celis, V.; Valiente-Echeverria, F.; Soto-Rifo, R.; Toro-Ascuy, D. New challenges of HIV-1 infection: How HIV-1 attacks and resides in the central nervous system. Cells 2019, 8, 1245. [Google Scholar] [CrossRef] [Green Version]
  140. Brake, D.A.; Goudsmit, J.; Krone, W.J.; Schammel, P.; Appleby, N.; Meloen, R.H.; Debouck, C. Characterization of murine monoclonal antibodies to the Tat protein from human immunodeficiency virus type 1. J. Virol. 1990, 64, 962–965. [Google Scholar] [CrossRef] [Green Version]
  141. Krone, W.J.; Debouck, C.; Epstein, L.G.; Heutink, P.; Meloen, R.; Goudsmit, J. Natural antibodies to HIV-Tat epitopes and expression of HIV-1 genes in vivo. J. Med. Virol. 1988, 26, 261–270. [Google Scholar] [CrossRef]
  142. Reiss, P.; Lange, J.M.; de Ronde, A.; de Wolf, F.; Dekker, J.; Debouck, C.; Goudsmit, J. Speed of progression to AIDS and degree of antibody response to accessory gene products of HIV-1. J. Med. Virol. 1990, 30, 163–168. [Google Scholar] [CrossRef]
  143. Re, M.C.; Furlini, G.; Vignoli, M.; Ramazzotti, E.; Roderigo, G.; De Rosa, V.; Zauli, G.; Lolli, S.; Capitani, S.; La Placa, M. Effect of antibody to HIV-1 Tat protein on viral replication in vitro and progression of HIV-1 disease in vivo. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 1995, 10, 408–416. [Google Scholar] [CrossRef]
  144. Zagury, J.F.; Sill, A.; Blattner, W.; Lachgar, A.; Le Buanec, H.; Richardson, M.; Rappaport, J.; Hendel, H.; Bizzini, B.; Gringeri, A.; et al. Antibodies to the HIV-1 Tat protein correlated with nonprogression to AIDS: A rationale for the use of Tat toxoid as an HIV-1 vaccine. J. Hum. Virol. 1998, 1, 282–292. [Google Scholar]
  145. Re, M.C.; Vignoli, M.; Furlini, G.; Gibellini, D.; Colangeli, V.; Vitone, F.; La Placa, M. Antibodies against full-length Tat protein and some low-molecular-weight Tat-peptides correlate with low or undetectable viral load in HIV-1 seropositive patients. J. Clin. Virol. 2001, 21, 81–89. [Google Scholar] [CrossRef]
  146. Robertson, D.L.; Anderson, J.P.; Bradac, J.A.; Carr, J.K.; Foley, B.; Funkhouser, R.K.; Gao, F.; Hahn, B.H.; Kalish, M.L.; Kuiken, C.; et al. HIV-1 nomenclature proposal. Science 2000, 288, 55–56. [Google Scholar] [CrossRef]
  147. Ratner, L.; Haseltine, W.; Patarca, R.; Livak, K.J.; Starcich, B.; Josephs, S.F.; Doran, E.R.; Rafalski, J.A.; Whitehorn, E.A.; Baumeister, K.; et al. Complete nucleotide sequence of the AIDS virus, HTLV-III. Nature 1985, 313, 277–284. [Google Scholar] [CrossRef]
  148. Huet, T.; Dazza, M.C.; Brun-Vezinet, F.; Roelants, G.E.; Wain-Hobson, S. A highly defective HIV-1 strain isolated from a healthy Gabonese individual presenting an atypical western blot. AIDS 1989, 3, 707–715. [Google Scholar] [CrossRef]
  149. Garcia, J.A.; Harrich, D.; Pearson, L.; Mitsuyasu, R.; Gaynor, R.B. Functional domains required for Tat-induced transcriptional activation of the HIV-1 long terminal repeat. EMBO J. 1988, 7, 3143–3147. [Google Scholar] [CrossRef]
  150. Foley, B.; Leitner, T.; Apetrei, C.; Hahn, B.; Mizrachi, I.; Mullins, J.; Rambaut, A.; Wolinsky, S.; Korber, B. HIV Sequence Compendium 2018; Theoretical Biology and Biophysics Group, Los Alamos National Laboratory: LA-UR, NM, USA, 2018; Volume 18, p. 25673. [Google Scholar]
  151. Pauza, C.D.; Trivedi, P.; Wallace, M.; Ruckwardt, T.J.; Le Buanec, H.; Lu, W.; Bizzini, B.; Burny, A.; Zagury, D.; Gallo, R.C. Vaccination with Tat toxoid attenuates disease in simian/HIV-challenged macaques. Proc. Natl. Acad. Sci. USA 2000, 97, 3515–3519. [Google Scholar] [CrossRef]
  152. Stittelaar, K.J.; Gruters, R.A.; Schutten, M.; van Baalen, C.A.; van Amerongen, G.; Cranage, M.; Liljestrom, P.; Sutter, G.; Osterhaus, A.D. Comparison of the efficacy of early versus late viral proteins in vaccination against SIV. Vaccine 2002, 20, 2921–2927. [Google Scholar] [CrossRef] [Green Version]
  153. Liang, X.; Casimiro, D.R.; Schleif, W.A.; Wang, F.; Davies, M.E.; Zhang, Z.Q.; Fu, T.M.; Finnefrock, A.C.; Handt, L.; Citron, M.P.; et al. Vectored Gag and Env but not Tat show efficacy against simian-human immunodeficiency virus 89.6p challenge in mamu-a*01-negative rhesus monkeys. J. Virol. 2005, 79, 12321–12331. [Google Scholar] [CrossRef] [Green Version]
  154. Cafaro, A.; Tripiciano, A.; Picconi, O.; Sgadari, C.; Moretti, S.; Buttò, S.; Monini, P.; Ensoli, B. Anti-Tat immunity in HIV-1 infection: Effects of naturally occurring and vaccine-induced antibodies against Tat on the course of the disease. Vaccines 2019, 7, 99. [Google Scholar] [CrossRef] [Green Version]
  155. Bellino, S.; Tripiciano, A.; Picconi, O.; Francavilla, V.; Longo, O.; Sgadari, C.; Paniccia, G.; Arancio, A.; Angarano, G.; Ladisa, N.; et al. The presence of anti-Tat antibodies in HIV-infected individuals is associated with containment of CD4+ T-cell decay and viral load, and with delay of disease progression: Results of a 3-year cohort study. Retrovirology 2014, 11, 49. [Google Scholar] [CrossRef] [Green Version]
  156. Demirhan, I.; Chandra, A.; Mueller, F.; Mueller, H.; Biberfeld, P.; Hasselmayer, O.; Chandra, P. Antibody spectrum against the viral transactivator protein in patients with human immunodeficiency virus type 1 infection and Kaposi’s sarcoma. J. Hum. Virol. 2000, 3, 137–143. [Google Scholar]
  157. Chen, Q.; Li, L.; Liao, W.; Zhang, H.; Wang, J.; Sheng, B.; Zhang, H.; Huang, X.; Ding, Y.; Zhang, T.; et al. Characterization of Tat antibody responses in Chinese individuals infected with HIV-1. PLoS ONE 2013, 8, e60825. [Google Scholar] [CrossRef] [Green Version]
  158. Butto, S.; Fiorelli, V.; Tripiciano, A.; Ruiz-Alvarez, M.J.; Scoglio, A.; Ensoli, F.; Ciccozzi, M.; Collacchi, B.; Sabbatucci, M.; Cafaro, A.; et al. Sequence conservation and antibody cross-recognition of clade B human immunodeficiency virus (HIV) type 1 Tat protein in HIV-1-infected italians, ugandans, and south africans. J. Infect. Dis. 2003, 188, 1171–1180. [Google Scholar] [CrossRef]
  159. Rezza, G.; Fiorelli, V.; Dorrucci, M.; Ciccozzi, M.; Tripiciano, A.; Scoglio, A.; Collacchi, B.; Ruiz-Alvarez, M.; Giannetto, C.; Caputo, A.; et al. The presence of anti-Tat antibodies is predictive of long-term nonprogression to AIDS or severe immunodeficiency: Findings in a cohort of HIV-1 seroconverters. J. Infect. Dis 2005, 191, 1321–1324. [Google Scholar] [CrossRef] [Green Version]
  160. Viglianti, G.A.; Mullins, J.I. Functional comparison of transactivation by simian immunodeficiency virus from rhesus macaques and human immunodeficiency virus type 1. J. Virol. 1988, 62, 4523–4532. [Google Scholar] [CrossRef] [Green Version]
  161. Kamori, D.; Ueno, T. HIV-1 Tat and viral latency: What we can learn from naturally occurring sequence variations. Front. Microbiol. 2017, 8, 80. [Google Scholar] [CrossRef] [Green Version]
  162. Rodman, T.C.; To, S.E.; Hashish, H.; Manchester, K. Epitopes for natural antibodies of human immunodeficiency virus (HIV)-negative (normal) and HIV-positive sera are coincident with two key functional sequences of HIV Tat protein. Proc. Natl. Acad. Sci. USA 1993, 90, 7719. [Google Scholar] [CrossRef] [Green Version]
  163. Shojania, S.; O’Neil J, D. HIV-1 Tat is a natively unfolded protein: The solution conformation and dynamics of reduced HIV-1 Tat1-72 by NMR spectroscopy. J. Biol. Chem. 2006, 281, 8347–8356. [Google Scholar] [CrossRef] [Green Version]
  164. Riddell, J.T.; Amico, K.R.; Mayer, K.H. HIV pre-exposure prophylaxis: A review. JAMA 2018, 319, 1261–1268. [Google Scholar] [CrossRef]
  165. Hong, F.F.; Mellors, J.W. Changes in HIV reservoirs during long-term antiretroviral therapy. Curr. Opin. HIV AIDS 2015, 10, 43–48. [Google Scholar] [CrossRef] [Green Version]
  166. Ghose, R.; Liou, L.Y.; Herrmann, C.H.; Rice, A.P. Induction of TAK (cyclin T1/P-TEFb) in purified resting CD4(+) T lymphocytes by combination of cytokines. J. Virol. 2001, 75, 11336–11343. [Google Scholar] [CrossRef] [Green Version]
  167. Sung, T.L.; Rice, A.P. Effects of prostratin on cyclin T1/P-TEFb function and the gene expression profile in primary resting CD4+ T cells. Retrovirology 2006, 3, 66. [Google Scholar] [CrossRef] [Green Version]
  168. Morgan, R.A.; Walker, R.; Carter, C.S.; Natarajan, V.; Tavel, J.A.; Bechtel, C.; Herpin, B.; Muul, L.; Zheng, Z.; Jagannatha, S.; et al. Preferential survival of CD4+ T lymphocytes engineered with anti-human immunodeficiency virus (HIV) genes in HIV-infected individuals. Hum. Gene Ther. 2005, 16, 1065–1074. [Google Scholar] [CrossRef]
Figure 1. (a) A schematic of Tat domains including acidic, cysteine (cys)-rich, core, basic (also called arginine rich), glutamine (glu)-rich and the second exon. (b) A model of transactivation where Tat recruits P-TEFb from the 7SK snRNP complex, where 7SK-RNA, LARP7, and MePCE make the core 7SK snRNP, which can bind to and inhibit CDK9 activity of P-TEFb by HEXIM. Tat recruits P-TEFb from sources that include the 7SK snRNP, P-TEFb-BRD4, and P-TEFb-SEC complexes. The Tat:P-TEFb complex binds to TAR RNA, where CDK9 phosphorylates NELF (leads to its release) and DSIF, as well as hyperphosphorylating the C-terminal domain (CTD) of RNAP II, which then undertakes efficient synthesis and elongation of full length viral mRNA transcripts.
Figure 1. (a) A schematic of Tat domains including acidic, cysteine (cys)-rich, core, basic (also called arginine rich), glutamine (glu)-rich and the second exon. (b) A model of transactivation where Tat recruits P-TEFb from the 7SK snRNP complex, where 7SK-RNA, LARP7, and MePCE make the core 7SK snRNP, which can bind to and inhibit CDK9 activity of P-TEFb by HEXIM. Tat recruits P-TEFb from sources that include the 7SK snRNP, P-TEFb-BRD4, and P-TEFb-SEC complexes. The Tat:P-TEFb complex binds to TAR RNA, where CDK9 phosphorylates NELF (leads to its release) and DSIF, as well as hyperphosphorylating the C-terminal domain (CTD) of RNAP II, which then undertakes efficient synthesis and elongation of full length viral mRNA transcripts.
Viruses 12 00415 g001
Figure 2. (a) Amino acid alignments for showing a consensus sequence for subtypes B and C [150], compared to the sequences of Tat-Oyi and Tat-BH10. Residues found in the subtype B consensus sequence (purple) and subtype C consensus (red) are indicated in the sequences of Tat-Oyi and Tat-BH10. Residues found in the consensus sequences of Tat-Oyi (green) and Tat-BH10 (blue) are also shown. The domains of Tat are demarcated.
Figure 2. (a) Amino acid alignments for showing a consensus sequence for subtypes B and C [150], compared to the sequences of Tat-Oyi and Tat-BH10. Residues found in the subtype B consensus sequence (purple) and subtype C consensus (red) are indicated in the sequences of Tat-Oyi and Tat-BH10. Residues found in the consensus sequences of Tat-Oyi (green) and Tat-BH10 (blue) are also shown. The domains of Tat are demarcated.
Viruses 12 00415 g002
Table 1. Anti-Tat agent summary.
Table 1. Anti-Tat agent summary.
Anti-Tat AgentTypeInhibits; Proposed Mechanism(s)Reference(s)
NullbasicMutant Tat protein1.Tat transactivation; binds to P-TEFb[64,68,69,70]
2. Reverse transcription; binds to reverse transcriptase[64,65]
3. Rev; binds to DDX1[64,66,67]
HT1HEXIM1-Tat fusionTat transactivation; binds to P-TEFb and inactivates CDK9[71,72]
dCASmall compound1. Tat transactivation; binds Tat basic domain and prevents interaction with TAR RNA[46,73,74]
2. HIV transcription levels; heterochromatin formation on HIV LTR promoter[75,76]
TriptolideSmall compound1. Decreases Tat steady state levels; mechanism unclear[77]
Tat-BH10Vaccine1. Tat; Tat neutralizing antibodies[78,79]
2. Virus production; anti-Tat cellular responses[78,80,81]
3. Provirus DNA load in blood lymphocytes; anti-Tat humoral/cellular responses[82,83]
4. Reduced immune activation; possibly due to reduced viral burden[82,84]
Tat-OyiVaccine1. Tat; Tat neutralizing antibodies[85,86]
2. Virus production, mechanism unclear[86]
3. Viral rebound after treatment interruption; mechanism unclear [86]
3. Provirus DNA; mechanism unclear[86,87]

Share and Cite

MDPI and ACS Style

Jin, H.; Li, D.; Lin, M.-H.; Li, L.; Harrich, D. Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure. Viruses 2020, 12, 415. https://doi.org/10.3390/v12040415

AMA Style

Jin H, Li D, Lin M-H, Li L, Harrich D. Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure. Viruses. 2020; 12(4):415. https://doi.org/10.3390/v12040415

Chicago/Turabian Style

Jin, Hongping, Dongsheng Li, Min-Hsuan Lin, Li Li, and David Harrich. 2020. "Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure" Viruses 12, no. 4: 415. https://doi.org/10.3390/v12040415

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop