Abstract
The human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (Tat) protein functions both intracellularly and extracellularly. Intracellularly the main function is to enhance transcription of the viral promoter. However, this process only requires a small amount of intracellular Tat. The majority of Tat is secreted through an unconventional mechanism by binding to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2), a phospholipid in the inner leaflet of the plasma membrane that is required for secretion. This interaction is mediated by the basic domain of Tat (residues 48–57) and a conserved tryptophan (residue 11). After binding to PtdIns(4,5)P2, Tat secretion diverges into multiple pathways, which we categorized as: oligomerization-mediated pore formation, spontaneous translocation, and incorporation into exosomes. Extracellular Tat has been shown to be neurotoxic and toxic to other cells of the CNS and periphery, able to recruit immune cells to the CNS and cerebrospinal fluid, and alter the gene expression and morphology of uninfected cells. The effects of extracellular Tat have been examined in HIV-1-associated neurocognitive disorders (HAND), however, only a small number of studies have focused on the mechanisms underlying Tat secretion. In this review, the molecular mechanisms of Tat secretion will be examined in a variety of biologically relevant cell types.
Keywords: HIV-1; Tat; PtdIns(4,5)P2; secretion; exosome; T-cell; monocyte-macrophage; astrocyte
Graphical Abstract
After the initial binding of residue 11 (light grey), a conserved tryptophan, inserts into the plasma membrane and stabilizes the interaction with PtdIns(4,5)P2. The specific region of PtdIns(4,5)P2 involved in this interaction is unknown. 3. After the interaction occurs, Tat is translocated extracellularly, but the exact process has yet to be elucidated. C) The process by which Tat is incorporated into exosomes is poorly understood. 1. Tat may bind to PtdIns(4,5)P2 at the exosomal membrane, such as during the formation of multiple vesicular bodies. 2. During invagination of the exosome, Tat may interact with HIV-1 TAR (red) and be incorporated at a higher rate. 3. During invagination of the exosome, Tat may interact with host proteins (yellow) and be incorporated at a higher rate. (INT, Intracellular; EXT, Extracellular; EXO, Exosome)

Functional properties of Tat
Human immunodeficiency virus type 1 (HIV-1) encodes for the transactivator of transcription (Tat) protein. Tat is a small, basic, non-structured protein (86 or 101 amino acid residues in length) that is responsible for enhancing the transcription of HIV-1 by recruiting the host positive transcription elongation factor b (P-TEFb) to the RNA stem-loop transactivation response (TAR) element, encoded by the viral long terminal repeat (LTR) 1,2. This, however, is not the only function of Tat, and many of its biological activities are related to the fact that the predominant proportion of Tat is transported extracellularly 3. Extracellular Tat has been shown to be neurotoxic and recruit additional immune cells to the central nervous system (CNS), causing low levels of chronic inflammation by activating bystander cells, such as endothelial cells, and causing the release of pro-inflammatory cytokines from immune cells, such as IL-1β from monocytes and macrophages 4–8. Additionally, Tat produced in the periphery can also traffic to the CNS and cross the blood-brain barrier 9. These, among other factors (including other HIV proteins such as gp120, Vpr, and Nef), contribute to the development of HIV-1-associated neurocognitive disorders (HAND)10. Furthermore, the mortality associated with HIV-1 has decreased since the development of antiretroviral therapy (ART), but there has been an increase in the prevalence of HAND and even patients that are adherent to ART are still at risk 11. While a comprehensive mechanism of HAND has yet to be elucidated, the secretion of viral proteins, such as gp120, Vpr, and Tat, have been demonstrated to alter behavior in vivo 12 and has been suggested to be crucial for productive viral infectivity 13. ART does not block the secretion of Tat and there is currently no approved therapy targeting Tat 14. Additionally, it is unknown which cells are responsible for releasing Tat and the process by which Tat is secreted within the CNS remains poorly understood.
The secretion of Tat is characterized as unconventional, meaning it does not traffic through the endoplasmic reticulum (ER) or Golgi apparatus. The first report that examined the source of extracellular Tat originated in 1990, when it was demonstrated that Tat was released from infected H9 and COS-1 cells in the absence of lysis 15. Disruption of microtubules, actin cytoskeleton, Na+/K+-ATPase activity, endosomal-formation, and Golgi apparatus integrity in CD4+ T cells did not significantly alter the secretion of Tat 5. Further investigations have led to the current understanding of the unconventional secretion of Tat, which is initiated by binding to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2), a phospholipid component of the inner leaflet of the plasma membrane3. The reported abundance of PtdIns(4,5)P2 varies 16–19, but is approximately 5% of the total phosphoinositides in a cell 20, however, it is only approximately 1% of the phospholipids in the inner leaflet of the plasma membrane 21. PtdIns(4,5)P2 can be synthesized by a few pathways, such as by phosphorylation of phosphatidylinositol 4-phosphate by phosphatidylinositol 4-phosphate 5-kinase (PIP5K) or de-phosphorylation of phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) by phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase 22. The major synthetic pathway responsible for production of PtdIns(4,5)P2 is generally accepted to be mediated by PIP5K. PtdIns(4,5)P2 is also responsible for a number of cellular functions including: regulation of cytoskeleton dynamics, endocytosis and exocytosis, and G protein-coupled receptor activation, where it is a substrate for phospholipase C and plays a critical role in the activation of protein kinase C and Ca2+ release 23.
The secretion mechanism of HIV-1 Tat has been frequently compared to fibroblast growth factor 2 (FGF2), a host cytokine that also binds to PtdIns(4,5)P2 and is translocated extracellularly 24. Furthermore, the mechanism of FGF2 secretion has been well characterized 25,26. The mechanism of FGF2 secretion is generally accepted to be mediated by oligomerization and formation of a pore in the plasma membrane 27,28. Other host proteins, such as IL-1β and galectins, are also unconventionally secreted, however, they are less well characterized compared to FGF2 26,29. The interaction between FGF2 and caspase-1 has been shown to be required for efficient secretion of both IL-1β and FGF2 30. Additionally, galectins and FGF2 have been detected within exosomes 31. If overlap between the secretion of host proteins exist, it can be rationalized that similarities exist for viral proteins as well.
Proteins that travel through the classical secretion pathway contain a stretch of 5–10 hydrophobic amino acids near the N-terminus, which mediates the initial transfer through the ER/Golgi apparatus 32. In this pathway, the protein will undergo folding, glycosylation, and insertion into COPII-coated vesicles, before reaching the plasma membrane and being secreted from the cell. Circumventing the ER/Golgi apparatus is essential for certain proteins because it allows for them to avoid modifications that would limit their biologic function 28,32. This was demonstrated by experimentally forcing FGF2 through the ER/Golgi apparatus, by synthesizing a fusion protein of FGF2 and fibroblast growth factor 4 (FGF4) 33. FGF4 naturally traffics through the ER/Golgi apparatus. The N-terminus signal peptide portion, derived from FGF4, caused the entire fusion protein to traffic through the ER/Golgi apparatus, which was then post-translationally modified by O-linked chondroitin sulfates and was secreted. This modification prevented interactions with the cell surface, which resulted in an accumulation of the secreted fusion protein in the supernatant 28,32,33. Tat could potentially prevent modifications that limit its biological activity by alteration of the corresponding acceptor sequences. This, however, is not likely to occur, as several modification sites reside within the basic domain, such as methylation by PRMT6 and acetylation by p300 and GCN5 34–37. Additionally, not all modifications are detrimental, acetylation of Tat via p300 has been demonstrated to increase transactivation of the HIV-1 LTR 37. However, alteration of the residues within the basic domain would more likely impact the secretion and extracellular functions of Tat more significantly than limitations caused by post-translational modifications.
It has been proposed that HIV-1 Tat, which also lacks a signal sequence, follows the same unconventional secretion pathway for exocytosis that is employed by FGF2, however, a direct comparison of the interaction kinetics of either Tat or FGF2 to PtdIns(4,5)P2 demonstrated dissimilar binding between the two proteins and the phospholipid, suggesting that the secretion mechanism may be analogous rather than homologous 38. Based on this information, we would propose that while there has been considerable evidence reported indicating the formation of membrane pores by oligomerization of Tat, it has also been shown that alternative mechanisms contribute, at least in part, to the secretion of Tat and must be further characterized. After binding to PtdIns(4,5)P2, Tat secretion can diverge into several pathways that we propose to be categorized as: oligomerization-mediated pore formation, spontaneous translocation, and incorporation into exosomes (Figure 1). In this review, each of these pathways will be examined in detail, the gaps in published knowledge will be addressed, and evidence will be presented for these mechanisms occurring in a variety of cells that contribute to the development of HAND.
Figure 1. An overview of the secretion mechanisms of HIV-1 Tat.
Tat can be secreted from cells by various mechanisms. Interaction with PtdIns(4,5)P2 allows Tat to be anchored or inserted into the plasma membrane. It has been proposed that Tat can be spontaneously translocated, though the final step has yet to be characterized. It may, however, be a precursor to oligomerization-mediated pore formation, which requires an increase in the concentration of cytosolic Tat. This is the most well characterized mechanism of Tat secretion. Finally, it has been suggested in the literature that Tat can be incorporated into multiple vesicular bodies and be incorporated into exosomes and be released. However, there is limited evidence on this mechanism and it requires more investigation. (INTRA, Intracellular; EXT, Extracellular; MVB, multiple vesicular body)
Oligomerization of Tat mediates the formation of pores in the plasma membrane
PtdIns(4,5)P2 is required for the efficient secretion of HIV-1 Tat and although Tat has been demonstrated to interact with other phospholipids, disruption of the interaction with PtdIns(4,5)P2 has been shown to attenuate secretion 3. This was determined by utilizing flotation gradient and lipid sedimentation assays to examine the interaction of Tat with phosphoinositides present in the plasma membrane 3. Tat bound with similar specificity to PtdIns(3,4,5)P3, however, the interaction occurred with a lower affinity. Isothermal titration calorimetry (ITC) of the interaction with Tat and PtdIns(3,4,5)P3 resulted in no detectable heat production 39. Tat did not bind to PtdIns(3,4,5)P3 in the ITC system, further supporting the low affinity interaction between the lipid and protein. In a different study, neomycin, which has been shown to inhibit protein binding to PtdIns(4,5)P2, decreased the secretion of Tat in a dose-dependent manner, demonstrating the specificity of Tat for PtdIns(4,5)P2 40. Further analysis of this interaction involved alanine substitutions, mutagenesis, and deletion studies of Tat, which determined that the basic domain (residues 48–57; GRKKRRQRRR) and a conserved tryptophan (residue 11; W) were required for efficient binding of Tat to PtdIns(4,5)P2, which agreed with previous reported studies 41,42. Overall, single alanine substitutions within the basic domain did not significantly alter secretion, such as substitutions at residue 50 or 51 alone, however, combinatorial substitutions, such as replacing residues 49 to 51 with alanine, were capable of ablating secretion. Alteration of W11 also impacted the stability of the interaction between Tat and PtdIns(4,5)P2. Replacement of W11 with a either a hydrophobic or hydrophilic residue prevented insertion of the protein into the plasma membrane, and thus attenuated Tat secretion 43.
HIV-1 Tat is a highly basic and hydrophilic protein; therefore, it does not easily cross the hydrophobic plasma membrane 44–46. The direct translocation of hydrophilic proteins like Tat across the plasma membrane may occur by formation of a water pore 32,38. The water pore is an opening through both bilayer leaflets 46. The rate and quantity of protein translocation across the pore has been demonstrated to be dependent on the number of basic residues, specifically the abundance of arginine residues 47. The basic domain of Tat contains two lysine and six arginine residues, causing the protein to be strongly cationic. Additionally, the arginine-rich motif has been demonstrated to be necessary and sufficient for translocation across the plasma membrane 44,47. Studies on Tat internalization have demonstrated that all the cationic residues are necessary for translocation across the plasma membrane in both Tat peptides and alanine-substituted Tat peptides 39. This is in direct opposition with the secretion of Tat, which can proceed with single alanine substitutions within the basic domain. The mechanisms of the secretion and internalization of Tat differ, though there are similarities, such as the basic domain of Tat being crucial for translocation through the plasma membrane. Furthermore, arginine is a charged amino acid, while alanine is hydrophobic, and substitution of varying amounts of arginine residues all impacted the translocation rate of Tat across the membrane, suggesting that the six arginine residues may be necessary for pore formation. Additionally, the importance of arginine side chains in membrane kinetics is not unique to Tat and has been associated with the opening and closing of voltage sensitive ion channels, which are responsible for transmembrane potentials 45.
The pore formation of Tat is initiated by recruitment of the viral protein to the plasma membrane and interaction with PtdIns(4,5)P2, which acts as a platform for multiple proteins involved in exocytosis (Figure 2A) 38,45,48. Tat binds to PtdIns(4,5)P2 with high affinity and multiple Tat proteins are recruited to the same location, resulting in oligomerization on the cytoplasmic side of the plasma membrane 38. Interaction with PtdIns(4,5)P2 has been shown to be crucial for Tat oligomerization and when PtdIns(4,5)P2 is not present Tat is not recruited, and is unable to form a pore 38,48. At low concentrations of Tat in the cytoplasm, the arginine and lysine side chains begin to interact with the phosphate and carbonyl groups of the plasma membrane, specifically PtdIns(4,5)P2. As the concentration of Tat increases, to approximately one protein per eight lipids, the attractive forces between the arginine residues and phosphates will intensify and cause a gradual thinning of the plasma membrane bilayer 44–47. Once this occurs, the arginine and lysine residues penetrate the bilayer, which will solvate the charged side chains 45,49. This causes the formation of a pore, allowing the secretion of free Tat molecules. As the viral protein is secreted, the cytoplasmic concentration of Tat decreases, causing a reduction in the cumulative attractive forces between the arginine residues and phosphates, resulting in closure of the pore. The pore is transient in nature, which additionally prevents leakage of other cellular proteins 44,45,47,49.
Figure 2. Detailing the mechanisms of HIV-1 Tat secretion.
A) The formation of pores in the plasma membrane has been shown to be mediated by Tat oligomerization. 1. The arginine and lysine side chains of Tat, within the basic domain, interact with the phosphate and carbonyl groups on PtdIns(4,5)P2. 2. As the concentration of Tat increases to one peptide per eight lipids, the plasma membrane gradually thins and, along with water, causes the formation of a pore. 3. As the viral protein is released, the cytoplasmic concentration of Tat decreases, resulting in the pore closing. B) The spontaneous translocation of Tat is mediated by interaction with PtdIns(4,5)P2. 1. The basic domain (light blue) of Tat (dark blue), residues 48-57, binds to PtdIns(4,5)P2 (orange). 2.
The secretion of FGF2 by formation of a membrane pore has been demonstrated to be reduced by pharmacological inhibition of Na/K-ATPase, a pump responsible for generating membrane potentials, using ouabain 27. ATP1A1, the α1-chain of Na/K-ATPase, was responsible for recruiting FGF2 to the plasma membrane, allowing for efficient secretion 50. It was recently demonstrated that Tat had a similar mechanism and by inhibiting the Na/K-ATPase using ouabain, the secretion of Tat was impaired 13. Secretion was not completely ablated, however, suggesting that pore formation is not the sole route for Tat secretion. The evidence in the literature supports the formation of a membrane pore as the predominant mechanism of FGF2 secretion and we would propose that this likely holds true for Tat, however, alternative mechanisms may contribute as well.
Secretion of Tat is mediated by interaction with PtdIns(4,5)P2 – spontaneous translocation
The spontaneous translocation of Tat across the plasma membrane is dependent upon interactions with PtdIns(4,5)P2 (Figure 2B). The current understanding is that translocation can be initiated when the basic domain of Tat binds to the polar head of PtdIns(4,5)P2 3,5. This interaction is then stabilized when the conserved W11 inserts into the plasma membrane, though it is not known what Tat is specifically interacting with to facilitate the interaction with the membrane 38,43. W11 may bind to a hydrophobic tail, but it is unclear whether this interaction occurs with the same molecule of PtdIns(4,5)P2 or with the tail of another phospholipid. After these events occur, Tat is transported extracellularly, however, this process has yet to be fully elucidated. To understand the mechanism more comprehensively, the secretion kinetics of Tat have been examined by altering pH and temperature. Tat secretion was determined to be temperature-dependent, reducing the temperature from 37°C to 16°C prevented secretion, and occur in the absence of cell lysis, as demonstrated by the lack of luciferase release, which was cotransfected into Jurkat cells along with Tat expression vectors 5,41. While investigating the insertion of Tat into endosomal membranes, it was demonstrated that the biologically relevant pH of 5.3 was able to induce a conformational change in Tat 43. This conformational change dramatically increased Tat insertion into the membrane by exposure of W11. It is important to note that the lipid monolayer utilized in these experiments was a model of the endosomal membrane for internalization of Tat and did not include PtdIns(4,5)P2, thus the results may differ if the lipid composition was modeled to the inner leaflet of the plasma membrane. Additionally, a molecular dynamic analysis of Tat suggested that residue 11 is not accessible without acidification 51. However, it has been verified that a reduction in pH is not required for insertion of Tat into the plasma membrane for efficient secretion, as it was shown that the mere presence of a lipid bilayer could alter the conformation of Tat at a neutral pH 38. Additionally, it has been demonstrated that acidification was not required for Tat secretion, as Tat was observed to insert into a lipid monolayer at a pH of 7.0 in the presence of PtdIns(4,5)P2 3,5. These distinctions, and others, between the intracellular and extracellular interaction of Tat and the plasma membrane demonstrates the complexity of this viral protein and factors that may contribute to spontaneous translocation. Furthermore, there is conflicting evidence on the order of the initiation for spontaneous translocation of Tat, which also applies to the other mechanisms. There are reports of W11 initiating the interaction with PtdIns(4,5)P2, although the majority appear to accept the basic domain is responsible for initiation 3,38,45. After the initial interaction with PtdIns(4,5)P2, the process by which Tat becomes extracellular remains to be determined. In addition, the kinetics of this interaction, such as the rate at which this occurs and if specific concentrations of Tat are required for this mechanism still need to be elucidated. Furthermore, it has yet to be conclusively shown that this mechanism is separate from oligomerization-mediated pore formation. Based on the lack of evidence relevant to the connection between these mechanisms, we would propose that they should be characterized separately until demonstrated otherwise.
The interaction of Tat with PtdIns(4,5)P2 does not always result in secretion, since other cellular pathways can also be altered. Interestingly, only Tat capable of binding to PtdIns(4,5)P2 exerts an effect on the exocytosis and secretion of the viral protein from neuroendocrine cells 48. Overexpression of PIP5K, which is the major enzyme synthesizing PtdIns(4,5)P2, rescued, to some degree, the overexpression of Tat, suggesting that Tat may sequester PtdIns(4,5)P2. Tat also inhibited the binding of annexin A2, which interacts with PtdIns(4,5)P2, providing in vivo evidence for direct binding to PtdIns(4,5)P2. The co-expression of Tat in PC12 cells with a drug-inducible type IV 5-phosphatase or the PtdIns(4,5)P2 5-phosphatase, synaptojanin, reduced the levels of plasma membrane PtdIns(4,5)P2. Reduction of PtdIns(4,5)P2 in both cases altered the plasma membrane distribution of Tat, which became largely cytosolic 48. Furthermore, Tat expression had significant effects on the actin cytoskeleton, which plays a vital role in maintaining the morphology of the cell. Feedback loops between the actin cytoskeleton and the plasma membrane has been proposed to have further effects on the organization of the plasma membrane phosphatidylinositol lipids in the cell and may influence exosome formation.
Incorporation of Tat into exosomes
Exosome-mediated trafficking of Tat is still a novel concept with only a limited number of studies reported in the literature investigating the mechanism. Evidence for this process is limited to the detection of Tat in exosomes isolated from transfected U-373MG cells, an astrocytic cell line 52. The results in this report suggested evidence for higher HIV-1 activation with exosome-associated Tat when compared to Tat not associated with an exosome. Interestingly, the basic domain of Tat did not affect packaging into exosomes, however, this result has not been examined in other biologically relevant cellular phenotypes. Furthermore, residue 11 was not examined, allowing for speculation on the importance of this residue for incorporation into exosomes.
Exosomes are small vesicles (30–100nm) that are released from cells through endocytic mechanisms. The biogenesis of these vesicles commences with the formation of early endosomes following endocytosis. Early endosomes mature into late endosomes, or multi-vesicular bodies (MVBs), marked by the presence of intraluminal vesicles (ILV) in the lumen of the endosome 53. Inward budding of endosome membrane forms ILVs and permits uptake of diverse cytosolic content into the vesicle and transmembrane, causing exosomes to have unique surface markers including: CD9+, CD63+, ALIX and TGS101 54–56. Tat incorporation and recruitment to exosomes can be rationalized by the interaction with PtdIns(4,5)P2 during exosome biogenesis. PtdIns(4,5)P2 is enriched in exosomes and Tat has been detected within exosomes secreted from transfected cells 52. In addition to PtdIns(4,5)P2 binding, HIV-1 Tat may associate with exosomes through other mechanisms. DNA, RNA, and protein, such as Tat, can be incorporated during the sorting of various ILVs 57–59. These ILVs can then either fuse with lysosomes for destruction or fuse with the plasma membrane to secrete exosomal vesicles that affect bystander cells 52,60,61. Tat can potentially enter the exosome during ILV formation, a process which uptakes contents from the cytosol (Figure 2C). Additionally, Tat may associate with other cellular content, such as RNA, to promote incorporation into exosomes. If the exosomal membrane is enriched in proteins that bind HIV-1 Tat, these interactions could potentially lead to enrichment of Tat on the inner or outer exosomal membrane. An important function of Tat, as mentioned earlier, is its ability to bind to TAR and recruit elongation factors for proviral transcription. It has been shown that the core domain of Tat (residues 38–48) can bind to TAR RNA 62–64. Evidence for TAR RNA elements found in exosomes has suggested another potential mechanism for HIV Tat incorporation into exosomes. Exosomes derived from HIV-1-infected cells or patients have been shown to contain TAR, which has been suggested to enhance HIV-1 replication in the recipient cell, via downregulation of apoptosis 65. The incorporation of TAR into the exosomes potentially provides a binding site to tether Tat for subsequent release and transport.
Moreover, HIV-1 Tat has been shown to bind to noncoding RNAs, specifically microRNAs (miRNA) that are also found in exosomes. RNAs are a required element for Tat to bind and inhibit Dicer function during miRNA biogenesis, highlighting the importance of the interaction between Tat and RNA as a mediator of cellular function 66. Within the basic domain of Tat, residue 51 has been shown to have functional roles in miRNA binding 67. Additionally, Tat has been demonstrated to interact and inhibit the activity of over 300 miRNAs, with a unique subset of these miRNAs binding to Tat with high affinity 68. Furthermore, the hairpin structure of HIV-1 TAR interacts with Dicer to yield TAR-derived miRNAs found in infected cells 69. During an HIV-1 infection, the cellular profile of miRNA expression is altered 70–74. These virus-associated miRNAs and virus-mediated miRNA changes may have unique interplay with Tat and influence loading into exosomes. Cells transfected with HIV-1 Tat secrete specific miRNAs, notably miR-132 and miR29b 72,75. Furthermore, during the formation of ILVs when miRNAs, proteins, and other contents are packaged into exosomes, the ability of Tat to bind to miRNA could promote Tat-miRNA packaging into vesicles for release. Tat has also been shown to interact with the Y-box 1 protein, a critical factor involved in the sorting of non-coding RNA into exosomes 76–78. This interaction may support the packaging of Tat into exosomes and further suggests that Tat may be able to alter the incorporation of miRNA and other non-coding RNA into exosomes.
Evidence of HIV-1 Tat incorporation into exosomes and delivery into surrounding cells offers new avenues for investigation, particularly in HAND-related neuropathogenesis. Several studies suggest that exosomes can facilitate horizontal transfer of cargo and stimulate intracellular signaling pathways 58,79,80. Exosomes have been shown to be involved in the regulation of many processes, including neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease 81. While the entire mechanism remains unclear, there is emerging evidence for exosome-mediated neuropathogenesis in HAND, specifically in the packaging of Tat. Many of these released vesicles contain DNA, RNA, and proteins, which may also confer pro-neoplastic and inflammatory effects 61,82,83. These extracellular vesicles carry the genetic and proteomic profile that mirror the cell it is secreted from 59. These mechanisms highlight the unique, indirect modes of transferring aberrant factors in HIV-1 CNS infection, a process that still requires more specific diagnostics and targeted treatment. We rationalize that exosomal trafficking of DNA and protein, specifically Tat, from HIV-1-infected cells to healthy cells could potentially lead to chronic inflammation and subsequent worsening of HAND. It has been demonstrated that Nef, another HIV-1 protein, was secreted via exosomes, supporting the rationale that Tat may be incorporated in exosomes as well 84. Furthermore, RNA contained within exosomes is stable, suggesting that the membrane of an exosome could protect the contents and potentially even increase the stability of HIV-1 Tat protein during transport 59,85. While there is limited evidence for the secretion of Tat in exosomes, we would propose that this mechanism may occur. Previous work suggested that FGF2 may be enriched in extracellular vesicles, specifically exosomes 86. However, quantification of FGF2 secretion via plasma membrane blebbing and release in exosomes derived from MVBs concluded that this was not the predominant mechanism for the efficient secretion of FGF2 31. However, recent work has demonstrated that Tat was not present in the exosomes isolated from the media of transfected HEK293T cells 87. The addition of a c-Myc nuclear localization signal to the C-terminus of Tat was required for detectable incorporation into exosomes. Furthermore, it was shown that Tat was not present in exosomes isolated from J1.1 or Jurkat cells 88. However, as previously stated, there has been data published that has demonstrated the presence of Tat within exosomes derived from U373.MG cells. Given these observations, the exact mechanisms that are operative to facilitate Tat secretion may be dependent on the type of cell involved in the secretion of Tat. To answer this question conclusively, additional secretion studies will be required to clearly demonstrate the exact mechanisms that may be involved in the secretion of Tat across a number of pathogenically relevant cellular phenotypes that may ultimately contribute to the development and severity of HAND.
Conclusion
PtdIns(4,5)P2 is relatively abundant in the mammalian plasma membrane, thus Tat secretion may occur in a variety of cell types. The secretion of Tat appears to be dependent on the intracellular concentration of Tat, thus restricting these mechanisms to cells that have active viral transcription. The investigations discussed in this review were predominantly performed in primary CD4+ T cells and T cell lines. CD4+ T cells are one of the main targets for HIV-1 infection and are the predominant HIV-1-susceptible cell in the periphery 89. Monocytes and macrophages are also susceptible and permissive to HIV-1 90. Additionally, studies suggest that astrocytes are susceptible to HIV-1 infection in the CNS and although histology of patient samples have detected Tat within astrocytes, it is still unclear whether infection of these cells occurs frequently enough to alter the microenvironment of the CNS 91,92. These cell types contribute to the development of HAND and there is limited literature on Tat secretion alterations that are cell-specific. While studies that have begun to define these three secretion mechanisms of Tat (Table 1), there is still additional research required to completely elucidate them. After the initial interaction of Tat with PtdIns(4,5)P2 there are many questions that remain to be addressed. What causes the release of Tat from binding to PtdIns(4,5)P2 and allows it to be spontaneously translocated? Do these mechanisms occur at different rates in different cells? Do different cells have a preferential mechanism? Do these mechanisms occur simultaneously? Are there additional mechanisms associated with Tat secretion that have not yet been elucidated, such as incorporation into microvesicles? Is a single molecule of Tat able to be secreted and then interact with the plasma membrane of the same cell to be re-internalized? Does the full-length Tat protein (101 residues) exhibit the same secretion kinetics as the truncated protein (86 residues) used to examine and elucidate these processes? These are but a few of many unanswered questions.
Table 1. Evidence for the specific mechanisms of HIV-1 Tat secretion by a variety of model systems.
Enzyme-linked immunosorbent assay (ELISA); Fourier-transform infrared spectroscopy (FTIR); Giant unilamellar vesicle (GUV); Confocal laser scanning microscopy (CLSM); Synchrotron X-ray scattering (SAXS); and Surface plasmon resonance (SPR)
| MECHANISM | MODEL SYSTEM(S) | METHOD(S) | REFERENCE |
|---|---|---|---|
| Spontaneous translocation | Jurkat | Pharmacological characterization; ELISA; Immunofluorescence | Rayne F. et al., Cell. Biol. Int., 2010 |
| Primary human CD4+ T-cell; Jurkat; Liposomes | Liposome sedimentation assay; ELISA; Immunofluorescence | Rayne F. et al., EMBO J., 2010 | |
| Jurkat; Liposomes; Lipid monolayer | Fluorescence spectroscopy; Immunofluorescence | Yezid H. et al., J. Biol. Chem., 2009 | |
| COS-1 | Western blot; Gel shift assay | Chang HC. et al., AIDS, 1997 | |
| Exosome | Primary mouse astrocytes; U373.MG; SHSY-5Y; HEK293T; TZM-bl | Western blot; Exosome fractionation | Rahimian P. et al., J. Neurovirol., 2016 |
| HEK293T (No Tat present in exosomes) | Western blot; Exosome fractionation | Tang X. et al, JCI Insight, 2018 | |
| J1.1; Jurkat (No Tat present in exosomes) | Western blot; | Narayanan A. et al., J Biol Chem., 2013 | |
| Pore formation | In silico | Molecular dynamic simulations | Herce H. et al., PNAS, 2007 |
| Liposomes | Flotation gradient; FTIR spectroscopy; Carboxyfluorescein | Zeitler M. et al., JBC Papers, 2015 | |
| GUV | Light microscopy; CLSM; Single molecule microscopy | Ciobanasu C. et al., Biophys. J., 2010 | |
| GUV | Confocal microscopy; SAXS | Mishra A. et al., Angew. Chem. Int. Ed., 2008 | |
| In silico | Molecular dynamics simulations | Huang K. et al., Biophys. J., 2013 | |
| In silico | Molecular dynamics simulations | Herce H. et al., Biophys. J., 2009 | |
| Primary human CD4+ T-cells; CHO K1; psgA-745; HEK293T; HeLa; U2OS | SPR binding assay; Phosphoimaging; Western blot | S. Agostini. et al., EBioMedicine., 2017 | |
| Undefined | Primary cells derived from Kaposi’s sarcoma lesions on HIV-1 infected patients; H9; Jurkat; COS-1; HLM1 | Western blot; Immunostaining | Ensoli B. et al., J. Virol., 1993 |
| Primary human macrophages; Primary rat hippocampal neurons; U-937; NG-108 mouse neuroblastoma | Western blot; ELISA | Johnston JB. et al., Ann. Neurol., 2001 | |
| Primary human astrocytes; Rat C6 glioma; SVGA; HeLa | ELISA; Immunofluorescence | Chauhan A. et al. J. Biol. Chem., 2003 |
While several studies have addressed some of these questions, there is still controversy regarding the isoforms of Tat and whether they are truly representative of what is occurring within HIV-1-infected individuals. Tat is encoded by two exons, which are alternatively spliced to produce the full-length 101-residue protein 93. There are, however, 72-residue and 86-residue isoforms of Tat that are commonly utilized in experimental systems, as well as various length peptides. Investigations have used expression constructs 94,95 and recombinant proteins 96–98 of the truncated 86-residue isoform of Tat to examine various biological processes. These points prompt the question that the experimental results one obtains may differ if performed with the full-length protein. Interestingly, the truncated isoforms are sometimes even referred to as the “full-length” protein. The first HIV-1 isolates (LAI, formerly LAV, and HTLV-IIIB) contained a pre-mature stop codon in the second exon of Tat, which encoded for an 86-residue protein 99. It was later discovered that full-length Tat was 101-residues in length. Additionally, it is now understood that the full-length protein is more prevalent in clinical isolates 100, and that variability within the second exon causes functional alterations, such as changes in viral gene expression and replication in specific cell populations 101,102. Furthermore, the full-length protein has not been studied as frequently as the truncated isoforms. Moreover, the majority of the functional domains are within the first exon 103, meaning the 72-residue and 86-residue isoforms of Tat transactivate the HIV-1 LTR similarly to the 101-residue full-length protein in most cell types 104,105. Previous investigations studying the mechanism of Tat secretion have commonly used the 86-residue isoform 5,38, however, the full-length protein has not yet been examined. Furthermore, the exact residues that comprise the basic domain of HIV-1 Tat are not consistent in the literature. The basic domain of Tat has been reported as either residues 49-57 106 or 48-57 107, though others, such as 47-57, have been used 108. A single residue could alter or even ablate the secretion of Tat 3, suggesting that the full-length protein may yield different results. Tat is considered to be non-structural, the secondary structure of the protein remains a controversial topic, and the conformation of the protein is flexible 109. Thus, we propose that it is likely the chemical properties of the peptide that are more relevant to secretion. Certain single residue substitutions may not alter the overall charge in the basic domain significantly enough to alter secretion 51. It has been suggested that only resides 49 to 51 and residue 11 are required for interaction with PtdIns(4,5)P2 5, however, utilization of different Tat isoforms between investigations makes comparisons difficult to interpret.
Combinatorial mutations are reasoned to alter the function of Tat considerably. These mutations are caused by several factors, including the error rate of HIV-1 reverse transcriptase, in conjunction with selective immune pressures 103,110. The impact of genetic variability on the function of Tat has been well documented in the context of LTR transactivation 94,111, but there are comparatively fewer publications examining the effect of genetic variation on Tat secretion. In silico analysis of Tat genetic signatures has been demonstrated to be correlated to co-receptor utilization 112. Tat sequences derived from patients diagnosed with HIV-1-associated dementia 7 and those without dementia have demonstrated differences in transactivation of the HIV-1 LTR 111. Furthermore, the individual influence of specific amino acid residues, such as the dicysteine motif C30C31 in Tat from HIV-1 subtype C 113,114, have been the subject of debate regarding their contribution to neuroprotection. Correlating the genetic variability of Tat to neurocognitive impairment and alterations in secretion kinetics has yet to be examined. This information would provide insight into the contribution of extracellular Tat and the severity of HAND, potentially leading to more comprehensive diagnostic tools for those chronically infected with HIV-1.
Synopsis.
The HIV-1 transactivator of transcription (Tat) protein causes a myriad of effects to bystander cells, such as apoptosis and gene expression alterations, once secreted from infected cells. Tat is accepted as a crucial component of HIV-1-associated neurocognitive disorders (HAND) by causing neurotoxicity within the CNS. The mechanisms associated with Tat secretion are still poorly understood. This review examines gaps in published knowledge and uses existing evidence to categorize the mechanisms of secretion and their relevance to cells influenced during HAND.
Acknowledgments
The authors were funded in part by the Public Health Service, National Institutes of Health, through grants from the National Institute of Neurological Disorders and Stroke (NINDS) R01 NS089435 (PI, Michael R. Nonnemacher), the NIMH Comprehensive NeuroAIDS Center (CNAC) P30 MH092177 (Kamel Khalili, PI; Brian Wigdahl, PI of the Drexel subcontract involving the Clinical and Translational Research Support Core) and under the Ruth L. Kirschstein National Research Service Award T32 MH079785 (PI, Jay Rappaport; with Brian Wigdahl serving as the PI of the Drexel University College of Medicine component and Olimpia Meucci as Co-Director). The contents of the paper are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.
Footnotes
Conflict of Interest Statement: The authors have no conflicts of interest to disclose.
References
- 1.Dingwall C, Ernberg I, Gait MJ, et al. Human immunodeficiency virus 1 tat protein binds trans-activation-responsive region (TAR) RNA in vitro. Proc Natl Acad Sci U S A. 1989;86(18):6925–6929. doi: 10.1073/pnas.86.18.6925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Li L, Aiamkitsumrit B, Pirrone V, et al. Development of co-selected single nucleotide polymorphisms in the viral promoter precedes the onset of human immunodeficiency virus type 1-associated neurocognitive impairment. J Neurovirol. 2011;17(1):92–109. doi: 10.1007/s13365-010-0014-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rayne F, Debaisieux S, Yezid H, et al. Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J. 2010;29(8):1348–1362. doi: 10.1038/emboj.2010.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Albini A, Ferrini S, Benelli R, et al. HIV-1 Tat protein mimicry of chemokines. Proc Natl Acad Sci U S A. 1998;95(22):13153–13158. doi: 10.1073/pnas.95.22.13153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rayne F, Debaisieux S, Bonhoure A, Beaumelle B. HIV-1 Tat is unconventionally secreted through the plasma membrane. Cell Biol Int. 2010;34(4):409–413. doi: 10.1042/CBI20090376. [DOI] [PubMed] [Google Scholar]
- 6.Bachani M, Sacktor N, McArthur JC, Nath A, Rumbaugh J. Detection of anti-tat antibodies in CSF of individuals with HIV-associated neurocognitive disorders. J Neurovirol. 2013;19(1):82–88. doi: 10.1007/s13365-012-0144-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hofman FM, Dohadwala MM, Wright AD, Hinton DR, Walker SM. Exogenous tat protein activates central nervous system-derived endothelial cells. J Neuroimmunol. 1994;54(1–2):19–28. doi: 10.1016/0165-5728(94)90226-7. [DOI] [PubMed] [Google Scholar]
- 8.Hudson L, Liu J, Nath A, et al. Detection of the human immunodeficiency virus regulatory protein tat in CNS tissues. J Neurovirol. 2000;6(2):145–155. doi: 10.3109/13550280009013158. [DOI] [PubMed] [Google Scholar]
- 9.Banks WA, Robinson SM, Nath A. Permeability of the blood-brain barrier to HIV-1 Tat. Exp Neurol. 2005;193(1):218–227. doi: 10.1016/j.expneurol.2004.11.019. [DOI] [PubMed] [Google Scholar]
- 10.Dahiya S, Irish BP, Nonnemacher MR, Wigdahl B. Genetic variation and HIV-associated neurologic disease. Adv Virus Res. 2013;87:183–240. doi: 10.1016/B978-0-12-407698-3.00006-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gannon P, Khan MZ, Kolson DL. Current understanding of HIV-associated neurocognitive disorders pathogenesis. Curr Opin Neurol. 2011;24(3):275–283. doi: 10.1097/WCO.0b013e32834695fb. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Carey AN, Sypek EI, Singh HD, Kaufman MJ, McLaughlin JP. Expression of HIV-Tat protein is associated with learning and memory deficits in the mouse. Behav Brain Res. 2012;229(1):48–56. doi: 10.1016/j.bbr.2011.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Agostini S, Ali H, Vardabasso C, et al. Inhibition of Non Canonical HIV-1 Tat Secretion Through the Cellular Na+,K+-ATPase Blocks HIV-1 Infection. EBioMedicine. 2017;21:170–181. doi: 10.1016/j.ebiom.2017.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mediouni S, Darque A, Baillat G, et al. Antiretroviral therapy does not block the secretion of the human immunodeficiency virus tat protein. Infect Disord Drug Targets. 2012;12(1):81–86. doi: 10.2174/187152612798994939. [DOI] [PubMed] [Google Scholar]
- 15.Ensoli B, Barillari G, Salahuddin SZ, Gallo RC, Wong-Staal F. Tat protein of HIV-1 stimulates growth of cells derived from Kaposi’s sarcoma lesions of AIDS patients. Nature. 1990;345(6270):84–86. doi: 10.1038/345084a0. [DOI] [PubMed] [Google Scholar]
- 16.Divecha N, Irvine RF. Phospholipid signaling. Cell. 1995;80(2):269–278. doi: 10.1016/0092-8674(95)90409-3. [DOI] [PubMed] [Google Scholar]
- 17.D’Santos CS, Clarke JH, Divecha N. Phospholipid signalling in the nucleus. Een DAG uit het leven van de inositide signalering in de nucleus. Biochim Biophys Acta. 1998;1436(1–2):201–232. doi: 10.1016/s0005-2760(98)00146-5. [DOI] [PubMed] [Google Scholar]
- 18.Hinchliffe KA, Ciruela A, Irvine RF. PIPkins1, their substrates and their products: new functions for old enzymes. Biochim Biophys Acta. 1998;1436(1–2):87–104. doi: 10.1016/s0005-2760(98)00140-4. [DOI] [PubMed] [Google Scholar]
- 19.Malek M, Kielkowska A, Chessa T, et al. PTEN Regulates PI(3,4)P2 Signaling Downstream of Class I PI3K. Mol Cell. 2017;68(3):566–580. e510. doi: 10.1016/j.molcel.2017.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Leslie NR, Downes CP. PTEN: The down side of PI 3-kinase signalling. Cell Signal. 2002;14(4):285–295. doi: 10.1016/s0898-6568(01)00234-0. [DOI] [PubMed] [Google Scholar]
- 21.Niemeyer MI, Cid LP, Paulais M, Teulon J, Sepulveda FV. Phosphatidylinositol (4,5)-bisphosphate dynamically regulates the K2P background K+ channel TASK-2. Sci Rep. 2017;7:45407. doi: 10.1038/srep45407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.van den Bout I, Divecha N. PIP5K-driven PtdIns(4,5)P2 synthesis: regulation and cellular functions. J Cell Sci. 2009;122(Pt 21):3837–3850. doi: 10.1242/jcs.056127. [DOI] [PubMed] [Google Scholar]
- 23.Gericke A, Leslie NR, Losche M, Ross AH. PtdIns(4,5)P2-mediated cell signaling: emerging principles and PTEN as a paradigm for regulatory mechanism. Adv Exp Med Biol. 2013;991:85–104. doi: 10.1007/978-94-007-6331-9_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Temmerman K, Ebert AD, Muller HM, Sinning I, Tews I, Nickel W. A direct role for phosphatidylinositol-4,5-bisphosphate in unconventional secretion of fibroblast growth factor 2. Traffic. 2008;9(7):1204–1217. doi: 10.1111/j.1600-0854.2008.00749.x. [DOI] [PubMed] [Google Scholar]
- 25.Nickel W, Seedorf M. Unconventional mechanisms of protein transport to the cell surface of eukaryotic cells. Annu Rev Cell Dev Biol. 2008;24:287–308. doi: 10.1146/annurev.cellbio.24.110707.175320. [DOI] [PubMed] [Google Scholar]
- 26.Schafer T, Zentgraf H, Zehe C, Brugger B, Bernhagen J, Nickel W. Unconventional secretion of fibroblast growth factor 2 is mediated by direct translocation across the plasma membrane of mammalian cells. J Biol Chem. 2004;279(8):6244–6251. doi: 10.1074/jbc.M310500200. [DOI] [PubMed] [Google Scholar]
- 27.Steringer JP, Lange S, Cujova S, et al. Key steps in unconventional secretion of fibroblast growth factor 2 reconstituted with purified components. Elife. 2017:6. doi: 10.7554/eLife.28985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Steringer JP, Muller HM, Nickel W. Unconventional secretion of fibroblast growth factor 2--a novel type of protein translocation across membranes? J Mol Biol. 2015;427(6 Pt A):1202–1210. doi: 10.1016/j.jmb.2014.07.012. [DOI] [PubMed] [Google Scholar]
- 29.Fettelschoss A, Kistowska M, LeibundGut-Landmann S, et al. Inflammasome activation and IL-1beta target IL-1alpha for secretion as opposed to surface expression. Proc Natl Acad Sci U S A. 2011;108(44):18055–18060. doi: 10.1073/pnas.1109176108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Keller M, Ruegg A, Werner S, Beer HD. Active caspase-1 is a regulator of unconventional protein secretion. Cell. 2008;132(5):818–831. doi: 10.1016/j.cell.2007.12.040. [DOI] [PubMed] [Google Scholar]
- 31.Seelenmeyer C, Stegmayer C, Nickel W. Unconventional secretion of fibroblast growth factor 2 and galectin-1 does not require shedding of plasma membrane-derived vesicles. FEBS Lett. 2008;582(9):1362–1368. doi: 10.1016/j.febslet.2008.03.024. [DOI] [PubMed] [Google Scholar]
- 32.Rabouille C. Pathways of Unconventional Protein Secretion. Trends Cell Biol. 2017;27(3):230–240. doi: 10.1016/j.tcb.2016.11.007. [DOI] [PubMed] [Google Scholar]
- 33.Wegehingel S, Zehe C, Nickel W. Rerouting of fibroblast growth factor 2 to the classical secretory pathway results in post-translational modifications that block binding to heparan sulfate proteoglycans. FEBS Lett. 2008;582(16):2387–2392. doi: 10.1016/j.febslet.2008.05.042. [DOI] [PubMed] [Google Scholar]
- 34.Boulanger MC, Liang C, Russell RS, et al. Methylation of Tat by PRMT6 regulates human immunodeficiency virus type 1 gene expression. J Virol. 2005;79(1):124–131. doi: 10.1128/JVI.79.1.124-131.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Col E, Caron C, Seigneurin-Berny D, Gracia J, Favier A, Khochbin S. The histone acetyltransferase, hGCN5, interacts with and acetylates the HIV transactivator, Tat. J Biol Chem. 2001;276(30):28179–28184. doi: 10.1074/jbc.M101385200. [DOI] [PubMed] [Google Scholar]
- 36.Kaehlcke K, Dorr A, Hetzer-Egger C, et al. Acetylation of Tat defines a cyclinT1-independent step in HIV transactivation. Mol Cell. 2003;12(1):167–176. doi: 10.1016/s1097-2765(03)00245-4. [DOI] [PubMed] [Google Scholar]
- 37.Ott M, Schnolzer M, Garnica J, et al. Acetylation of the HIV-1 Tat protein by p300 is important for its transcriptional activity. Curr Biol. 1999;9(24):1489–1492. doi: 10.1016/s0960-9822(00)80120-7. [DOI] [PubMed] [Google Scholar]
- 38.Zeitler M, Steringer JP, Muller HM, Mayer MP, Nickel W. HIV-Tat Protein Forms Phosphoinositide-dependent Membrane Pores Implicated in Unconventional Protein Secretion. J Biol Chem. 2015;290(36):21976–21984. doi: 10.1074/jbc.M115.667097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wender PA, Mitchell DJ, Pattabiraman K, Pelkey ET, Steinman L, Rothbard JB. The design, synthesis, and evaluation of molecules that enable or enhance cellular uptake: peptoid molecular transporters. Proc Natl Acad Sci U S A. 2000;97(24):13003–13008. doi: 10.1073/pnas.97.24.13003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arbuzova A, Martushova K, Hangyas-Mihalyne G, et al. Fluorescently labeled neomycin as a probe of phosphatidylinositol-4, 5-bisphosphate in membranes. Biochim Biophys Acta. 2000;1464(1):35–48. doi: 10.1016/s0005-2736(99)00243-6. [DOI] [PubMed] [Google Scholar]
- 41.Chang HC, Samaniego F, Nair BC, Buonaguro L, Ensoli B. HIV-1 Tat protein exits from cells via a leaderless secretory pathway and binds to extracellular matrix-associated heparan sulfate proteoglycans through its basic region. AIDS. 1997;11(12):1421–1431. doi: 10.1097/00002030-199712000-00006. [DOI] [PubMed] [Google Scholar]
- 42.Ensoli B, Buonaguro L, Barillari G, et al. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. J Virol. 1993;67(1):277–287. doi: 10.1128/jvi.67.1.277-287.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yezid H, Konate K, Debaisieux S, Bonhoure A, Beaumelle B. Mechanism for HIV-1 Tat insertion into the endosome membrane. J Biol Chem. 2009;284(34):22736–22746. doi: 10.1074/jbc.M109.023705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Durzynska J, Przysiecka L, Nawrot R, et al. Viral and other cell-penetrating peptides as vectors of therapeutic agents in medicine. J Pharmacol Exp Ther. 2015;354(1):32–42. doi: 10.1124/jpet.115.223305. [DOI] [PubMed] [Google Scholar]
- 45.Herce HD, Garcia AE. Molecular dynamics simulations suggest a mechanism for translocation of the HIV-1 TAT peptide across lipid membranes. Proc Natl Acad Sci U S A. 2007;104(52):20805–20810. doi: 10.1073/pnas.0706574105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Huang K, Garcia AE. Free energy of translocating an arginine-rich cell-penetrating peptide across a lipid bilayer suggests pore formation. Biophys J. 2013;104(2):412–420. doi: 10.1016/j.bpj.2012.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ciobanasu C, Siebrasse JP, Kubitscheck U. Cell-penetrating HIV1 TAT peptides can generate pores in model membranes. Biophys J. 2010;99(1):153–162. doi: 10.1016/j.bpj.2010.03.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tryoen-Toth P, Chasserot-Golaz S, Tu A, et al. HIV-1 Tat protein inhibits neurosecretion by binding to phosphatidylinositol 4,5-bisphosphate. J Cell Sci. 2013;126(Pt 2):454–463. doi: 10.1242/jcs.111658. [DOI] [PubMed] [Google Scholar]
- 49.Herce HD, Garcia AE, Litt J, et al. Arginine-rich peptides destabilize the plasma membrane, consistent with a pore formation translocation mechanism of cell-penetrating peptides. Biophys J. 2009;97(7):1917–1925. doi: 10.1016/j.bpj.2009.05.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zacherl S, La Venuta G, Muller HM, et al. A direct role for ATP1A1 in unconventional secretion of fibroblast growth factor 2. J Biol Chem. 2015;290(6):3654–3665. doi: 10.1074/jbc.M114.590067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pantano S, Carloni P. Comparative analysis of HIV-1 Tat variants. Proteins. 2005;58(3):638–643. doi: 10.1002/prot.20323. [DOI] [PubMed] [Google Scholar]
- 52.Rahimian P, He JJ. Exosome-associated release, uptake, and neurotoxicity of HIV-1 Tat protein. J Neurovirol. 2016 doi: 10.1007/s13365-016-0451-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sotelo JR, Porter KR. An electron microscope study of the rat ovum. J Biophys Biochem Cytol. 1959;5(2):327–342. doi: 10.1083/jcb.5.2.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol. 2012;83(11):1484–1494. doi: 10.1016/j.bcp.2011.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8(19):4083–4099. doi: 10.1002/pmic.200800109. [DOI] [PubMed] [Google Scholar]
- 56.Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6(3):267–283. doi: 10.1586/epr.09.17. [DOI] [PubMed] [Google Scholar]
- 57.Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285(23):17442–17452. doi: 10.1074/jbc.M110.107821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Muralidharan-Chari V, Clancy JW, Sedgwick A, D’Souza-Schorey C. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123(Pt 10):1603–1611. doi: 10.1242/jcs.064386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–1476. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Woodman PG, Futter CE. Multivesicular bodies: co-ordinated progression to maturity. Curr Opin Cell Biol. 2008;20(4):408–414. doi: 10.1016/j.ceb.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 62.Bayer P, Kraft M, Ejchart A, Westendorp M, Frank R, Rosch P. Structural studies of HIV-1 Tat protein. J Mol Biol. 1995;247(4):529–535. doi: 10.1006/jmbi.1995.0158. [DOI] [PubMed] [Google Scholar]
- 63.Rice AP, Carlotti F. Structural analysis of wild-type and mutant human immunodeficiency virus type 1 Tat proteins. J Virol. 1990;64(12):6018–6026. doi: 10.1128/jvi.64.12.6018-6026.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sodroski J, Rosen C, Wong-Staal F, et al. Trans-acting transcriptional regulation of human T-cell leukemia virus type III long terminal repeat. Science. 1985;227(4683):171–173. doi: 10.1126/science.2981427. [DOI] [PubMed] [Google Scholar]
- 65.Narayanan A, Iordanskiy S, Das R, et al. Exosomes derived from HIV-1-infected cells contain trans-activation response element RNA. J Biol Chem. 2013;288(27):20014–20033. doi: 10.1074/jbc.M112.438895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bennasser Y, Jeang KT. HIV-1 Tat interaction with Dicer: requirement for RNA. Retrovirology. 2006;3:95. doi: 10.1186/1742-4690-3-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bennasser Y, Le SY, Benkirane M, Jeang KT. Evidence that HIV-1 encodes an siRNA and a suppressor of RNA silencing. Immunity. 2005;22(5):607–619. doi: 10.1016/j.immuni.2005.03.010. [DOI] [PubMed] [Google Scholar]
- 68.Sardo L, Vakil PR, Elbezanti W, El-Sayed A, Klase Z. The inhibition of microRNAs by HIV-1 Tat suppresses beta catenin activity in astrocytes. Retrovirology. 2016;13:25. doi: 10.1186/s12977-016-0256-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Klase Z, Kale P, Winograd R, et al. HIV-1 TAR element is processed by Dicer to yield a viral micro-RNA involved in chromatin remodeling of the viral LTR. BMC Mol Biol. 2007;8:63. doi: 10.1186/1471-2199-8-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yeung ML, Bennasser Y, Myers TG, Jiang G, Benkirane M, Jeang KT. Changes in microRNA expression profiles in HIV-1-transfected human cells. Retrovirology. 2005;2:81. doi: 10.1186/1742-4690-2-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dreux M, Garaigorta U, Boyd B, et al. Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host Microbe. 2012;12(4):558–570. doi: 10.1016/j.chom.2012.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hu G, Yao H, Chaudhuri AD, et al. Exosome-mediated shuttling of microRNA-29 regulates HIV Tat and morphine-mediated neuronal dysfunction. Cell Death Dis. 2012;3:e381. doi: 10.1038/cddis.2012.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci U S A. 2010;107(14):6328–6333. doi: 10.1073/pnas.0914843107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 2012;1820(7):940–948. doi: 10.1016/j.bbagen.2012.03.017. [DOI] [PubMed] [Google Scholar]
- 75.Rahimian P, He JJ. HIV-1 Tat-shortened neurite outgrowth through regulation of microRNA-132 and its target gene expression. J Neuroinflammation. 2016;13(1):247. doi: 10.1186/s12974-016-0716-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ansari SA, Safak M, Gallia GL, Sawaya BE, Amini S, Khalili K. Interaction of YB-1 with human immunodeficiency virus type 1 Tat and TAR RNA modulates viral promoter activity. J Gen Virol. 1999;80(Pt 10):2629–2638. doi: 10.1099/0022-1317-80-10-2629. [DOI] [PubMed] [Google Scholar]
- 77.Shurtleff MJ, Temoche-Diaz MM, Karfilis KV, Ri S, Schekman R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. Elife. 2016:5. doi: 10.7554/eLife.19276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Shurtleff MJ, Yao J, Qin Y, et al. Broad role for YBX1 in defining the small noncoding RNA composition of exosomes. Proc Natl Acad Sci U S A. 2017;114(43):E8987–E8995. doi: 10.1073/pnas.1712108114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Demory Beckler M, Higginbotham JN, Franklin JL, et al. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Mol Cell Proteomics. 2013;12(2):343–355. doi: 10.1074/mcp.M112.022806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lee TH, D’Asti E, Magnus N, Al-Nedawi K, Meehan B, Rak J. Microvesicles as mediators of intercellular communication in cancer--the emerging science of cellular ‘debris’. Semin Immunopathol. 2011;33(5):455–467. doi: 10.1007/s00281-011-0250-3. [DOI] [PubMed] [Google Scholar]
- 81.De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front Immunol. 2015;6:203. doi: 10.3389/fimmu.2015.00203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hong BS, Cho JH, Kim H, et al. Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. BMC Genomics. 2009;10:556. doi: 10.1186/1471-2164-10-556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Pisetsky DS, Gauley J, Ullal AJ. Microparticles as a source of extracellular DNA. Immunol Res. 2011;49(1–3):227–234. doi: 10.1007/s12026-010-8184-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lenassi M, Cagney G, Liao M, et al. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 2010;11(1):110–122. doi: 10.1111/j.1600-0854.2009.01006.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.O’Driscoll L, Kenny E, Mehta JP, et al. Feasibility and relevance of global expression profiling of gene transcripts in serum from breast cancer patients using whole genome microarrays and quantitative RT-PCR. Cancer Genomics Proteomics. 2008;5(2):94–104. [PubMed] [Google Scholar]
- 86.Ceccarelli S, Visco V, Raffa S, Wakisaka N, Pagano JS, Torrisi MR. Epstein-Barr virus latent membrane protein 1 promotes concentration in multivesicular bodies of fibroblast growth factor 2 and its release through exosomes. Int J Cancer. 2007;121(7):1494–1506. doi: 10.1002/ijc.22844. [DOI] [PubMed] [Google Scholar]
- 87.Tang X, Lu H, Dooner M, Chapman S, Quesenberry PJ, Ramratnam B. Exosomal Tat protein activates latent HIV-1 in primary, resting CD4+ T lymphocytes. JCI Insight. 2018;3(7) doi: 10.1172/jci.insight.95676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Narayanan A, Iordanskiy S, Das R, et al. Exosomes derived from HIV-1-infected cells contain trans-activation response element RNA. J Biol Chem. 2013;288(27):20014–20033. doi: 10.1074/jbc.M112.438895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Nonnemacher MR, Quiterio S, Allen AG, Mele AR, Pirrone V, Wigdahl B. Myelomonocytic Cell Lines in Modeling HIV-1 Infection of the Bone Marrow. In: Ghosh A, editor. Biology of Myelomonocytic Cells. InTech; 2017. pp. 129–162. 2017/5/10 ed. [Google Scholar]
- 90.Baxter AE, Russell RA, Duncan CJ, et al. Macrophage infection via selective capture of HIV-1-infected CD4+ T cells. Cell Host Microbe. 2014;16(6):711–721. doi: 10.1016/j.chom.2014.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Takahashi K, Wesselingh SL, Griffin DE, McArthur JC, Johnson RT, Glass JD. Localization of HIV-1 in human brain using polymerase chain reaction/in situ hybridization and immunocytochemistry. Ann Neurol. 1996;39(6):705–711. doi: 10.1002/ana.410390606. [DOI] [PubMed] [Google Scholar]
- 92.Trillo-Pazos G, Diamanturos A, Rislove L, et al. Detection of HIV-1 DNA in microglia/macrophages, astrocytes and neurons isolated from brain tissue with HIV-1 encephalitis by laser capture microdissection. Brain Pathol. 2003;13(2):144–154. doi: 10.1111/j.1750-3639.2003.tb00014.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Purcell DF, Martin MA. Alternative splicing of human immunodeficiency virus type 1 mRNA modulates viral protein expression, replication, and infectivity. J Virol. 1993;67(11):6365–6378. doi: 10.1128/jvi.67.11.6365-6378.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ronsard L, Ganguli N, Singh VK, et al. Impact of Genetic Variations in HIV-1 Tat on LTR-Mediated Transcription via TAR RNA Interaction. Front Microbiol. 2017;8:706. doi: 10.3389/fmicb.2017.00706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Srinivasakumar N, Schuening F. Novel Tat-encoding bicistronic human immunodeficiency virus type 1-based gene transfer vectors for high-level transgene expression. J Virol. 2000;74(14):6659–6668. doi: 10.1128/jvi.74.14.6659-6668.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Chaloin O, Peter JC, Briand JP, et al. The N-terminus of HIV-1 Tat protein is essential for Tat-TAR RNA interaction. Cell Mol Life Sci. 2005;62(3):355–361. doi: 10.1007/s00018-004-4477-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Dandekar DH, Ganesh KN, Mitra D. HIV-1 Tat directly binds to NFkappaB enhancer sequence: role in viral and cellular gene expression. Nucleic Acids Res. 2004;32(4):1270–1278. doi: 10.1093/nar/gkh289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Donahue DA, Kuhl BD, Sloan RD, Wainberg MA. The viral protein Tat can inhibit the establishment of HIV-1 latency. J Virol. 2012;86(6):3253–3263. doi: 10.1128/JVI.06648-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chang SY, Bowman BH, Weiss JB, Garcia RE, White TJ. The origin of HIV-1 isolate HTLV-IIIB. Nature. 1993;363(6428):466–469. doi: 10.1038/363466a0. [DOI] [PubMed] [Google Scholar]
- 100.Lopez-Huertas MR, Callejas S, Abia D, et al. Modifications in host cell cytoskeleton structure and function mediated by intracellular HIV-1 Tat protein are greatly dependent on the second coding exon. Nucleic Acids Res. 2010;38(10):3287–3307. doi: 10.1093/nar/gkq037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kukkonen S, Martinez-Viedma Mdel P, Kim N, Manrique M, Aldovini A. HIV-1 Tat second exon limits the extent of Tat-mediated modulation of interferon-stimulated genes in antigen presenting cells. Retrovirology. 2014;11:30. doi: 10.1186/1742-4690-11-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lopez-Huertas MR, Mateos E, Sanchez Del Cojo M, et al. The presence of HIV-1 Tat protein second exon delays fas protein-mediated apoptosis in CD4+ T lymphocytes: a potential mechanism for persistent viral production. J Biol Chem. 2013;288(11):7626–7644. doi: 10.1074/jbc.M112.408294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Li L, Dahiya S, Kortagere S, et al. Impact of Tat Genetic Variation on HIV-1 Disease. Adv Virol. 2012;2012:123605. doi: 10.1155/2012/123605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Demarchi F, Gutierrez MI, Giacca M. Human immunodeficiency virus type 1 tat protein activates transcription factor NF-kappaB through the cellular interferon-inducible, double-stranded RNA-dependent protein kinase, PKR. J Virol. 1999;73(8):7080–7086. doi: 10.1128/jvi.73.8.7080-7086.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kameoka M, Rong L, Gotte M, Liang C, Russell RS, Wainberg MA. Role for human immunodeficiency virus type 1 Tat protein in suppression of viral reverse transcriptase activity during late stages of viral replication. J Virol. 2001;75(6):2675–2683. doi: 10.1128/JVI.75.6.2675-2683.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Xie X, Colberg-Poley AM, Das JR, et al. The basic domain of HIV-tat transactivating protein is essential for its targeting to lipid rafts and regulating fibroblast growth factor-2 signaling in podocytes isolated from children with HIV-1-associated nephropathy. J Am Soc Nephrol. 2014;25(8):1800–1813. doi: 10.1681/ASN.2013070710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mujeeb A, Bishop K, Peterlin BM, Turck C, Parslow TG, James TL. NMR structure of a biologically active peptide containing the RNA-binding domain of human immunodeficiency virus type 1 Tat. Proc Natl Acad Sci U S A. 1994;91(17):8248–8252. doi: 10.1073/pnas.91.17.8248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ziegler A, Seelig J. Interaction of the protein transduction domain of HIV-1 TAT with heparan sulfate: binding mechanism and thermodynamic parameters. Biophys J. 2004;86(1 Pt 1):254–263. doi: 10.1016/S0006-3495(04)74101-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Pantano S, Tyagi M, Giacca M, Carloni P. Amino acid modification in the HIV-1 Tat basic domain: insights from molecular dynamics and in vivo functional studies. J Mol Biol. 2002;318(5):1331–1339. doi: 10.1016/s0022-2836(02)00217-6. [DOI] [PubMed] [Google Scholar]
- 110.Dampier W, Nonnemacher MR, Mell J, et al. HIV-1 Genetic Variation Resulting in the Development of New Quasispecies Continues to Be Encountered in the Peripheral Blood of Well-Suppressed Patients. PLoS One. 2016;11(5):e0155382. doi: 10.1371/journal.pone.0155382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Boven LA, Noorbakhsh F, Bouma G, et al. Brain-derived human immunodeficiency virus-1 Tat exerts differential effects on LTR transactivation and neuroimmune activation. J Neurovirol. 2007;13(2):173–184. doi: 10.1080/13550280701258399. [DOI] [PubMed] [Google Scholar]
- 112.Antell GC, Dampier W, Aiamkitsumrit B, et al. Utilization of HIV-1 envelope V3 to identify X4- and R5-specific Tat and LTR sequence signatures. Retrovirology. 2016;13(1):32. doi: 10.1186/s12977-016-0266-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.de Almeida SM, Rotta I, Jiang Y, et al. Biomarkers of chemotaxis and inflammation in cerebrospinal fluid and serum in individuals with HIV-1 subtype C versus B. J Neurovirol. 2016;22(6):715–724. doi: 10.1007/s13365-016-0437-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Rao VR, Neogi U, Talboom JS, et al. Clade C HIV-1 isolates circulating in Southern Africa exhibit a greater frequency of dicysteine motif-containing Tat variants than those in Southeast Asia and cause increased neurovirulence. Retrovirology. 2013;10:61. doi: 10.1186/1742-4690-10-61. [DOI] [PMC free article] [PubMed] [Google Scholar]


