Abstract
Objectives
HIV-1 enters the central nervous system (CNS) early in infection, and a significant proportion of people with HIV experience CNS complications despite anti-retroviral therapy. Chronic immune dysfunction, inflammatory cytokines and chemokines, and viral proteins like Tat and gp120 released by HIV-1-infected immune cells are implicated in the pathogenesis of HIV-1-associated neurocognitive disorders (HAND).
Methods
To elucidate the contribution of non-viral factors to CNS complications in people with HIV-1, a comparative analysis of neurovirulent subtype B (HIV-1ADA) and non-neurovirulent subtype C (HIV-1Indie-C1) isolates was performed. Culture supernatants from HIV-1-infected PBMCs, either with or without immunodepletion of Tat and gp120, were used to treat SH-SY5Y neuroblastoma cells.
Results
HIV-1ADA-infected PBMC media showed significantly higher neurotoxicity than HIV-1IndieC1-infected PBMC media, notwithstanding the depletion of Tat and gp120, highlighting the role of non-viral factors (e.g., cytokines) contributing to neurotoxicity. A comparison of inflammatory profiles revealed that HIV-1ADA media contained elevated levels of cytokines (IL-1α, IL-1β, IL-6 and TNFα) and chemokines (CCL2, CCL3, CCL4 and IP-10). Given the involvement of Tat in upregulating immune mediators, we tested the role of purified Tat proteins from HIV-1 subtypes B and C in inducing an inflammatory phenotype in PBMCs. PBMCs from healthy subjects were treated with recombinant purified Tat from subtype B or C. Subtype B Tat induced a stronger inflammatory response than subtype C Tat.
Conclusions
These results confirm that both viral and non-viral immune factors mediate neuronal damage in people with HIV.
Introduction
HIV-associated neurocognitive disorders (HAND) comprise a spectrum of neurodegenerative pathologies that arise from the viral infection of the central nervous system (CNS) in subjects with chronic HIV infection. Despite the success of combination of antiretroviral therapy (cART), HAND remains a significant clinical challenge in up to 50 % of HIV-infected individuals, with its adverse effects on patient survival, quality-of-life metrics, and activities of daily living [1]. While subjects on cART show low levels of systemic and CNS viral replication, the residual viral proliferation can induce the secretion of several pro-inflammatory cytokines and chemokines that, in turn, cause augmented neurological damage. Several chemokines and cytokines, including IP-10, IL-1α, IL-1β, and TNF-α, have been implicated in the etiology of HAND and detected at elevated levels in CSF and serum in patients suffering from HAND [2]. Although most studies primarily focused on macrophages and microglia as the source of secreted chemokines and cytokines, other reports demonstrated the migration of both HIV-1-infected T-cells and monocytes to the CNS, implicating their role in neuronal damage in HAND [3], 4].
This prompted us to employ peripheral blood mononuclear cells (PBMCs), consisting of both CD14 monocytes and T-cells, that were infected with relatively low titers of HIV-1 and examine the contribution of non-viral neurotoxic factors to HAND. Neurotoxic factors other than those of viral origin (e.g., Tat and gp120), are believed to be present in detectible but low levels in the CNS of people with HIV (PWH) who are virologically suppressed via cART. Previously, we demonstrated that intra- and inter-subtype genetic variations in HIV-1 Tat and gp120 influence neurovirulence [5], [6], [7] and we evaluated the effects on neurocognitive functions in a SCID-HIV encephalitis mouse model [8]. Using cell models, we monitored neurotoxicity, pro-inflammatory cytokine induction, and monocyte recruitment. Our previous comparative analyses demonstrated significantly higher levels of neurovirulence by HIV-1ADA (a representative neurovirulent strain of subtype B) compared to HIV-1IndieC1 (a representative non-neurovirulent strain of subtype C) [7], 8].
In this study, using HIV-infected PBMCs, we compared the relative effects of chronic inflammation and immune dysfunction mediated by HIV-1ADA and HIV-1Indie infections to HAND. Using spent media of PBMC infected with either of the viral strains, we ascertained the augmented neurotoxic properties of HIV-1ADA over HIV-1Indie. Additionally, after immunodepleting Tat and gp120 proteins from the spent media, we showed sustained, although reduced, neurotoxic properties of the HIV-1ADA, which we ascribe to pro-inflammatory cytokines and chemokines secreted into the media. Furthermore, we demonstrated that a higher level of induced secretion of pro-inflammatory cytokines and chemokines by treating PBMC with recombinant Tat proteins derived from HIV-1 subtypes B when compared to that from subtype C. Thus, from our data, a pre-deterministic role of Tat in promoting neurotoxicity is evident and this property appears to be subtype-specific.
Materials and methods
Materials
PBMCs were purified from leukopaks from New York Blood Center. SHSY-5Y neuroblastoma cells were obtained from ATCC (Catalog number – CRL-2266) and maintained in a 1:1 mixture of Eagle’s Minimum Essential Medium and F12 Medium with 10 % serum. Recombinant purified Tat proteins were from HIV-1BL43 [9], a subtype C virus and HIV-1YU2, a subtype B virus and were generated as described previously [9].
Isolation of peripheral blood mononuclear cells
Peripheral blood mononuclear cells (PBMCs) were isolated from HIV-1 negative donor leukopaks (New York Blood Center) using density gradient centrifugation. Leukopaks were diluted in RPMI/FBS and overlaid over a Ficoll-Hypaque (Sigma-Aldrich, St. Louis, MO) gradient. Following centrifugation at 400×g for 30–40 min at room temperature with no brakes, PBMCs were recovered from the Ficoll-Hypaque interface, diluted in PBS, and centrifuged at 200×g for 10 min at room temperature to eliminate platelets. PBMCs were re-suspended in Gibco RPMI1640 (ThermoFisher), supplemented with 10 % FBS (Gibco) and 1 % Penicillin and Streptomycin. Cells were incubated at 37 °C in T75 flasks.
HIV-1 infection of PBMCs
PBMCs were activated with PHA (5 μg/ml) and IL-2 (100 IU/ml) for 48 h prior to infection. HIV-1ADA and HIV-1IndieC1 virus stocks were generated by transfecting the HEK 293T cells with infectious molecular clone DNAs. Virus released into the medium was quantitated by the p24 Enzyme-linked immune-sorbent assay (ELISA; Applied Biosystems, Inc. Waltham, MA), as described previously [6]. Approximately 50 × 106 PBMCs were infected with viral stocks at low multiplicity of infection (5 and 10 ng of HIV-1ADA and HIV-1IndieC1 respectively) to achieve comparable levels of viral infectivity at the assay end point 4-h or five days following infection as described previously [6].
Immunodepleting Tat and gp120
HIV-1 Tat and gp120 were immuno-depleted from HIV-infected spent media from PBMCs using broadly neutralizing anti-gp120 (VRC 03, NIH AIDS Reagent Repository) and anti-Tat E1.1 [9] antibodies, as described previously [6], 7]. VRC03 was chosen because of its broadly neutralizing capacity and its availability and E1.1 monoclonal antibodies were selected because of their ability to neutralize both subtype B and C Tat proteins [7]. E1.1 mAbs were raised in BALB/c mice using recombinant full-length Tat protein. E1.1 mAb was of IgG1 isotype. Immune characterization showed that E1.1 displayed a Tat binding affinity, with a Kd value of 1.9 nM and in Tat-neutralization properties in preventing the Tat-mediated transactivation in HLM-1 cells (Dr. Ranga, unpublished results).
For immunodepletion, Pansorbin beads (Millipore Sigma, Burlington, MA; Cat no. 507861) were incubated with anti-gp120 and anti-Tat antibodies for an hour on ice. Antibodies were used in excess, at 3-fold (for VRC03) or 100-fold excess (for E1.1), concentrations (VRC03 and E1.1 antibodies were both used at 300 ng respectively) to ensure complete protein depletion from spent media. The beads were centrifuged, re-suspended in 25 μL RPMI, and added to 10 ml of spent media. Following incubation for an hour on ice, the Pansorbin beads are centrifuged at 3,000 rpm for 5 min at 4 °C. The antigen-depletion step was repeated with a fresh round of antibodies to ensure the complete removal of gp120 and Tat proteins from the supernatant. The antibody-treated supernatants were used in the in vitro neurotoxicity assays.
Neurotoxicity assay
SH-SY5Y neuroblastoma cells were obtained from ATCC (CRL-2266) and maintained in a 1:1 mixture of Eagle’s Minimal Essential Medium and F12 Medium supplemented with 10 % fetal bovine serum. Approximately 250,000 cells/well were plated in 12-well plates and allowed to stabilize for three days. Three hundred µl (300 µL) of HIV-infected PBMC supernatants (with or without immuno-depletion) were added to 600 µL of SH-SY5Y media and then added to previously plated SH-SY5Y cells for 24 h as described previously [6]. Neuronal apoptosis and viability were assessed, in triplicate wells, using the WST-1 assay (Roche, Basel, Switzerland), which measures mitochondrial activity, providing a robust indication of cell viability and death. The percentage of cell death, compared to control cells treated with uninfected PBMC supernatant, was calculated as previously described [6].
To ensure comparable levels of viral particles in the spent media, we first measured virus replication via p24 ELISA in the HIV-1ADA and HIV-1IndieC1 infected PBMC cultures. This calibration allowed for equivalent exposure in the neurotoxicity assays. Viral release was standardized by infecting PBMCs with pre-determined virus inputs (5 and 10 ng of p24 for HIV-1ADA and HIV-1IndieC1, respectively) and confirmed by measuring p24 levels in the media at peak virus production points (day 15). These combined approaches ensured that observed neuronal death was attributable to the effects of viral strain differences and associated cytokine-chemokine profiles, rather than variability in infection parameters.
Luminex ELISA and CCL2 ELISA assays
Thirteen different cytokines/chemokines (Interferon-α, CCL-2, CCL-3, CCL-4, IL1-α, IL-1β, IL-4, IL-6, IL-8, IL-10, TNF-α, IP-10 and RANTES) were measured using a high-sensitivity human cytokine array kit. Each sample was assayed in triplicate wells, and the cytokine standards supplied by the manufacturer were included on each plate. Data was acquired using a Luminex-100 system and analyzed using the Bio-Plex Manager software. As CCL2 levels were above the limit of detection of the cytokine array kit, a separate CCL-2 ELISA was performed using the Invitrogen Human CCL2 ELISA kit, as described previously [7], 8].
Tat treatment of PBMCs and real-time PCR
Purification of recombinant HIV-1 Tat proteins from the neurovirulent Subtype-B YU-2 strain and non-neurovirulent Subtype-C BL-43 strain has been reported previously [9]. Neurotoxic properties of these Tat proteins were described previously [10]. PBMCs of healthy subjects were activated with PHA (5 μg/ml) for 24 h, before treating them with 200 ng/ml recombinant Tat protein for 4 h at 37 °C. PBMCs were pelleted, and RNA was extracted using RNAeasy Kit (Qiagen, Germantown, Maryland). cDNA was generated from the RNA using a Taqman reverse transcriptase cDNA kit (Applied Biosystems Inc). cDNA was quantitated, and a qRT-PCR was performed using a Taqman cytokine gene expression SuperArray plate (Applied Biosystems Inc). The delta delta-CT method was used to calculate fold differences between treatments, using GAPDH as a control house-keeping gene. RNA extracted from PBMCs treated with a buffer was used as a control, and the relative fold changes were determined in relation to control CT values.
Statistical analysis
Statistical significance was determined using one-way ANOVA followed by post hoc tests to identify specific group differences, with p-values indicated in the figure legends to denote significance levels.
Results
Contribution of non-viral factors to HIV neuropathogenesis in vitro
To understand the contribution of non-viral immune factors to the pathogenesis underlying HAND, we investigated whether HIV-spent media immunodepleted for viral Tat and gp120 proteins could still retain neurotoxicity. PBMCs of healthy donors were infected with the neurovirulent HIV-1ADA (5 ng/ml p24) or the non-neurovirulent HIV-1Indie (10 ng/ml p24) strain to achieve comparable levels of viral infection five days after infection. Following viral infection, p24 levels in the spent media were quantitated using a commercial p24 assay. Spent media of HIV-1ADA and HIV-1Indie contained 25 ng/ml and 22 ng/ml of p24, respectively, confirming comparable levels of infection. Spent media were then incubated with VRC03 and E1.1 antibodies to deplete gp120 and Tat antigens from the samples, respectively. Subsequently, undifferentiated SHSY-5Y neuroblastoma cells were treated with immuno-depleted supernatants or an uninfected control medium. Following immuno-depletion of Tat and gp120 (the two primary viral factors responsible for neurotoxicity), the neurovirulent HIV-1ADA isolate retained about half of its neurotoxicity compared to the non-neurovirulent HIV-1IndieC1 isolate (Figure 1), that exhibited barely any change in its already low overall neurotoxicity (Figure 1 inset). This result pointed at host factors other than viral proteins, such as pro-inflammatory cytokines and chemokines, as the potential source of cytotoxicity of SHSY-5Y cells.
![Figure 1:
Contribution of cytokines and chemokines to neurotoxicity: 300 µl of the spent media of HIV-1ADA- or HIV-1Indie-infected PBMCs were incubated with pooled VRC03 and E1.1 (anti-gp120 and anti-Tat, respectively) antibodies for 24 h to deplete both gp120 and Tat, via two rounds of immunodepletion. Neuronal SH-SY5Y cells were incubated with the spent media, with or without (inset) immuno-depletion. WST-1 cell viability was performed using the WST-1 assay (Inset-adapted from reference # [6]). This experiment was performed 3 times.](/document/doi/10.1515/nipt-2024-0020/asset/graphic/j_nipt-2024-0020_fig_001.jpg)
Contribution of cytokines and chemokines to neurotoxicity: 300 µl of the spent media of HIV-1ADA- or HIV-1Indie-infected PBMCs were incubated with pooled VRC03 and E1.1 (anti-gp120 and anti-Tat, respectively) antibodies for 24 h to deplete both gp120 and Tat, via two rounds of immunodepletion. Neuronal SH-SY5Y cells were incubated with the spent media, with or without (inset) immuno-depletion. WST-1 cell viability was performed using the WST-1 assay (Inset-adapted from reference # [6]). This experiment was performed 3 times.
Differential induction of cytokines and chemokine by HIV-1ADA and HIV-1IndieC1
In order to examine the types of cytokines that were induced by the infection of subtypes B and C HIV-1, we next infected PBMCs with HIV-1ADA or HIV-1IndieC1. Upon infection of PBMCs, HIV-1ADA induced significantly higher levels of pro-inflammatory cytokines IL-1α, IL-1β, IL-6 and TNF-α as measured via Luminex assay (Figure 2). These factors have been implicated [6], [11], [12], [13], [14], [15] in causing neuronal damage in various neuroimmune disorders. In contrast, HIV-1IndieC1 induced a higher expression level of IL-8, which is an exception in not being a neurotoxic cytokine. Further, HIV-1ADA also induced higher levels of CC family of chemokines, CCL2, CCL3, and CCL4, and also IP-10 (CXCL-10), a CXC family chemokine. Although the CC-chemokines are conditionally neuroprotective [16], 17], at higher levels, they can promote increased immune cell trafficking to the brain, leading to augmented neuronal damage. Furthermore, HIV-1ADA infection of PBMCs also resulted in a more significant down-regulation of anti-inflammatory cytokines IL-4 and IL-10, compared to HIV-1IndieC1-infection or control uninfected sample. Additionally, HIV-1IndieC1 also suppressed the induction of IL-10 to a greater extent than that of HIV-1ADA. These results suggest that the cytokines and chemokines are vital in causing neurotoxicity, which is subjective to subtype-specific differences.

Differential induction of chemokines and cytokines upon HIV-1 infection of PBMCs. A panel of thirteen cytokines/chemokines (Interferon-α, CCL-2, CCL-3, CCL-4, IL-1α, IL-1β, IL-4, IL-6, IL-8, IL-10, TNF-α, IP-10, and RANTES) was measured in the spent media from HIV-1ADA- or HIV-1Indie-infected PBMCs, using a high-sensitivity human cytokine array kit. Each sample was assayed in triplicate wells, and data was acquired using a Luminex-100 system and analyzed using the Bio-Plex Manager software. This experiment was performed 3 times, each time with triplicates.
Tat plays a key role in the differential induction of chemokines and cytokines
It is well-known that HIV-1 Tat protein transactivates numerous cellular genes in addition to HIV-1 LTR. In fact, Tat from subtypes B and C are known to differentially transactivate cytokine genes in primary monocytes [18]. In order to test whether the differential neurotoxicities of spent media from the PBMCs infected with HIV-1ADA and HIV-1IndieC1 are likely a reflection of the types of cytokines induced by Tat protein in PBMCs, PBMCs were treated with recombinant Tat proteins from HIV-1YU-2 and HIV-1BL-43 strains representing subtypes B and C, respectively. After a 4-h treatment, RNA was extracted from PBMCs, and gene expression changes were compared using quantitative real-time PCR on a macroarray of selected cytokine/chemokine genes. The neurovirulent HIV-1YU-2 Tat protein induced 5- to 30-fold increases in the level of pro-inflammatory cytokine transcripts compared to the control or the non-neurovirulent HIV-1BL-43 Tat (Figure 3). Similar elevated patterns for subtype B Tat-treated PBMCs were observed with neurotoxic host factors, such as TNF-α and IP-10.

Differential secretion of cytokines and chemokines by PBMC treated with Tat: PBMCs of healthy donors were treated with 200 ng/ml of recombinant Tat proteins of HIV-1YU-2 (clade B) or HIV-1BL-43 (clade C) viruses. Transcription of selected cytokine and chemokine genes was evaluated using real-time PCR. This experiment was performed two times with triplicates.
The Tat-induced patterns of the anti-inflammatory cytokines, IL-4 and IL-10, differed from those of the pro-inflammatory cytokines. Both Tat proteins exerted a suppressive effect on the expression of the two anti-inflammatory cytokines, compared to a ‘no Tat’ control. Importantly, HIV-1BL-43 Tat exerted a weaker inhibitory effect on the secretion of these two anti-inflammatory cytokines than the HIV-1YU-2 Tat, thus inducing still significantly higher levels of these host factors. Thus, compared to subtype B, subtype C Tat induced relatively low levels of pro-inflammatory cytokines and relatively higher levels of anti-inflammatory cytokines, resulting in an overall lower potential for neurotoxicity
Discussion
Neurodegeneration persists in HIV-infected individuals despite cART and viral suppression due to multifactorial etiopathogenesis (Winston and Spudich, 2020). Neurotoxic cytokines and chemokines secreted by HIV-1 infected cells and bystander cells play a central role in developing neuronal damage observed in HAND [2]. Using in vitro HIV-infection of macrophages and the measurement of neurocognitive functions in SCID-HIVE mice, we previously established differential neurovirulent properties of HIV-1 B and C subtypes and furthermore demonstrated that subtype-specific genetic variations in Tat or gp120 primarily underlie the observed differences in neurotoxicity.
A potential role of non-viral factors in neuroinflammation
The spent media of virus-infected PBMC retained neurotoxic properties even after depleting Tat and gp120, suggesting an important role for immune dysregulation in the causation of adverse CNS outcomes. Using the highly sensitive Luminex assay, several pro- and anti-inflammatory cytokines and chemokines could be measured in such media. Importantly, the neurovirulent subtype B strain up-regulated the cytokines higher than the non-neurovirulent subtype C strain. Consistent with these results, differential expression of the same cytokines was also shown in PBMCs treated with recombinant Tat proteins.
The analysis of chemokines and cytokines in Figures 2 and 3 highlights distinct inflammatory profiles associated with subtype B (HIV-1ADA/HIV-1YU2) and subtype C (HIV-1IndieC1/HIV-1BL43). In Figure 2, the depleted media from PBMCs infected with subtype B HIV-1ADA shows significantly higher levels of pro-inflammatory chemokines (CCL2, CCL3, CCL4, and CXCL10/IP-10) and cytokines (IL-1α, IL-1β, IL-6, TNF-α) compared to subtype C HIV-1IndieC1, which induces a less pronounced inflammatory response. Subtype B also suppresses anti-inflammatory cytokines (IL-4 and IL-10) to a greater extent, shifting the balance toward a more pro-inflammatory state. The results in Figure 3, where recombinant Tat proteins were used, reveal subtype-specific effects of Tat proteins with HIV-1B Tat inducing significantly higher levels of pro-inflammatory markers, including CCL2, CCL3, CCL4, IL-1β, IL-6, and TNF-α, compared to subtype C Tat. Both Tat proteins suppress IL-4 and IL-10, but subtype C Tat exhibits a weaker suppressive effect, potentially altering the intensity of inflammation. The comparison underscores that subtype B HIV-1 strains (ADA/YU2) drive a more aggressive pro-inflammatory and neurotoxic response than subtype C strains (IndieC1/BL43), reflecting their differential contributions to neuropathogenesis.
Both purified Tat proteins and HIV-infection display subtype-specific differences
Our observations with HIV-infected PBMCs correlated well with the findings using recombinant purified Tat proteins added to PBMCs. Subtype C Tat protein induced low levels of pro-inflammatory cytokines than subtype B Tat protein and higher levels of anti-inflammatory cytokines. Furthermore, previously published studies [17], [18], [19] on IL-10 levels induced by the subtype B and C Tat proteins in monocytes have yielded inconsistent results suggesting the involvement of additional viral or host factors affecting IL-10 regulation. It is well known that Tat protein is secreted and is taken up by bystander cells [2]. In the uninfected bystander cells, HIV-1 Tat can transcriptionally transactivate proinflammatory cytokine genes. We previously showed that the differential uptake of Tat B and C proteins is likely the basis of differential transactivation of such inflammatory host genes and the resulting differential neuroinflammation [20]. These observations indicate that differences between neurovirulent and non-neurovirulent HIV-1 strains are reflected in their Tat protein, inducing the differential regulation of pro- and anti-inflammatory chemokines/cytokines in PBMCs.
Among the factors up-regulated, both IL-1α and IL-1β have been implicated in causing and enhancing neuronal damage [21]. These variant forms of Interleukin-1 act through the IL-1 receptor. Several models of brain injury demonstrated that IL-1 receptor agonist (IL-1Rα) reduces neuronal damage caused by IL-1α and IL-1β [11]. IL-6 has been implicated in enhancing NMDA-mediated neurotoxicity, leading to increased membrane depolarization [22]. In the CNS, TNF-α orchestrates a diverse array of functions, including but not limited to immune surveillance, cellular homeostasis, and protection against specific neurological insults. However, in the context of HIV-1 infection, TNF-α enhances HIV-1 Tat-induced neurotoxicity by oxidative stress leading to neuronal apoptosis [23]. TNF-α can also inhibit glutamate uptake by astrocytes, leading to an extracellular buildup of glutamate, causing neuronal excitotoxicity [24]. Furthermore, inhibiting TNF-α activity abrogates HIV-1-induced neurotoxicity in vitro [25]. IL-1β and TNF-α dysregulate glutamate production in neurons through the induction of glutaminase [15]. IP-10, while serving as a T cell chemoattractant, can cause neurotoxicity in primary neurons due to elevated intracellular calcium levels [12], leading to caspase activation and neuronal death [26].
We found that infection of PBMC by the neurovirulent HIV-1ADA and treatment of PBMC by recombinant Tat of HIV-1YU-2 strain, both up-regulated chemokines CCL-2, CCL-3, and CCL-4 to significant levels. CCL-2 (MCP-1), although typically neuroprotective [16], can cause monocyte migration to the CNS [27], thus exacerbating neuronal damage. Likewise, CCL-3 (MIP1α) and CCL-4 (MIP1β) also can enhance the migration of immune cells of diverse lineages to the CNS [28]. All the processes can contribute to increased chronic inflammation and progressive neuronal damage, finally manifesting as HAND. Neuronal injury is further augmented by the suppression of anti-inflammatory chemokines, IL-4 and IL-10, as shown in vitro in neuronal cultures and in vivo in patients. Despite viral suppression with cART, indirect neuronal damage is persistent and is mediated by upregulated inflammatory cytokines and chemokines, particularly in individuals with chronic HIV infection. Concomitant up-regulation of several pro-inflammatory cytokines, such as TNF-α and IL-1α, and down-regulation of anti-inflammatory cytokines, such as IL-4 and IL-10, have been documented in HIV-1 patients suffering from HAND [13].
Our findings suggest that in addition to using cART to control viral replication, adjunctive therapies incorporating neuroprotective agents, such as minocycline [29], 30] and memantine [31] might be beneficial in suppressing CNS inflammation. Alternative approaches to reducing CNS inflammation by targeting leukocyte trafficking across the blood-brain barrier with Natalizumab, an anti-alpha-4-integrin monoclonal antibody [32], 33] can potentially improve CNS outcomes in people with HIV. Additionally, novel therapies targeting HIV-1 Tat function [34] could reduce overall neuronal damage, both from the direct neurotoxicity of Tat and the up-regulation of pro-inflammatory cytokines and chemokines induced by Tat. Although substantial in vitro and in vivo evidence demonstrates attenuation of neuronal damage using adjunctive approaches, therapeutic interventions targeting CNS dysfunction in HIV-infected individuals remain clinically ineffective, necessitating further research to develop interventions that can improve patient outcomes and quality of life.
Funding source: National Institutes of Health
Award Identifier / Grant number: R01 AI153008
Award Identifier / Grant number: R01 MH083570
Award Identifier / Grant number: T32 AI007501
Acknowledgments
The authors wish to thank the Einstein-Rockefeller-CUNY Center for AIDS research for use of Biomarkers and Advanced Technologies core services; Arthur Ruiz and David Ajasin for critically reading the manuscript and the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH for providing valuable reagents.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: VR generated the neurotoxicity data, helped draft the original manuscript and helped develop the overall content, MR generated the data on the infection of PBMCs and measurement of cytokine and chemokine transcript levels, SB purified the Tat proteins and generated the data on the cytokine protein levels from PBMCs treated with Tat, UR supervised the work on Tat purification and cytokine protein quantitation and for the manuscript revisions, VR helped develop the overall content, supervised most of the work reported here and contributed to drafting the original manuscript. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: The results reported in this manuscript were supported by NIH grants R01 MH083570 and R01 AI153008 to VRP. MAR wishes to acknowledge support from the institutional training grant NIH T32 AI007501.
-
Data availability: Not applicable.
References
1. Saylor, D, Dickens, AM, Sacktor, N, Haughey, N, Slusher, B, Pletnikov, M, et al.. HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment. Nat Rev Neurol 2016;12:234–48. https://doi.org/10.1038/nrneurol.2016.27.Suche in Google Scholar PubMed PubMed Central
2. Rao, VR, Ruiz, AP, Prasad, VR. Viral and cellular factors underlying neuropathogenesis in HIV associated neurocognitive disorders (HAND). AIDS Res Ther 2014;11:13. https://doi.org/10.1186/1742-6405-11-13.Suche in Google Scholar PubMed PubMed Central
3. Coffin, J, Swanstrom, R. HIV pathogenesis: dynamics and genetics of viral populations and infected cells. Cold Spring Harbor Perspect Med 2013;3:a012526. https://doi.org/10.1101/cshperspect.a012526.Suche in Google Scholar PubMed PubMed Central
4. Schnell, G, Joseph, S, Spudich, S, Price, RW, Swanstrom, R. HIV-1 replication in the central nervous system occurs in two distinct cell types. PLoS Pathog 2011;7:e1002286. https://doi.org/10.1371/journal.ppat.1002286.Suche in Google Scholar PubMed PubMed Central
5. Ranga, U, Shankarappa, R, Siddappa, NB, Ramakrishna, L, Nagendran, R, Mahalingam, M, et al.. Tat protein of human immunodeficiency virus type 1 subtype C strains is a defective chemokine. J Virol 2004;78:2586–90. https://doi.org/10.1128/jvi.78.5.2586-2590.2004.Suche in Google Scholar PubMed PubMed Central
6. Rao, VR, Neogi, U, Eugenin, E, Prasad, VR. The gp120 protein is a second determinant of decreased neurovirulence of Indian HIV-1C isolates compared to southern African HIV-1C isolates. PLoS One 2014;9:e107074. https://doi.org/10.1371/journal.pone.0107074.Suche in Google Scholar PubMed PubMed Central
7. Rao, VR, Neogi, U, Talboom, JS, Padilla, L, Rahman, M, Fritz-French, C, 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. https://doi.org/10.1186/1742-4690-10-61.Suche in Google Scholar PubMed PubMed Central
8. Rao, VR, Sas, AR, Eugenin, EA, Siddappa, NB, Bimonte-Nelson, H, Berman, JW, et al.. HIV-1 clade-specific differences in the induction of neuropathogenesis. J Neurosci 2008;28:10010–6. https://doi.org/10.1523/jneurosci.2955-08.2008.Suche in Google Scholar PubMed PubMed Central
9. Siddappa, NB, Venkatramanan, M, Venkatesh, P, Janki, MV, Jayasuryan, N, Desai, A, et al.. Transactivation and signaling functions of Tat are not correlated: biological and immunological characterization of HIV-1 subtype-C Tat protein. Retrovirology 2006;3:53. https://doi.org/10.1186/1742-4690-3-53.Suche in Google Scholar PubMed PubMed Central
10. Mishra, M, Vetrivel, S, Siddappa, NB, Ranga, U, Seth, P. Clade-specific differences in neurotoxicity of human immunodeficiency virus-1 B and C Tat of human neurons: significance of dicysteine C30C31 motif. Ann Neurol 2008;63:366–76. https://doi.org/10.1002/ana.21292.Suche in Google Scholar PubMed
11. Rothwell, N. Interleukin-1 and neuronal injury: mechanisms, modification, and therapeutic potential. Brain Behav Immun 2003;17:152–7. https://doi.org/10.1016/s0889-1591(02)00098-3.Suche in Google Scholar PubMed
12. Sui, Y, Potula, R, Dhillon, N, Pinson, D, Li, S, Nath, A, et al.. Neuronal apoptosis is mediated by CXCL10 overexpression in simian human immunodeficiency virus encephalitis. Am J Pathol 2004;164:1557–66. https://doi.org/10.1016/s0002-9440(10)63714-5.Suche in Google Scholar PubMed PubMed Central
13. Wesselingh, SL, Glass, J, McArthur, JC, Griffin, JW, Griffin, DE. Cytokine dysregulation in HIV-associated neurological disease. Adv Neuroimmunol 1994;4:199–206. https://doi.org/10.1016/s0960-5428(06)80258-5.Suche in Google Scholar PubMed
14. Yadav, A, Collman, RG. CNS inflammation and macrophage/microglial biology associated with HIV-1 infection. J Neuroimmune Pharmacol 2009;4:430–47. https://doi.org/10.1007/s11481-009-9174-2.Suche in Google Scholar PubMed PubMed Central
15. Ye, L, Huang, Y, Zhao, L, Li, Y, Sun, L, Zhou, Y, et al.. IL-1beta and TNF-alpha induce neurotoxicity through glutamate production: a potential role for neuronal glutaminase. J Neurochem 2013;125:897–908. https://doi.org/10.1111/jnc.12263.Suche in Google Scholar PubMed PubMed Central
16. Eugenin, EA, D’Aversa, TG, Lopez, L, Calderon, TM, Berman, JW. MCP-1 (CCL2) protects human neurons and astrocytes from NMDA or HIV-tat-induced apoptosis. J Neurochem 2003;85:1299–311. https://doi.org/10.1046/j.1471-4159.2003.01775.x.Suche in Google Scholar PubMed
17. Kitai, R, Zhao, ML, Zhang, N, Hua, LL, Lee, SC. Role of MIP-1beta and RANTES in HIV-1 infection of microglia: inhibition of infection and induction by IFNbeta. J Neuroimmunol 2000;110:230–9. https://doi.org/10.1016/s0165-5728(00)00315-5.Suche in Google Scholar PubMed
18. Gandhi, N, Saiyed, Z, Thangavel, S, Rodriguez, J, Rao, KV, Nair, MP. Differential effects of HIV type 1 clade B and clade C Tat protein on expression of proinflammatory and antiinflammatory cytokines by primary monocytes. AIDS Res Hum Retroviruses 2009;25:691–9. https://doi.org/10.1089/aid.2008.0299.Suche in Google Scholar PubMed PubMed Central
19. Wong, JK, Campbell, GR, Spector, SA. Differential induction of interleukin-10 in monocytes by HIV-1 clade B and clade C Tat proteins. J Biol Chem 2010;285:18319–25. https://doi.org/10.1074/jbc.m110.120840.Suche in Google Scholar PubMed PubMed Central
20. Ruiz, AP, Ajasin, DO, Ramasamy, S, DesMarais, V, Eugenin, EA, Prasad, VR. A naturally occurring polymorphism in the HIV-1 tat basic domain inhibits uptake by bystander cells and leads to reduced neuroinflammation. Sci Rep 2019;9:3308. https://doi.org/10.1038/s41598-019-39531-5.Suche in Google Scholar PubMed PubMed Central
21. Thornton, P, Pinteaux, E, Gibson, RM, Allan, SM, Rothwell, NJ. Interleukin-1-induced neurotoxicity is mediated by glia and requires caspase activation and free radical release. J Neurochem 2006;98:258–66. https://doi.org/10.1111/j.1471-4159.2006.03872.x.Suche in Google Scholar PubMed
22. Baune, BT, Konrad, C, Grotegerd, D, Suslow, T, Birosova, E, Ohrmann, P, et al.. Interleukin-6 gene (IL-6): a possible role in brain morphology in the healthy adult brain. J Neuroinflammation 2012;9:125. https://doi.org/10.1186/1742-2094-9-125.Suche in Google Scholar PubMed PubMed Central
23. Buscemi, L, Ramonet, D, Geiger, JD. Human immunodeficiency virus type-1 protein Tat induces tumor necrosis factor-alpha-mediated neurotoxicity. Neurobiol Dis 2007;26:661–70. https://doi.org/10.1016/j.nbd.2007.03.004.Suche in Google Scholar PubMed PubMed Central
24. Fine, SM, Angel, RA, Perry, SW, Epstein, LG, Rothstein, JD, Dewhurst, S, et al.. Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes. Implications for pathogenesis of HIV-1 dementia. J Biol Chem 1996;271:15303–6. https://doi.org/10.1074/jbc.271.26.15303.Suche in Google Scholar PubMed
25. Theodore, S, Cass, WA, Nath, A, Steiner, J, Young, K, Maragos, WF. Inhibition of tumor necrosis factor-alpha signaling prevents human immunodeficiency virus-1 protein Tat and methamphetamine interaction. Neurobiol Dis 2006;23:663–8. https://doi.org/10.1016/j.nbd.2006.05.005.Suche in Google Scholar PubMed
26. Mehla, R, Bivalkar-Mehla, S, Nagarkatti, M, Chauhan, A. Programming of neurotoxic cofactor CXCL-10 in HIV-1-associated dementia: abrogation of CXCL-10-induced neuro-glial toxicity in vitro by PKC activator. J Neuroinflammation 2012;9:239. https://doi.org/10.1186/1742-2094-9-239.Suche in Google Scholar PubMed PubMed Central
27. Eugenin, EA, Osiecki, K, Lopez, L, Goldstein, H, Calderon, TM, Berman, JW. CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigration of human immunodeficiency virus (HIV)-infected leukocytes across the blood-brain barrier: a potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci 2006;26:1098–106. https://doi.org/10.1523/jneurosci.3863-05.2006.Suche in Google Scholar PubMed PubMed Central
28. Mocchetti, I, Campbell, LA, Harry, GJ, Avdoshina, V. When human immunodeficiency virus meets chemokines and microglia: neuroprotection or neurodegeneration? J Neuroimmune Pharmacol 2013;8:118–31. https://doi.org/10.1007/s11481-012-9353-4.Suche in Google Scholar PubMed PubMed Central
29. Follstaedt, SC, Barber, SA, Zink, MC. Mechanisms of minocycline-induced suppression of simian immunodeficiency virus encephalitis: inhibition of apoptosis signal-regulating kinase 1. J Neurovirol 2008;14:376–88. https://doi.org/10.1080/13550280802199898.Suche in Google Scholar PubMed PubMed Central
30. Sacktor, N, Miyahara, S, Deng, L, Evans, S, Schifitto, G, Cohen, BA, et al.. Minocycline treatment for HIV-associated cognitive impairment: results from a randomized trial. Neurology 2011;77:1135–42. https://doi.org/10.1212/wnl.0b013e31822f0412.Suche in Google Scholar PubMed PubMed Central
31. Nath, A, Haughey, NJ, Jones, M, Anderson, C, Bell, JE, Geiger, JD. Synergistic neurotoxicity by human immunodeficiency virus proteins Tat and gp120: protection by memantine. Ann Neurol 2000;47:186–94. https://doi.org/10.1002/1531-8249(200002)47:2<186::aid-ana8>3.3.co;2-v.10.1002/1531-8249(200002)47:2<186::AID-ANA8>3.3.CO;2-VSuche in Google Scholar
32. Campbell, JD, McQueen, RB, Miravalle, A, Corboy, JR, Vollmer, TL, Nair, K. Comparative effectiveness of early natalizumab treatment in JC virus-negative relapsing-remitting multiple sclerosis. Am J Manag Care 2013;19:278–85.Suche in Google Scholar
33. Spudich, SS, Ances, BM. Neurologic complications of HIV infection: highlights from the 2013 conference on retroviruses and opportunistic infections. Top Antiviral Med 2013;21:100–8.Suche in Google Scholar
34. Darbinian, N, Khalili, K, Amini, S. Neuroprotective activity of pDING in response to HIV-1 Tat. J Cell Physiol 2014;229:153–61. https://doi.org/10.1002/jcp.24392.Suche in Google Scholar
© 2025 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Oligonucleotide therapeutics for neurodegenerative diseases
- Rheumatoid arthritis drugs and the risk of Parkinson’s disease – a meta-analysis
- Research Articles
- HIV-1 infection facilitates Alzheimer’s disease pathology in humanized APP knock-in immunodeficient mice
- Neurovirulent and non-neurovirulent strains of HIV-1 and their Tat proteins induce differential cytokine-chemokine profiles
- Cannabis use is associated with a lower likelihood of presence of HIV drug resistance mutations in a retrospective cohort of adults with HIV
- Sex dependent correlation of spleen atrophy and behavior deficits induced by binge treatment with ethanol in rodent models
- Brief Reports
- The 28th Scientific Conference of the Society on NeuroImmune Pharmacology: Charleston, SC – March 10-13, 2024
- The 29th Scientific Conference of the Society on NeuroImmune Pharmacology in Omaha, NE, June 8–12, 2025
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Oligonucleotide therapeutics for neurodegenerative diseases
- Rheumatoid arthritis drugs and the risk of Parkinson’s disease – a meta-analysis
- Research Articles
- HIV-1 infection facilitates Alzheimer’s disease pathology in humanized APP knock-in immunodeficient mice
- Neurovirulent and non-neurovirulent strains of HIV-1 and their Tat proteins induce differential cytokine-chemokine profiles
- Cannabis use is associated with a lower likelihood of presence of HIV drug resistance mutations in a retrospective cohort of adults with HIV
- Sex dependent correlation of spleen atrophy and behavior deficits induced by binge treatment with ethanol in rodent models
- Brief Reports
- The 28th Scientific Conference of the Society on NeuroImmune Pharmacology: Charleston, SC – March 10-13, 2024
- The 29th Scientific Conference of the Society on NeuroImmune Pharmacology in Omaha, NE, June 8–12, 2025