Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

IL-23 signaling in Th17 cells is inhibited by HIV infection and is not restored by HAART: Implications for persistent immune activation

  • Jason R. Fernandes,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Software, Validation, Writing – original draft

    Affiliations The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, Department of Biochemistry, Microbiology, and Immunology, The University of Ottawa, Ottawa, Ontario, Canada

  • Tamara K. Berthoud,

    Roles Writing – original draft, Writing – review & editing

    Affiliations The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, Department of Biochemistry, Microbiology, and Immunology, The University of Ottawa, Ottawa, Ontario, Canada

  • Ashok Kumar,

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision

    Affiliations Department of Biochemistry, Microbiology, and Immunology, The University of Ottawa, Ottawa, Ontario, Canada, The Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada

  • Jonathan B. Angel

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Validation, Writing – review & editing

    jangel@ohri.ca

    Affiliations The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, Department of Biochemistry, Microbiology, and Immunology, The University of Ottawa, Ottawa, Ontario, Canada, Division of Infectious Diseases, The Ottawa Hospital, Ottawa, Ontario, Canada

Abstract

Objectives

HIV infection causes a profound depletion of gut derived Th17 cells, contributing to loss of mucosal barrier function and an increase in microbial translocation, thus driving systemic immune activation. Despite normalization of circulating CD4+ T cell counts with highly active antiretroviral therapy (HAART), Th17 frequency and function often remain impaired. Given the importance of interleukin (IL)-23 in the generation and stabilization of Th17 cells we hypothesized that impaired IL-23 signaling causes persistent Th17 dysfunction in HIV infection.

Methods

The effects of in vitro HIV infection on responses to IL-23 in Th17 cells were examined. These included the production of IL-17, phosphorylated STAT3 (pSTAT3) and the transcription of retinoic acid orphan receptor C (RORC) gene. Blood derived Th17 cells from untreated and HAART-treated HIV-infected individuals were also examined for the IL-23 induced production of phosphorylated STAT3 (pSTAT3) and the expression of the IL-23 receptors.

Results

In vitro HIV infection significantly inhibited IL-17 production and IL-23 induced pSTAT3 while expression of RORC RNA was unaffected. Th17 cells isolated from untreated and HAART-treated HIV-infected individuals showed complete loss of IL-23 induced pSTAT3 without a decrease in the expression of the IL-23 receptors.

Conclusions

This study is the first to demonstrate an effect of HIV on the IL-23 signaling pathway in Th17 cells. We show that in vitro and in vivo HIV infection results in impaired IL-23 signaling which is not reversed by HAART nor is it a result of reduced receptor expression, suggesting that HIV interferes with IL-23-activated signaling pathways. These findings may explain the inability of HAART to restore Th17 frequency and function and the resulting persistent chronic immune activation observed in HIV infected individuals.

Introduction

Among the CD4+ T cells in gut associated lymphoid tissue (GALT), the Th17 subset has been identified as a critical regulator of homeostasis and antimicrobial defense [13]. Found predominantly at mucosal surfaces, Th17 cells secrete a unique spectrum of cytokines that help co-ordinate adaptive and innate immune responses [47], and have direct effects on mucosal epithelial cells [8] that act to maintain normal mucosal homeostasis. Studies of HIV-infected individuals and SIV-infected rhesus macaques have demonstrated that the early phases of SIV and HIV infection are characterized by massive losses of Th17 cells from the GALT [914], facilitated by the fact that HIV preferentially infects CD4+ T cells that express the Th17 cell marker CCR6 [15]. Loss of GALT Th17 cells is associated with microbial translocation, permeability to intestinal pathogens, and damage to the mucosal epithelium [12,1618]. Thus, Th17 deficiency is a major contributor to the systemic immune activation typical of chronic HIV infection. Despite the ability of highly-active antiretroviral therapy (HAART) to suppress viral replication and restore peripheral CD4+ T cell counts, the recovery of Th17 cells in the GALT is frequently incomplete [11,1921].

Mouse studies have shown that terminal Th17 differentiation is dependent on chromatin remodeling of the IL-17 gene which is regulated by IL-23 [2224], a recently described IL-12 cytokine family member. However in humans, IL-23 is believed to act by maintaining and expanding already-differentiated Th17 cells [23,2529]. IL-23 signals through a heterodimeric receptor composed of the IL-12 receptor, beta 1 (IL-12Rβ1) chain and a unique IL-23 receptor (IL-23R) chain [30]. IL-23 signaling through its receptor requires tyrosine kinase 2 (TYK2) and Janus kinase 2 (JAK2) activity [30], and results in phosphorylation of Signal transducer and activator of transcription 3 (STAT3) which then binds to the IL-17 promoter [3133], resulting in expression of IL-17. STAT3 phosphorylation also promotes transcription of the RAR related orphan receptor C (RORC) gene, which encodes the Th17-specific transcriptional regulators RORγt and RORα [3436], and upregulates IL-23R and STAT3 transcription in an autocrine fashion [37,38]. Th17 cells can be programmed away from IL-17 production towards secretion of other cytokines [3941], thus, IL-23 seems to perform a critical role in maintaining the key characteristics by which Th17 cells are identified transcriptionally and functionally.

Although HAART enables control of viral replication in the periphery, evidence suggests that viral suppression in GALT is highly variable [19]. Thus, even in well suppressed patients, ongoing viral replication in the gut may limit recovery of Th17 cells. Recently, HIV was shown to change the cytokine secretion profile of Th17 cells in the absence of overt cell death, suggesting that HIV infection may also cause Th17 dysfunction [42]. Although IL-23 has a demonstrated impact on maintaining human Th17 cell function, little is known about how HIV infection may affect the ability of IL-23 to maintain Th17 activity or key signaling pathways and transcription factors activated downstream of IL-23. We therefore sought to determine whether HIV inhibits the responsiveness of human Th17 cells to IL-23, thus contributing to ongoing Th17 deficits in HAART-treated patients.

Materials and methods

Study participants

All research on human blood was approved by the Ottawa Health Sciences Network Research Ethics Board. All participants provided written consent prior to participation in the study. Blood was collected from healthy volunteers, HAART-treated or untreated HIV infected individuals in heparin-containing tubes. Blood drawn from untreated individuals was collected either at a initial clinical visits at a pre-treatment time point or from individuals who had interrupted treatment. The clinical characteristics of HIV-infected patients are listed in Table 1.

thumbnail
Table 1. Clinical characteristics of HIV-infected study subjects.

https://doi.org/10.1371/journal.pone.0186823.t001

Blood Th17 cell isolation and Th17 generation from naïve CD4+ T cells

Peripheral blood mononuclear cells (PBMC) were isolated from blood by Ficoll-Paque Plus (GE Healthcare) density gradient centrifugation. Blood Th17 cells, defined as CD4+ CXCR3-CCR6+ as previously described [43,44], were isolated from PBMC by magnetic isolation using the a human Th17 cell enrichment kit according to the manufacturer’s instructions (Stemcell Technologies). Briefly, unwanted cells were extracted by negative selection using EasySep Human CD4+CXCR3- T Cell Pre-Enrichment Cocktail. Tetrameric Antibody Complexes (TAC) recognizing CD8, CD14, CD16, CD19, CD20, CD36, CD56, CD66b, CD123, TCRγ/δ, glycophorin A, CD45RAhigh, CXCR3 and dextran-coated magnetic particles, were added to PBMC and the undesired cells separated out with a magnet. Following this step which produces a highly enriched population of CD4+ T cells, CCR6+ cells were enriched by positive selection. TAC recognizing CCR6 and dextran-coated magnetic particles were added to the cells and the labeled cells then separated with a magnet. The enriched Th17 cells were then rested for 12hrs at 37 degrees to ensure the dextran-coated magnetic particles had completely dissociated from the Th17 cells. The enriched Th17 cells are known not to be activated by the enrichment process, IFN-γ and IL-17 production following the enrichment process was analyzed by StemCell and shown to be negligible without further stimulation (Magdalena Maslowski, Stem Cell, personal communication).

For some experiments, Th17 cells were generated in vitro from peripheral blood CD4+ T cells as previously described [45]. CD4+ T cells were isolated from PBMC using the CD4 positive selection kit (Stemcell Technologies) and activated using the T cell activation and expansion kit (Miltenyi Biotec) in RPMI-1640 supplemented with IL-1β (10 ng/mL) and IL-6 (50 ng/mL) and in the presence of neutralizing antibodies against IL-4 and IFN-γ (both at 10 μg/mL). After 5 days of priming the cells were washed and placed back into culture in RPMI-1640 supplemented with IL-2 (20 IU/mL) and IL-23 (20 ng/mL) for 7 days. All cytokines and neutralizing antibodies were purchased from R&D Systems, Inc.

HIV infection of Th17 cells

In vitro HIV infection of Th17 cells was performed using a dual-tropic clinical HIV isolate grown in our laboratory (HIVCS204), or a mock preparation as a control. Cells were incubated with HIV (MOI = 0.1) for 24 hours in the absence of exogenous stimuli. Following the infection period, input virus was washed out with phosphate-buffered saline. The presence of HIV in these cultures was confirmed by PCR to detect HIV p24 DNA in infected cells. Viability of infected cultures was assessed by trypan blue exclusion and found to be >85% at time of analysis.

IL-17 intracellular cytokine staining

Th17 cells were stimulated with PMA (5 ng/mL) (Sigma-Aldrich) and ionomycin (50 ng/mL) (Sigma-Aldrich) in RPMI-1640 for 6 hours, in the presence of the protein transport inhibitor brefeldin A (Golgiplug) as per the manufacturer’s instructions (BD Biosciences). Following stimulation the cells were washed with 1% BSA-PBS and fixed and permeabilized using the Invitrogen Fix & Perm kit, according to the manufacturer’s instructions. Permeabilized cells were stained for 30 minutes at room temperature with anti-IL-17-Alexa Fluor 647 (BD Biosciences). Excess antibody was washed out and the cells were resuspended for analysis. A minimum of 30,000 events per condition were acquired on an FC500 flow cytometer (Beckman Coulter). Negative fluorescence cut-offs were defined using fluorescence minus-one (FMO) controls.

Measurement of cytokine secretion

HIV-infected or uninfected Th17 cells were activated with beads coated with monoclonal antibodies against CD3 and CD28 in the presence of IL-23 (20 ng/mL). Supernatants, collected following 3 days of activation and secretion of IL-17 by Th17 cells, were measured using the Ready-Set-Go IL-17 ELISA (R&D Systems). Developed ELISA plates were read using a SpectraMax 190 (Molecular Devices). Cytokine concentrations were determined using the regression line from a standard curve performed in triplicate with each assay. All samples were assayed in triplicate and IL-17 concentrations were expressed as ng/mL, mean ± SEM.

STAT3 phosphorylation analysis

For the detection of phosphorylated STAT3 (pSTAT3), enriched Th17 cells were cultured in RPMI-1640 alone or in the presence of either IL-23 or IL-6 (both at 50 ng/mL; R&D Systems) for 15 minutes at 37°C. The incubation time of 15 minutes was determined to be the optimal time for pSTAT3 expression. Incubation times longer than 15mins were not shown to further enhance the pSTAT3 detection in either HIV+ve or HIV-ve Th17 cultures. Following stimulation the cells were immediately fixed by the addition of paraformaldehyde at 37°C for 10 minutes. The cells were then washed and permeabilized on ice for 30 minutes using BD Phosflow buffer III (BD Biosciences). Following permeabilization, the cells were washed and stained with anti-pSTAT3-AlexaFluor 488 (BD Biosciences) for 30 minutes at room temperature, then washed, and resuspended for analysis. A minimum of 30,000 events in the lymphocyte scatter gate was collected for each sample on an FC500 flow cytometer (Beckman Coulter). Negative fluorescence cutoffs were defined using unstimulated controls. Data presented represent the proportion of cells that stain positive for pSTAT3.

qRT-PCR

RNA was isolated from cells using the RNeasy mini kit (Qiagen) and reverse transcribed into cDNA using the Invitrogen Superscript III First Strand Synthesis kit (Invitrogen). qRT-PCR reactions were set up using commercially available primers for the human RORC gene (Qiagen). 18S rRNA was amplified using the following specific primers: 5’-CTGCCATTAAGGGTGTGG-3’ [forward] and 5’-TCCATCCTTTACATCCTTCTG-3’ [reverse]. Reactions were prepared with iQ SYBR green supermix (Bio-Rad). Amplification was assessed by measuring the fluorescent signal of SYBR green using an iCycler Real-time PCR thermocycler (Bio-Rad). mRNA levels were calculated using the ΔΔCt method, normalized to S18 rRNA. Results are expressed as fold change relative to unstimulated, uninfected cells.

IL-23R staining

For the flow cytometric detection of IL-23R Th17 cells, enriched with the StemCell Th17 enrichment kit as previously described, were fixed in paraformaldehyde at room temperature for 20 minutes. The cells were washed and permeabilized in 0.05% saponin in PBS. IL-23R was stained using a rabbit polyclonal antibody (EMD Millipore), which stains for the cytoplasmic domain of the IL-23R, or a rabbit IgG isotype control (Jackson Immunoresearch) for one hour at room temperature. Excess antibody was washed out, and the bound primary antibody was stained using a donkey-anti-rabbit-AlexaFluor 488 conjugate for one hour at room temperature (Life Technologies). Excess antibody was washed out, and the cells were resuspended in PBS/BSA buffer for analysis. A minimum of 10,000 events in the lymphocyte gate were collected using an FC500 flow cytometer (Beckman Coulter). Negative cut-offs were defined using isotype controls.

Western blots

For detection of the IL-23 receptor protein IL-12Rβ1, blood Th17 cells were lysed in RIPA buffer in the presence of protease inhibitors for 30 minutes on ice, and centrifuged at 20,000 x g for 30 minutes at 4°C. Lysates were separated on a 12% polyacrylamide gel and transferred to PVDF membranes. IL-12Rβ1 protein was labeled using a monoclonal antibody (Santa Cruz Biotechnology) and detected with horseradish peroxidase-conjugated goat anti-rabbit IgG (R&D Systems). β-actin was similarly detected for use as a loading control. Images were acquired using an AlphaImager imaging system and AlphaView software (ProteinSimple).

Statistical analysis

Differences between in vitro infected/uninfected groups were tested using paired two-tailed Student’s t tests. Comparisons of healthy control and HIV patient data were tested using one-way ANOVA and Tukey’s multiple comparison tests. Data was analyzed using Graphpad Prism software v 6.0, with a P level < 0.05 considered to be statistically significant.

Results

In vitro HIV infection reduces IL-17 secretion and expression by Th17 cells

We examined the effects of in vitro HIV infection on the production of the canonical Th17 cytokine IL-17 by Th17 cells. Spontaneous IL-17 secretion by blood-derived Th17 enriched cells and in vitro-generated Th17 cells was minimal and was not significantly altered by HIV infection (Fig 1A). Infection with HIV significantly reduced anti-CD3/anti-CD28-induced secretion of IL-17 by blood-derived (p < 0.001) and in vitro-generated (p = 0.046) Th17 cells (Fig 1A). HIV-infected Th17 cells were stimulated with PMA/ionomycin in the presence of brefeldin A to determine the effect of HIV infection on the frequency of IL-17+ cells. Infection with HIV significantly impaired PMA/ionomycin induced expression of IL-17 by in vitro-generated Th17 cells (p = 0.013, Fig 1B/1C). Similar results were observed for blood-derived Th17 cells (p = 0.026, Fig 1B/1C). These results indicate that even short-term exposure to HIV impairs IL-17 expression by Th17 cells.

thumbnail
Fig 1. HIV reduces IL-17 secretion and intracellular expression in blood-derived and in vitro-generated Th17 cells.

Blood-derived and in vitro generated Th17 cells were infected with HIV for 24 hours. (A) Th17 cells were cultured for 3 days with anti-CD3 and anti-CD28 mAbs. Supernatants were harvested and assayed for secreted IL-17 by ELISA; * p < 0.001, n = 9; ** p = 0.045, n = 9. Data shown are mean ± SEM. (B) Representative histograms showing intracellular expression of IL-17 in Th17 cells stimulated for 6 hours with PMA and onomycin in the presence of brefeldin A. (C) Summary of the proportion of blood-derived and in vitro generated Th17 cells expressing IL-17 following PMA and ionomycin stimulation is shown with mean frequency indicated; + p = 0.026, n = 9, ++ p = 0.012, n = 5.

https://doi.org/10.1371/journal.pone.0186823.g001

HIV infection inhibits IL-23 induced phosphorylation of STAT3 in Th17 cells

IL-23 has been identified as a key cytokine for the generation and maintenance of human Th17 cells, and it has been hypothesized that lack of IL-23 signals leads to loss of Th17 function, and in particular the propensity to secrete IL-17[23,2529]. Phosphorylated STAT3 (pSTAT3) is the primary transcription factor associated with IL-23 signaling in Th17 cells [3133]. Phosphorylated STAT3 can also be induced through the binding of IL-6 to its receptor [46]. Therefore, to determine whether HIV infection influences IL-23 signaling in Th17 cells, HIV-infected cells were stimulated with IL-23 or IL-6 and pSTAT3 was assessed by flow cytometry.

Unstimulated cells expressed a negligible amount of pSTAT3 (1.8 ± 0.1% of cells stained positive for pSTAT3). Stimulation of uninfected Th17 cells with IL-23 induced a robust pSTAT3 response (57.8 ± 4.8%), was significantly reduced by in vitro HIV infection (32.7 ± 2.5%, p = 0.001, Fig 2A and 2B). In contrast, Th17 responsiveness to IL-6, which also induces pSTAT3, was comparable between uninfected (71.9 ± 3.9%) and HIV-infected Th17 cells (68.8 ± 3.9%, Fig 2A).

thumbnail
Fig 2. Phosphorylation of STAT3 within Th17 cells in response to IL-23 is inhibited by in vitro and in vivo HIV infection and is not restored by HAART.

Blood-derived Th17 cells from healthy donors were infected with HIV for 24 hours. Input virus was washed out, and cells were stimulated with IL-23 or IL-6 for 15 minutes. (A) Representative histograms showing STAT3 phosphorylation in response to IL-23 or IL-6 in HIV-infected and uninfected Th17 cells. (B) Summary of frequency of Th17 cells responding to IL-23 or IL-6 by phosphorylation of STAT3 (%pSTAT3+). * p = 0.001, n = 5. (C) Representative histograms demonstrating STAT3 phosphorylation following IL-23 (top) and IL-6 (bottom) stimulation in Th17 cells isolated from patients and healthy controls. (D) Summary of IL-23 induced pSTAT3 responses in circulating Th17 cells from HIV seronegative, HIV-infected untreated and HIV-infected HAART donors (* p < 0.001, n = 7) and IL-6-induced pSTAT3 responses in Th17 cells isolated from HIV infected donors.

https://doi.org/10.1371/journal.pone.0186823.g002

Th17 cells from HIV-infected individuals do not express phosphorylated STAT3 in response to IL-23

Having demonstrated that HIV interferes with IL-23 signaling in Th17 cells infected with HIV in vitro, we next examined whether circulating Th17 cells from HIV infected patients displayed similar IL-23 signaling defects. Circulating Th17 cells were enriched with the STEM cell Th17 enrichment kit and stimulated for 15 minutes with IL-23 or IL-6 and pSTAT3 was assessed by flow cytometry. IL-23 induced pSTAT3 was completely absent in circulating Th17 cells from untreated HIV-infected patients (2.1 ± 0.3% vs. 57.8 ± 4.8% in uninfected controls, p < 0.001 Fig 2C and 2D), suggesting that in vivo HIV infection interferes with the initial events in IL-23 signaling. Surprisingly, IL-23 induced pSTAT3 responses within circulating Th17 cells from HAART-treated patients was similarly diminished (3.7 ± 0.6%, p < 0.001; Fig 2C and 2D). Responses to IL-6 were not significantly different among untreated patients, treated patients and uninfected controls (65.3 ± 6.1%, 68.1 ± 9.8% and 73.2 ± 2.8%, respectively). These results are consistent with the in vitro results and demonstrate that HIV infection results in selective interference with IL-23 signaling in human Th17 cells while the cellular phosphorylation machinery related to STAT3 remains intact.

In vitro HIV infection has no effect on RORC expression

Central Th17 transcription factors RORα and RORγt are encoded by the RORC gene, the transcription of which is triggered by anti-CD3/CD28 stimulation [47]. To identify whether HIV inhibits expression of RORC, blood-derived Th17 cells were infected with HIV and stimulated for 3 days with beads coated with monoclonal antibodies against CD3 and CD28. Relative expression of RORC mRNA was determined by qRT-PCR. The transcription of RORC was not affected by infection with HIV (Fig 3).

thumbnail
Fig 3. In vitro HIV infection does not affect RORC, RNA levels in blood-derived Th17 cells.

Blood-derived Th17 cells were infected with HIV for 24 hours. Input virus was washed out and cells were stimulated with anti-CD3 and anti-CD28 mAbs for 3 days. mRNA was isolated and quantified by real-time qPCR for RORC genes. Relative mRNA levels were calculated using the ΔΔCt method, normalized to 18S rRNA. Results are expressed as fold change relative to unstimulated, uninfected cells. n = 9.

https://doi.org/10.1371/journal.pone.0186823.g003

IL-23 receptor expression is not downregulated in HIV infection

The IL-23 receptor complex is a heterodimer composed of the IL-12 receptor beta 1 chain (IL-12Rβ1) and a unique polypeptide chain called IL-23R [30]. HIV infection has previously been shown to alter cytokine responsiveness through multiple mechanisms [4851], and so it is possible that the observed hyporesponsiveness to IL-23 was due to downregulation of one or both of the receptor chains on circulating Th17 cells. In HIV-seronegative donors, nearly all circulating Th17 cells were found to express IL-23R (95.1 ± 1.4%), comparable to values observed in untreated HIV infected individuals (92.2 ± 2.6%) (Fig 4B). No decrease in the geometric mean fluorescence intensity (MFI) of the IL-23R expression on Th17 cells was found in the HIV infected untreated donors compared to the HIV seronegative donors. Expression of the IL-12Rβ1 chain was similarly unaffected in HIV seropositive individuals (Fig 4C). Taken together, these observations confirm that HIV-induced defects in IL-23 signaling are not due to lack of IL-23R or IL-12Rβ1 expression.

thumbnail
Fig 4. HIV infection does not downregulate IL-23R expression.

Circulating Th17 cells were isolated from HIV-seronegative or untreated HIV-infected individuals and expression of IL-23R was assessed by flow cytometry. (A) Representative histograms demonstrating IL-23R expression on circulating Th17 cells isolated from HIV-seronegative controls, HIV-infected untreated, HIV-infected HAART treated individuals and the matched IL-23R isotype control. (B) Summary of % cells expressing IL-23R on circulating Th17 cells from HIV seronegative and untreated HIV-infected patients. (C) Western blot demonstrating expression of IL-12Rβ1 on blood Th17 cell lysates from HIV-seronegative and HIV infected, untreated donors. Figure is representative of 3 out of 6 donors tested.

https://doi.org/10.1371/journal.pone.0186823.g004

Discussion

While Th17 cell expression and function are impaired in HIV infection, the mechanisms by which this occurs are yet to be identified. Here we describe for the first time the effect of HIV on the IL-23 signaling pathway in Th17 cells. The principle findings of this study were as follows: (1) expression of the canonical Th17 cytokine IL-17 is significantly inhibited by in vitro HIV infection; (2) both in vivo and in vitro HIV infection significantly reduce IL-23-induced pSTAT3 expression which is not reversed in the setting of effective HAART; and (3) the observed reduction in IL-17 is not due to the effect of HIV infection on RORC mRNA expression in Th17 cells nor its effect on the IL-23 receptor expression. The apparent irreversibility of the IL-23 signaling defects with effective HAART underscores the challenges in restoring mucosal homeostasis in HIV infected individuals and may explain in part why immune activation persists despite suppression of viral replication.

IL-17 production in response to TCR (anti-CD3/CD28) stimulation was shown to be significantly impaired by in vitro HIV infection. This is consistent with previous reports indicating that HIV proteins can cause reductions in CD3/CD28 activation of CD4+ T cells. In particular, the HIV accessory protein Nef interferes with the strength of CD3-mediated signaling [5254], potentially providing an explanation for the observed reduction in IL-17 secretion in response to TCR stimulation. However, this does explain the reduction in IL-17 expression induced by PMA/ionomycin, which bypasses the need for TCR activation.

HIV infection significantly reduced IL-23 induced pSTAT3 responses in both Th17 cells isolated from HIV infected individuals and HIV infected Th17 cultures. When stimulated with IL-6, the pSTAT3 responses in Th17 cells remained intact. These robust pSTAT3 responses to IL-6 indicate that STAT3 protein is present and functional within the in vitro-infected Th17 cells, as well as the Th17 cells isolated from HIV patients. A greater reduction in the pSTAT3 responses to IL-23 was detected in the Th17 cells isolated from HIV infected patients compared to the Th17 cells cultured with HIV (Fig 2B and 2D). Host and environmental factors such as the cytokine milieu may have contributed to the differences seen.

The IL-23 receptor (IL-23R and IL-12Rβ1) also remained unaffected by HIV infection indicating that receptor downregulation is not the cause of impaired IL-23 responses. HIV may therefore be interfering with intracellular signaling events downstream of the IL-23 receptor, but upstream of STAT3 phosphorylation.

On binding to the IL-23 receptor complex, IL-23 induces the phosphorylation of JAK2 and TYK2 with which the IL-23 receptor is constitutively associated [30], leading to the phosphorylation of STAT3 [55]. It is possible that HIV may interfere with the phosphorylation of JAK-2 and TYK-2 proteins and explain the reduction of IL-23 induced pSTAT3 we observed. However while IL-23 has been shown to induce the phosphorylation of JAK-2 and TYK-2 in human CD4 T-cell clones [30], IL-23 induced phosphorylation of JAK-2 and TYK-2 has not been documented in human primary Th17 cells. In fact, in our model, IL-23 was unable to induce phosphorylation of JAK-2 and TYK-2.

HIV may also induce negative regulators of the IL-23 signaling pathway. Recent studies in rhesus macaques by Bixler SL et al. showed that during acute SIV infection, IL-17 mRNA expression in CD4+ T cells was significantly reduced. This reduction was correlated with the increased expression of the negative regulators: protein inhibitor of activated STAT3 (PIAS3), Src Homology Phosphatase 2 (SHP2) and suppressor of cytokine signaling 3 (SOCS3)[56]. HIV infection has also been shown to induce SOCS1 and SOCS3 mRNA in CD4+ T cells in HIV infection [49], and the HIV trans-activator protein (tat) impairs IFN-γ expression by inhibiting STAT1 activation via a SOCS2-dependent pathway [57]. Thus, induction of negative regulators may contribute to an impaired potential to produce IL-17, though IL-23 is not known to induce such negative regulation in Th17 cells.

The RORC gene products, RORγt and RORα, promote Th17 differentiation [58] and IL-17 production [59]. Although RORC mRNA levels are not altered by HIV infection, it is still possible that HIV induces protein-protein interactions that limit the activity of RORC gene products. In mice, the protein FoxP3 has been shown to inactivate RORγt [60] and a similar interaction has been described for RORα [61]. In humans, HIV infection has been shown to induce higher levels of FoxP3 expression in CD4+ T cells. HIV infection has also been shown to increase STAT1 levels in human T-cells [62], and to induce STAT1 activation in monocyte-derived macrophages and human brain microvascular endothelial cells (HBMECs) [63,64]. STAT1 induction then leads to the inhibition of RORγt via the transcription of T-bet. Thus HIV infection may induce the expression of host proteins that antagonize transcription factors involved in the control of Th17 differentiation and function.

In the context of HIV infection, persistent immune activation has been implicated in much of the morbidity that remains despite the effectiveness of HAART. Studies have demonstrated that when Th17 reconstitution has been observed, a reduction in microbial translocation and systemic immune activation markers is also observed [11,6567], and that preservation of intestinal Th17 cells in SIV-infected rhesus macaques is associated with reduced microbial translocation and systemic immune activation [68]. Consistent with the hypothesis that HIV alters Th17 function and generation, we have shown that Th17 cells lose the ability to express IL-17 following infection with HIV. Both in vitro and in vivo HIV infection significantly inhibit the ability of Th17 cells to respond to IL-23, a key cytokine implicated in the generation of Th17 cells and the maintenance of their function. Importantly, this lack of responsiveness to IL-23 does not appear to be reversed by HAART. This irreversible defect in IL-23 signaling in Th17 cells may be a driving factor for microbial translocation and persistent immune activation, and thus should be a focus for therapeutic targets to reduce immune activation-associated morbidity in HIV infection.

Acknowledgments

This work was supported by the Canadian Institutes of Health Research (CIHR) (JBA and AK, HOP-98830; operating grant). The funding agency played no role in study design, collection or analysis of data, writing of the manuscript, or decision to submit for publication.

References

  1. 1. Dandekar S, George MD, Bäumler AJ. Th17 cells, HIV and the gut mucosal barrier. Curr Opin HIV AIDS. 2010 Mar;5(2):173–8. pmid:20543596
  2. 2. Conti HR, Shen F, Nayyar N, Stocum E, Sun JN, Lindemann MJ, et al. Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med. 2009 Feb 16;206(2):299–311. pmid:19204111
  3. 3. Hernández-Santos N, Huppler AR, Peterson AC, Khader SA, McKenna KC, Gaffen SL. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol. 2013 Sep;6(5):900–10. pmid:23250275
  4. 4. Cao AT, Yao S, Gong B, Elson CO, Cong Y. Th17 cells upregulate polymeric Ig receptor and intestinal IgA and contribute to intestinal homeostasis. J Immunol Baltim Md 1950. 2012 Nov 1;189(9):4666–73.
  5. 5. Duan M-C, Huang Y, Zhong X-N, Tang H-J. Th17 cell enhances CD8 T-cell cytotoxicity via IL-21 production in emphysema mice. Mediators Inflamm. 2012;2012:898053. pmid:23319833
  6. 6. Hirota K, Turner J-E, Villa M, Duarte JH, Demengeot J, Steinmetz OM, et al. Plasticity of Th17 cells in Peyer’s patches is responsible for the induction of T cell-dependent IgA responses. Nat Immunol. 2013 Apr;14(4):372–9. pmid:23475182
  7. 7. Jaffar Z, Ferrini ME, Herritt LA, Roberts K. Cutting edge: lung mucosal Th17-mediated responses induce polymeric Ig receptor expression by the airway epithelium and elevate secretory IgA levels. J Immunol Baltim Md 1950. 2009 Apr 15;182(8):4507–11.
  8. 8. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004 Oct;21(4):467–76. pmid:15485625
  9. 9. Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, Asher TE. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood [Internet]. 2008;112. Available from: http://dx.doi.org/10.1182/blood-2008-05-159301
  10. 10. Xu H, Wang X, Liu DX, Moroney-Rasmussen T, Lackner AA, Veazey RS. IL-17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected macaques. Mucosal Immunol. 2012 Nov;5(6):658–69. pmid:22669579
  11. 11. Chege D, Sheth PM, Kain T, Kim CJ, Kovacs C, Loutfy M. Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS [Internet]. 2011;25. Available from: http://dx.doi.org/10.1097/QAD.0b013e328344cefb
  12. 12. Marchetti G, Bellistrì GM, Borghi E, Tincati C, Ferramosca S, La Francesca M, et al. Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS Lond Engl. 2008 Oct 1;22(15):2035–8.
  13. 13. El Hed A, Khaitan A, Kozhaya L, Manel N, Daskalakis D, Borkowsky W, et al. Susceptibility of human Th17 cells to human immunodeficiency virus and their perturbation during infection. J Infect Dis. 2010 Mar 15;201(6):843–54. pmid:20144043
  14. 14. Singh A, Vajpayee M, Ali SA, Mojumdar K, Chauhan NK, Singh R. HIV-1 diseases progression associated with loss of Th17 cells in subtype “C” infection. Cytokine. 2012 Oct;60(1):55–63. pmid:22840497
  15. 15. Alvarez Y, Tuen M, Shen G, Nawaz F, Arthos J, Wolff MJ. Preferential HIV Infection of CCR6+ Th17 Cells Is Associated with Higher Levels of Virus Receptor Expression and Lack of CCR5 Ligands. J Virol [Internet]. 2013;87. Available from: http://dx.doi.org/10.1128/JVI.01838-13
  16. 16. Cecchinato V, Trindade CJ, Laurence A, Heraud JM, Brenchley JM, Ferrari MG, et al. Altered balance between Th17 and Th1 cells at mucosal sites predicts AIDS progression in simian immunodeficiency virus-infected macaques. Mucosal Immunol. 2008 Jul;1(4):279–88. pmid:19079189
  17. 17. Klatt NR, Estes JD, Sun X, Ortiz AM, Barber JS, Harris LD. Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection. Mucosal Immunol [Internet]. 2012;5. Available from: http://dx.doi.org/10.1038/mi.2012.38
  18. 18. Raffatellu M, Santos RL, Verhoeven DE, George MD, Wilson RP, Winter SE, et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat Med. 2008 Apr;14(4):421–8. pmid:18376406
  19. 19. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, Hogan C, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004 Sep 20;200(6):761–70. pmid:15365095
  20. 20. He Y, Li J, Zheng Y, Luo Y, Zhou H, Yao Y, et al. A randomized case-control study of dynamic changes in peripheral blood Th17/Treg cell balance and interleukin-17 levels in highly active antiretroviral-treated HIV type 1/AIDS patients. AIDS Res Hum Retroviruses. 2012 Apr;28(4):339–45. pmid:21767239
  21. 21. Ndhlovu LC, Chapman JM, Jha AR, Snyder-Cappione JE, Pagán M, Leal FE, et al. Suppression of HIV-1 plasma viral load below detection preserves IL-17 producing T cells in HIV-1 infection. AIDS Lond Engl. 2008 May 11;22(8):990–2.
  22. 22. McGeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, Blumenschein WM, et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol. 2009 Mar;10(3):314–24. pmid:19182808
  23. 23. Chen Z, Tato CM, Muul L, Laurence A, O’Shea JJ. Distinct regulation of interleukin-17 in human T helper lymphocytes. Arthritis Rheum. 2007 Sep;56(9):2936–46. pmid:17763419
  24. 24. Akimzhanov AM, Yang XO, Dong C. Chromatin remodeling of interleukin-17 (IL-17)-IL-17F cytokine gene locus during inflammatory helper T cell differentiation. J Biol Chem. 2007 Mar 2;282(9):5969–72. pmid:17218320
  25. 25. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005 Jan 17;201(2):233–40. pmid:15657292
  26. 26. Monteleone I, Pallone F, Monteleone G. Interleukin-23 and Th17 cells in the control of gut inflammation. Mediators Inflamm. 2009;2009:297645. pmid:19503799
  27. 27. Morrison PJ, Ballantyne SJ, Kullberg MC. Interleukin-23 and T helper 17-type responses in intestinal inflammation: from cytokines to T-cell plasticity. Immunology. 2011 Aug;133(4):397–408. pmid:21631495
  28. 28. Ness-Schwickerath KJ, Morita CT. Regulation and function of IL-17A- and IL-22-producing γδ T cells. Cell Mol Life Sci CMLS. 2011 Jul;68(14):2371–90. pmid:21573786
  29. 29. Stritesky GL, Yeh N, Kaplan MH. IL-23 promotes maintenance but not commitment to the Th17 lineage. J Immunol Baltim Md 1950. 2008 Nov 1;181(9):5948–55.
  30. 30. Parham C, Chirica M, Timans J, Vaisberg E, Travis M, Cheung J, et al. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 and a novel cytokine receptor subunit, IL-23R. J Immunol Baltim Md 1950. 2002 Jun 1;168(11):5699–708.
  31. 31. Chen Z, Laurence A, Kanno Y, Pacher-Zavisin M, Zhu BM, Tato C. Selective regulatory function of Socs3 in the formation of IL-17-secreting T cells. Proc Natl Acad Sci U A [Internet]. 2006;103. Available from: http://dx.doi.org/10.1073/pnas.0600666103
  32. 32. Caruso R, Fina D, Paoluzi OA, Del Vecchio Blanco G, Stolfi C, Rizzo A, et al. IL-23-mediated regulation of IL-17 production in Helicobacter pylori-infected gastric mucosa. Eur J Immunol. 2008 Feb;38(2):470–8. pmid:18200634
  33. 33. de Beaucoudrey L, Puel A, Filipe-Santos O, Cobat A, Ghandil P, Chrabieh M, et al. Mutations in STAT3 and IL12RB1 impair the development of human IL-17-producing T cells. J Exp Med. 2008 Jul 7;205(7):1543–50. pmid:18591412
  34. 34. Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007 Jun;8(6):639–46. pmid:17486092
  35. 35. Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol [Internet]. 2007;8. Available from: http://dx.doi.org/10.1038/ni1488
  36. 36. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity. 2008 Jan;28(1):29–39. pmid:18164222
  37. 37. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 2007 Mar 30;282(13):9358–63. pmid:17277312
  38. 38. Che Mat NF, Zhang X, Guzzo C, Gee K. Interleukin-23-induced interleukin-23 receptor subunit expression is mediated by the Janus kinase/signal transducer and activation of transcription pathway in human CD4 T cells. J Interferon Cytokine Res Off J Int Soc Interferon Cytokine Res. 2011 Apr;31(4):363–71.
  39. 39. Basu R, Hatton RD, Weaver CT. The Th17 family: flexibility follows function. Immunol Rev. 2013 Mar;252(1):89–103. pmid:23405897
  40. 40. Peck A, Mellins ED. Plasticity of T-cell phenotype and function: the T helper type 17 example. Immunology. 2010 Feb;129(2):147–53. pmid:19922424
  41. 41. Lee YK, Mukasa R, Hatton RD, Weaver CT. Developmental plasticity of Th17 and Treg cells. Curr Opin Immunol. 2009 Jun;21(3):274–80. pmid:19524429
  42. 42. Kim CJ, McKinnon LR, Kovacs C, Kandel G, Huibner S, Chege D, et al. Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation. J Immunol Baltim Md 1950. 2013 Sep 1;191(5):2164–73.
  43. 43. Lurati A, Laria A, Gatti A, Brando B, Scarpellini M. Different T cells’ distribution and activation degree of Th17 CD4+ cells in peripheral blood in patients with osteoarthritis, rheumatoid arthritis, and healthy donors: preliminary results of the MAGENTA CLICAO study. Open Access Rheumatol Res Rev. 2015;7:63–8.
  44. 44. Morita R, Schmitt N, Bentebibel S-E, Ranganathan R, Bourdery L, Zurawski G, et al. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011 Jan 28;34(1):108–21. pmid:21215658
  45. 45. Crome SQ, Clive B, Wang AY, Kang CY, Chow V, Yu J, et al. Inflammatory effects of ex vivo human Th17 cells are suppressed by regulatory T cells. J Immunol Baltim Md 1950. 2010 Sep 15;185(6):3199–208.
  46. 46. Goodman WA, Young AB, McCormick TS, Cooper KD, Levine AD. Stat3 phosphorylation mediates resistance of primary human T cells to regulatory T cell suppression. J Immunol Baltim Md 1950. 2011 Mar 15;186(6):3336–45.
  47. 47. Diveu C, McGeachy MJ, Boniface K, Stumhofer JS, Sathe M, Joyce-Shaikh B, et al. IL-27 blocks RORc expression to inhibit lineage commitment of Th17 cells. J Immunol Baltim Md 1950. 2009 May 1;182(9):5748–56.
  48. 48. Honda M, Yamamoto S, Cheng M, Yasukawa K, Suzuki H, Saito T, et al. Human soluble IL-6 receptor: its detection and enhanced release by HIV infection. J Immunol Baltim Md 1950. 1992 Apr 1;148(7):2175–80.
  49. 49. Miller RC, Schlaepfer E, Baenziger S, Crameri R, Zeller S, Byland R, et al. HIV interferes with SOCS-1 and -3 expression levels driving immune activation. Eur J Immunol. 2011 Apr;41(4):1058–69. pmid:21337543
  50. 50. Popik W, Hesselgesser JE, Pitha PM. Binding of human immunodeficiency virus type 1 to CD4 and CXCR4 receptors differentially regulates expression of inflammatory genes and activates the MEK/ERK signaling pathway. J Virol. 1998 Aug;72(8):6406–13. pmid:9658081
  51. 51. Rozmyslowicz T, Murphy SL, Conover DO, Gaulton GN. HIV-1 infection inhibits cytokine production in human thymic macrophages. Exp Hematol. 2010 Dec;38(12):1157–66. pmid:20817073
  52. 52. Haller C, Rauch S, Fackler OT. HIV-1 Nef employs two distinct mechanisms to modulate Lck subcellular localization and TCR induced actin remodeling. PloS One. 2007 Nov 21;2(11):e1212. pmid:18030346
  53. 53. Neri F, Giolo G, Potestà M, Petrini S, Doria M. The HIV-1 Nef protein has a dual role in T cell receptor signaling in infected CD4+ T lymphocytes. Virology. 2011 Feb 20;410(2):316–26. pmid:21176845
  54. 54. Rudolph JM, Eickel N, Haller C, Schindler M, Fackler OT. Inhibition of T-cell receptor-induced actin remodeling and relocalization of Lck are evolutionarily conserved activities of lentiviral Nef proteins. J Virol. 2009 Nov;83(22):11528–39. pmid:19726522
  55. 55. Sohn SJ, Barrett K, Van Abbema A, Chang C, Kohli PB, Kanda H, et al. A restricted role for TYK2 catalytic activity in human cytokine responses revealed by novel TYK2-selective inhibitors. J Immunol Baltim Md 1950. 2013 Sep 1;191(5):2205–16.
  56. 56. Bixler SL, Sandler NG, Douek DC, Mattapallil JJ. Suppressed Th17 levels correlate with elevated PIAS3, SHP2, and SOCS3 expression in CD4 T cells during acute simian immunodeficiency virus infection. J Virol. 2013 Jun;87(12):7093–101. pmid:23596301
  57. 57. Cheng SM, Li JCB, Lin SS, Lee DCW, Liu L, Chen Z, et al. HIV-1 transactivator protein induction of suppressor of cytokine signaling-2 contributes to dysregulation of IFN{gamma} signaling. Blood. 2009 May 21;113(21):5192–201. pmid:19279332
  58. 58. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006 Sep 22;126(6):1121–33. pmid:16990136
  59. 59. Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol [Internet]. 2008;9. Available from: http://dx.doi.org/10.1038/ni.1610
  60. 60. Zhou L, Lopes JE, Chong MMW, Ivanov II, Min R, Victora GD, et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008 May 8;453(7192):236–40. pmid:18368049
  61. 61. Du J, Huang C, Zhou B, Ziegler SF. Isoform-specific inhibition of ROR alpha-mediated transcriptional activation by human FOXP3. J Immunol Baltim Md 1950. 2008 Apr 1;180(7):4785–92.
  62. 62. Le Saout C, Hasley RB, Imamichi H, Tcheung L, Hu Z, Luckey MA, et al. Chronic exposure to type-I IFN under lymphopenic conditions alters CD4 T cell homeostasis. PLoS Pathog. 2014 Mar;10(3):e1003976. pmid:24603698
  63. 63. Federico M, Percario Z, Olivetta E, Fiorucci G, Muratori C, Micheli A, et al. HIV-1 Nef activates STAT1 in human monocytes/macrophages through the release of soluble factors. Blood. 2001 Nov 1;98(9):2752–61. pmid:11675348
  64. 64. Chaudhuri A, Yang B, Gendelman HE, Persidsky Y, Kanmogne GD. STAT1 signaling modulates HIV-1-induced inflammatory responses and leukocyte transmigration across the blood-brain barrier. Blood. 2008 Feb 15;111(4):2062–72. pmid:18003888
  65. 65. Gordon SN, Cervasi B, Odorizzi P, Silverman R, Aberra F, Ginsberg G, et al. Disruption of intestinal CD4+ T cell homeostasis is a key marker of systemic CD4+ T cell activation in HIV-infected individuals. J Immunol Baltim Md 1950. 2010 Nov 1;185(9):5169–79.
  66. 66. Macal M, Sankaran S, Chun T-W, Reay E, Flamm J, Prindiville TJ, et al. Effective CD4+ T-cell restoration in gut-associated lymphoid tissue of HIV-infected patients is associated with enhanced Th17 cells and polyfunctional HIV-specific T-cell responses. Mucosal Immunol. 2008 Nov;1(6):475–88. pmid:19079215
  67. 67. Nakayama K, Nakamura H, Koga M, Koibuchi T, Fujii T, Miura T, et al. Imbalanced production of cytokines by T cells associates with the activation/exhaustion status of memory T cells in chronic HIV type 1 infection. AIDS Res Hum Retroviruses. 2012 Jul;28(7):702–14. pmid:21902582
  68. 68. Pallikkuth S, Micci L, Ende ZS, Iriele RI, Cervasi B, Lawson B, et al. Maintenance of intestinal Th17 cells and reduced microbial translocation in SIV-infected rhesus macaques treated with interleukin (IL)-21. PLoS Pathog. 2013;9(7):e1003471. pmid:23853592