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

HIV replication is associated to inflammasomes activation, IL-1β, IL-18 and caspase-1 expression in GALT and peripheral blood

  • Manuel Gerónimo Feria,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia

  • Natalia Andrea Taborda,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia, Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia

  • Juan C. Hernandez,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Supervision, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia, Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia

  • Maria Teresa Rugeles

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    maria.rugeles@udea.edu.co

    Affiliation Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia

Abstract

Background

Human immunodeficiency virus (HIV) promotes an inflammatory process, leading to the progressive loss of the functional capacity of the immune system. The HIV infection induces alterations in several tissues, but mainly in the gut-associated lymphoid tissue (GALT). However, the degree of GALT deterioration varies among infected individuals. In fact, it has been shown that HIV-controllers, who spontaneously control viral replication, exhibit a lower inflammatory response, and a relative normal frequency and function of most of the immune cells. Inflammasomes are molecular complexes involved in the inflammatory response, being NLRP1, NLRP3, NLRC4, AIM2 and Pyrin inflammasomes, the best characterized so far. These complexes regulate the maturation of cytokines of the IL-1 family, including IL-1β and IL-18. These cytokines have been associated with immune activation and expansion of HIV target cells, promoting viral replication. Interesting, some reports indicate that HIV induces the activation of the NLRP3 inflammasome, but the role of this, and other inflammasomes during HIV infection, especially in GALT, remains unclear.

Objective

To compare the relative expression of inflammasome components and the proinflammatory response related to their activity, between HIV-progressors and HIV-controllers.

Methods

GALT biopsies and peripheral blood mononuclear cells (PBMCs) from 15 HIV-controllers and 15 HIV-progressors were obtained. The relative expression of the following inflammasome components were evaluated by RT-PCR: NLRP3, NLRC4, NLRP1, AIM2, ASC, Caspase-1, IL-1β and IL-18. In addition, plasma concentration of IL-18 was evaluated as an indicator of baseline proinflammatory status. Finally, in supernatants of PBMCs in vitro stimulated with inflammasome agonists, the concentrations of IL-1β and IL-18 were quantified by ELISA.

Results

HIV-progressors exhibited higher expression of IL-1β, IL-18 and caspase-1 genes in GALT and PBMCs compared with HIV-controllers. In addition, HIV-progressors had also increased expression of ASC in PBMCs. When plasma levels were evaluated, IL-18 was increased in HIV-progressors. Interesting, these patients also showed an increased production of IL-1β in supernatants of PBMCs stimulated in vitro with the agonists of AIM2, NLRP1 and NLRC4 inflammasomes. Finally, the expression of caspase-1, NLRP1, IL-1β and IL-18 in GALT or peripheral blood was correlated with CD4+ T-cell count and viral load.

Conclusion

Our results suggest that during HIV-infection, the required signals to induce the expression of different components of the inflammasomes are produced, both in GALT and in periphery. The activation of these molecular complexes could increase the number of target cells, favoring HIV replication and cell death, promoting the disease progression.

Introduction

The pathogenesis of HIV involves a complex interaction between several viral and host factors. So far, it is recognized that the gut-associated lymphoid tissue (GALT) is the main affected organ, as a consequence of the massive elimination of CD4+ T-cells, in particular Th17 cells that play an essential role in the mucosa homeostasis [1, 2]. The elimination of these and other cells such as enterocytes [35], induces structural damage in GALT, allowing microbial translocation from the intestinal lumen to systemic circulation, promoting excessive immune activation [6]. This state, currently recognize as the main immunopathogenic mechanism during HIV infection, is established during the acute phase and remains throughout the course of the infection, leading to cell exhaustion and activation-induced apoptosis [7]. Myeloid cells, including monocytes/macrophages and dendritic cells, are also affected by immune activation, as there is an increased level of microbial ligands recognized by Toll (TLR) and NOD (NLR) receptors inducing activation of NF-κB and the gene expression of proinflammatory cytokines such as IL-1β and IL-18 [8, 9]. In addition, endosomal TLR7 and TLR8 could detect HIV, increasing the inflammatory response [10, 11]. IL-1β and IL-18 are involved in the differentiation of naïve CD4+ T cells to Th1 and Th17 profiles [1214], increasing even more the immune activation, and promoting viral replication since these cells are viral targets; in fact, these cytokines are increased in plasma of infected patients [15, 16], suggesting their involvement in HIV pathogenesis [10, 11].

Several molecules participate during the inflammatory process, including the large multimolecular complexes, so-called inflammasomes. These molecules activate caspase-1 and cytokines of the IL-1 family, including IL-1β and IL-18 [17]. Although several inflammasomes have been described, the best characterized are the nucleotide-binding domain leucine-rich repeat-containing (NLR) family (NLRP1, NLRP3, and NLRC4); the protein absent in melanoma 2 (AIM2), belonging to the PYHIN protein family; and the recently described Pyrin [18].

The excessive response of inflammasomes has been associated with some chronic inflammatory diseases such as type 2 diabetes, asthma, atherosclerosis, kidney disease and autoimmune disorders [19, 20]. In the context of HIV, some studies have found that HIV induces NLRP3 inflammasome activation as an alternative mechanism to maintain the inflammatory process. In fact, it has been previously published that HIV is able to induce the primary signal for NLRP3 inflammasome activation in macrophages [15]. In addition, other investigations have reported that dendritic cells exhibit high mRNA expression of NLRP3, IL-1β and caspase-1 in response to in vitro HIV infection, suggesting that this inflammasome might be involved in the pathogenesis of this infection [21, 22]. However, the role of these molecular complexes, especially in GALT, is unclear. Therefore, in a cohort of HIV-progressors and HIV-controllers, we compared the relative expression of inflammasome components and the proinflammatory response, related to their activity. The results were correlated with viral load and CD4+ T-cell count in both, peripheral blood and GALT, in order to determine the potential association between inflammasome activation and AIDS progression.

Materials and methods

Study population

Fifteen HIV-controllers, based on previously defined criteria [23, 24] and 15 HIV-progressors with viral loads between 10,000 and 100,000 copies/mL and CD4+ T-cell count >350 cells/μL, were included. Peripheral blood samples from all donors and GALT samples from 11 HIV-controllers and 13 HIV-progressors were obtained. All the enrolled individuals were antiretroviral therapy naïve and recruited from health insurance programs in Medellin, Colombia. The individuals enrolled signed a written informed consents prepared according to the colombian legislation, resolution 008430/1993 and approved by Ethical committee of the University of Antioquia (certificate 14-08-567) files.

Viral load in plasma and CD4+ T-cell count

Plasma viral load was determined using the commercial assay RT-PCR Ampliprep-Cobas Amplicor (Roche, Indianapolis, IN; detection limit of 20 copies/mL), following the manufacturer’s protocol. The frequency of peripheral blood and GALT CD4+ T-cells was determined by flow cytometry. Briefly, peripheral blood was incubated with specific monoclonal antibodies at room temperature in the dark. The erythrocytes were lysed and the cells were washed twice with PBS and fixed with 2% paraformaldehyde. For GALT, cells were first incubated with IgG-blocking antibodies 20 μg/mL (eBioscience, San Diego, USA); then, cells were washed twice with PBS and incubated with specific monoclonal antibodies at room temperature in the dark. In this study, the following fluorescence labeled monoclonal antibodies were obtained from eBioscience (San Diego, USA): CD3 (Anti-CD3-FITC; clone UCHT1), CD4 (Anti-CD4-APC; Clone: RPA-T4) and CD8 -PE, clone: RPA-T8). The lymphocyte region was selected by "Size (SSC) vs. Forward (FSC) light scatter" parameters. CD8+ and CD4+ T-cells were selected from the CD3+ gate. The acquisition was performed on the FACS CANTO-II (BD) cytometer, using the software BD FACSDiva version 6.1.2.

Isolation of mononuclear cells from GALT biopsies

Rectosigmoidoscopy and biopsies were performed as previously reported [25], using a flexible sigmoidoscope with single-use biopsy forceps FB-24K-1 (Olympus America Corp, Melville, NY, USA); from each subject, tissue samples were obtained from the rectum at 10 cm from anal verge. Four fragments were digested with collagenase type II from Clostridium histolyticum (Sigma; 0.5 mg/mL) diluted in RPMI 1640% and 7.5% fetal bovine serum (FBS) plus 100 U/mL penicillin and 100 mg/mL streptomycin (Gibco-BRL, Grand Island, NY) for 30 minutes at 37°C with shaking. After collagenase digestion, biopsy fragments were further disrupted by repeated passage through a syringe with a 16-gauge blunt-end needle (Stem Cell Technologies, Vancouver, BC, Canada). Rectal cells (RCs) were isolated from the fragments by passage through a 70 mM nylon strainer (Falcon, Lincoln Park, NJ). RCs were washed with PBS (Sigma-Aldrich, San Luis, MO) to remove excess of collagenase. Subjects with nodular lymphoid hyperplasia, ulcers, diverticulitis, adenoma and other benign or malignant growths were excluded from the study.

In vitro activation of inflammasomes

Peripheral blood mononuclear cells (PBMCs) were isolated using Ficoll-hypaque gradient. Then, 1x105 PBMCs were primed for 2 hours with 50 pg/mL ultrapure lipopolysaccharide (LPS) from Escherichia coli. The second activation signal was induced with specific inflammasome agonists, including ATP (2mM) for NLRP3; flagellin of Salmonella typhimurium (500 ng/mL) for NLRC4; poly(dA;dT) (50 μg/mL) for AIM2; and Muramyl dipeptide (MDP, 0.1 μg/mL) for NLRP1. After four hours of incubation, or 2 hours for ATP-treated PBMCs, the supernatants were harvested. All the agonists used were from Invivogen (California, USA). To minimize the effects of variability among individuals, the results were normalized according to the primed control cells (cells treated only with 50 pg/mL LPS), and expressed as fold increases.

ELISA

The IL-1β production was quantified following the recommendations of the manufacturer of the OptEIA ™ Set commercial kit (BD Biosciences, San Diego, USA); for IL-18, the Human IL-18 Matched Antibody Pairs BMS267/2MST kit (eBioscience, Vienna, Austria) was used. IL-1β and IL-18 detection was performed in plasma of HIV infected individuals, and in supernatants of agonists-stimulated PBMCs.

Gene expression of inflammasome components

To determine the transcriptional expression of genes associated with the inflammasomes (NLRP3, NLRP1, NLRC4, NLRP6, AIM2, ASC, and caspase-1) and their products (IL-1β and IL-18), total RNA extraction from PBMCs and GALT biopsies was performed using the RNeasy Mini Kit (QIAGEN, Inc., Valencia, CA, USA), following the manufacturer's instructions. RNA was treated with DNAse (DNase I, RNase-free, Qiagen, Hilden, Germany) and the cDNA was synthesized using the RevertAid H Minus First Strand cDNA Synthesis Kit (Thermo scientific, Waltham, Massachusetts, USA).

Gene expression was quantified by real-time PCR, using Maxima SYBR Green/ROX qPCR Master Mix (Thermo scientific, Waltham, Massachusetts, USA), specific oligonucleotides (S1 Table) and 2 μL cDNA of each sample in a final volume of 20 μL. A melting curve was included to confirm the specificity of the PCR product. All real-time PCR amplifications were performed using the CFX96 real-time system and data analysis using the software CFX Manager Version 1.5.534.0511 (Bio-Rad,Hercules, CA). Relative expression was calculated using ubiquitin to normalize the RNA content and the ΔCt method (1,8 (ct(target gene)-ct(ubiquitin gene)) was applied. The same amount de cDNA was used in all cases [26].

Statistical analysis

To compare data from HIV-controllers vs. HIV-progressors, Mann–Whitney U–2-tailed test was used. A p value <0.05 was considered statistically significant. Correlation analyses were based on Spearman correlation coefficient calculations. The statistical tests were performed using the GraphPad Software version 7.02.

Results

Prolonged control of viral infection in HIV-controllers

Demographic characteristics of HIV-infected donors are shown in Table 1. No significant differences were observed in the nadir CD4+ T cells between HIV- controllers and HIV-progressors. However, HIV-controllers exhibited lower viral loads and a higher CD4+ T cell count, compared with HIV-progressors. In addition, the mean time since HIV diagnosis was 62 months in HIV-controllers, evidencing their stable ability for exerting viral control; in fact, some of these individuals, could be classified as long-term non progressors.

thumbnail
Table 1. Demographic characteristics of HIV-infected donors.

https://doi.org/10.1371/journal.pone.0192845.t001

Higher expression of IL-1β, IL-18 and caspase-1 genes in GALT from HIV-progressors

Considering that GALT is the main affected tissue during HIV infection, the expression of the inflammasome components in this organ was evaluated. Compared to HIV-controllers, HIV-progressors had a higher expression of IL-1β (Fig 1A), IL-18 (Fig 1B) and caspase-1 (Fig 1C). In contrast, we did not observe any differences in the expression of other inflammasome components, including, ASC, NLRP3, NLRC4, NLRP1, and AIM2 (Fig 1D–1H), between groups.

thumbnail
Fig 1. Relative expression of the inflammasomes components in GALT.

The RNA levels of IL-1β (A), IL-18 (B), caspase- 1 (C), ASC (D), NLRP3 (E), NLRC4 (F), NLRP1 (G), and AIM2 (H) were quantified in GALT from HIV-1-controllers and HIV-1-progressors, by qPCR. The ubiquitin gene was used as constitutive gene to normalize the RNA content. Relative expression is referred to ubiquitin expression. The results are presented as medians and interquartile ranges. Statistical comparison was performed using a Mann-Whitney U test with a 95% confidence level. Significant differences are indicated at the top of the figure (* p <0.05).

https://doi.org/10.1371/journal.pone.0192845.g001

Increased expression of IL-1β, IL-18, caspase-1 and ASC genes in PBMCs from HIV-progressors

The relative expression of IL-1β (Fig 2A), IL-18 (Fig 2B), caspase-1 (Fig 2C) and ASC (Fig 2D) genes was higher in PBMCs from HIV-progressors than HIV-controllers. No significant differences were observed in other inflammasome components, including NLRP3, NLRC4, NLRP1, AIM2 (Fig 2E–2H).

thumbnail
Fig 2. Relative expression of the inflammasomes components in PBMCs.

The RNA levels of IL-1β (A), IL-18 (B), caspase- 1 (C), ASC (D), NLRP3 (E), NLRC4 (F), NLRP1 (G), and AIM2 (H) were quantified in PBMCs from HIV-1-controllers and HIV-1-progressors, by qPCR. The ubiquitin gene was used as constitutive gene to normalize the RNA content. Relative expression is referred to ubiquitin expression. The results are presented as medians and interquartile ranges. Statistical comparison was performed using a Mann-Whitney U test with a 95% confidence level. Significant differences are indicated at the top of the figure (* p <0.05) (**p<0.01) (***p<0,001).

https://doi.org/10.1371/journal.pone.0192845.g002

Higher levels of IL-18 in plasma from HIV-progressors

In order to determine the basal inflammatory status associated with inflammasomes activity, IL-18 was quantified in plasma from HIV-infected donors. HIV-progressors have a higher IL-18 concentration compared to HIV-controllers (131.6 pg/mL vs. 4.8 pg/mL, Fig 3).

thumbnail
Fig 3. Plasma levels of IL-18 in HIV-1-infected donors.

IL-18 was quantified in plasma samples by ELISA. The results are presented as median and interquartile ranges. Statistical comparison between groups was performed using a Mann-Whitney U test with a confidence level of 95%. Significant differences are indicated at the top of the figure (** p <0.01).

https://doi.org/10.1371/journal.pone.0192845.g003

HIV-progressors exhibit increased production of IL-1β through AIM2, NLRP1 and NLRC4 inflammasomes, in an in vitro model

To determine the specific inflammasomes associated to HIV progression, through the IL-1β and IL-18 production, an in vitro assay was performed. A higher production of IL-1β in PBMCs from HIV-progressors compared to HIV-controllers, was observed when the cells were treated with agonist of NLRP1 (Fig 4B), NLRC4 (Fig 4C) and AIM2 (Fig 4D) inflammasomes. In the case of the NLRP3 inflammasome (Fig 4A), there were no significant differences in IL-1β release between both groups of patients.

thumbnail
Fig 4. Activation of inflammasomes in PBMCs from HIV-1-infected donors.

LPS-primed PBMCs were treated with inflammasomes agonists (ATP for NLRP3 (A), MDP for NLRP1 (B), Flagellin for NLRC4 (C) and poly(dA:dT) for AIM2 (D), and the IL-1β release was quantified by ELISA in the supernatants. Fold increase is referred to primed cells (cells treated only with 50 pg/mL LPS). The results are presented as median and interquartile ranges. Statistical comparison between groups was performed using a Mann-Whitney U test with a confidence level of 95%. Significant differences are indicated at the top of the figure (*p <0.05).

https://doi.org/10.1371/journal.pone.0192845.g004

The expression of caspase-1, NLRP1, IL-1β and IL-18 in GALT or peripheral blood correlates with CD4+ T cell count and viral load

Initially, we compared the frequency of CD4+ T cells in GALT and peripheral blood between the HIV-infected donors. As expected, HIV-controllers exhibit higher frequency of CD4+ T-cells in GALT (Fig 5A) and peripheral blood (Fig 5B). Then, in order to determine if the altered expression of inflammasome components and their products were involved in HIV progression, a correlation analysis was performed. In GALT, the relative expression of NLRP1 (Fig 5C) was negatively correlated with CD4+ T-cell count. Likewise, caspase-1 was negatively correlated with CD4+ T-cell count (Fig 5D) and positively correlated with viral load (Fig 5E).

thumbnail
Fig 5. Correlation between the expressions of the inflammasome components in GALT with the CD4+ T-cell count and viral load.

Higher frequency of CD4+-T cells in GALT (A) and peripheral blood (B) in HIV-controllers compared to HIV-progressors. The relative expression of NLRP1 in GALT was negatively correlated with the CD4+ T-cell in peripheral blood (C). Equally, the relative expression of caspase-1 was negatively correlated with the CD4+ T cells in peripheral blood (D) and positively with the viral load (E). Statistical comparison between groups was performed using a Mann-Whitney U test with a confidence level of 95%. Significant differences are indicated at the top of the figure (**p <0.01) (***p<0,001). The correlations were performed with a Spearman test. The r value and the p-value of the correlations are indicated at the top of each figure; a p-value less than 0.05 was considered as a significant correlation.

https://doi.org/10.1371/journal.pone.0192845.g005

Finally, in peripheral blood, viral load was positively correlated with the relative expression of IL-1β and IL-18 (Fig 6A and 6B). Interestingly, the protein level of IL-18 was positively correlated with viral load and negatively correlated with the frequency of CD4+ T cells in GALT (Fig 6C and 6D). Finally, the relative expression of caspase-1 and the protein level of IL-18 were positively correlated (Fig 6E).

thumbnail
Fig 6. Correlation between the expression of the inflammasome components and products in peripheral with the CD4+ T-cell count and the viral load.

The relative expression of IL-1β and IL-18 in PBMCs was positively correlated with viral load (A) (B). The plasma concentration of IL-18 was negatively correlated with the CD4+ T-cell frequency in GALT (C) and positively with the viral load (D). The expression of caspase-1 was positively correlated with plasma concentration of IL-18 (E). The correlations were performed with the Spearman test. The r value and the p-value of the correlations are indicated at the top of each figure; a p-value less than 0.05 was considered as a significant correlation.

https://doi.org/10.1371/journal.pone.0192845.g006

Discussion

HIV is characterized by an unregulated inflammatory response and the consequent immune exhaustion [27]. Inflammasomes could be one of the immune components involved in this process. They are important in the regulation of the caspase-1 activity to induce the proteolytic maturation of the IL-1β and IL-18. These proinflammatory cytokines are particularly increased in HIV infected individuals, suggesting their role in AIDS progression [15, 28]. In fact, it has been shown that HIV has the ability to activate the NLRP3 inflammasome [15]; however, the definite role of this, and other inflammasomes during the pathogenesis of HIV is still unclear. Here, we evaluated the role of different inflammasomes in the pathogenesis of this infection, by studying two different groups of infected patients: HIV-controllers and HIV-progressors.

We observed that HIV-progressors exhibit higher relative expression of IL-1β, IL-18 and caspase-1 in GALT, and in PBMCs, compared with HIV-controllers. In addition, HIV-progressors had also increased expression of ASC in PBMCs, which could reflect the fact that this protein is mainly expressed by monocytes and lymphocytes [29]. In relation to our findings, ex vivo studies have demonstrated that HIV induces the expression of caspase-1, NLRP3, IL-1β and IL-18 in cerebral white matter and microglial cells [30, 31], promoting an inflammatory process at the central nervous system, and HIV-associated neurocognitive disorders [32]. In addition, in vitro HIV-infected dendritic cells and monocytes-derived macrophages have shown increased expression of others inflammasome components such as NLRP3, ASC and AIM2 [21, 22, 33]. Similar results have been observed in primary cells or HIV infected cell lines [34, 35], indicating the ability of the virus for inducing the priming signal for the activation of the inflammasomes in different tissues, as one of the mechanisms responsible for the systemic hyperactivation observed in infected individuals, even in those under highly active antiretroviral therapy (HAART) [36].

It is clear now that immune activation is established early during the acute phase of the infection, leading to diverse alterations in GALT, favoring viral replication and eventually, immune exhaustion [37]. Transcriptome studies in simian immunodeficiency virus (SIV)-infected monkeys have shown that several proinflammatory genes are upregulated, including those coding for various inflammasome components such as NLRs, TLRs, and caspases [38]. Supporting this evidence, here we show that HIV-progressors had a higher relative expression of IL-1β, IL-18 and caspase-1, compared with HIV-controllers in GALT and PBMCs samples. These findings might suggest that the inflammasomes might indeed influence the clinical course of the HIV infection, by participating in the inflammatory process, recognized as one of the main pathogenic mechanisms associated to AIDS progression. In fact, the massive elimination, in GALT, of HIV non-permissive cells occur by pyroptosis, which is induced mainly by the activation of the caspase-1. Interestingly, a higher expression of caspase-1 in both, PBMCs and GALT samples was observed in HIV-progressors compared with HIV controllers. In addition, the relative expression of the caspase-1 in GALT had a negative and a positive correlation with CD4+ T-cell count and with load viral, respectively. These results support the potential role of the caspasa-1 in promoting immune deterioration during HIV infection and viral replication.

The lack of differences between both study groups in the relative expression of the PRRs, NLRP3, NLRC4, NLRP1 and AIM2 might indicate that these proteins do not influence HIV progression, although the impact of the low sample size cannot be ruled-out. Similar results were reported in studies comparing HIV infected patients with uninfected individuals [31], where a higher expression of IL-1β, IL-18, and caspase-1 were found in cerebral white matter from HIV- infected individuals.

Another characteristic of HIV-infected individuals is the expression of high levels of the proinflammatory cytokines IL-1β and IL-18 [15, 39]. In fact, we observed that HIV-progressors had higher levels of circulating IL-18 compared with HIV-controllers. During HIV infection, low plasma concentrations of the IL-18-binding protein (which modulate the proinflammatory activity of the IL-18) have been reported [40]; this finding might contribute to the exacerbated inflammatory process that characterizes HIV-progressors [25]. It is important to note that IL-18 is involved in the differentiation of naïve T cells to Th1 and Th17 cells [13, 14, 41, 42], which are highly susceptible to HIV infection; therefore, high levels of this cytokine could promote massive viral replication, immune activation and the consequent mucosal damage. Exploring the frequencies of different CD4+ T cell subpopulations in HIV patients, exhibiting different progression patterns, could support or ruled-out this hypothesis.

Unfortunately, we did not detect IL-1β, although it is also processed by the inflammasome. The presence of IL-1β inhibitors in the plasma samples is the most likely explanation for this result, since exogenous addition of this cytokine to the plasma did not result in the subsequent detection of this protein. In fact, higher levels of the IL-1RII, natural inhibitor of this cytokine, have been previously reported in HIV-infected [43]. Since previous reports have indeed indicated the presence of high levels of this cytokine in HIV infected individuals, further studies evaluating its correlation with progression are required.

Previously, in vitro studies demonstrated that HIV or its products (viral proteins Vpr and Tat or the non-integrated viral cDNA) activates NLRP3 and AIM2 inflammasomes [15, 30, 35, 44, 45]. However, the inflammasomes most likely responsible for the high production of IL-1β and IL-18 are still unclear. In this study, we observed that HIV-progressors produce increased levels of IL-1β through AIM2, NLRC4 and NLRP1 inflammasomes, in response to the specific agonists. In contrast to other studies, in cell cultures we did not detect IL-18 [46]. It seems that the level of production of this cytokine in cultures is very low, even below the detection limit of the commercial kit used; in fact, in previous studies, the protein had to be concentrated for the ELISA determination [47].

In relation with the higher production of IL-1β through the AIM2 inflammasome observed in HIV-progressors, it has been previously demonstrated that the proteins of the HIN200 family, AIM2 and IFI16 (interferon-γ inducible protein 16) interact with the HIV cDNA present in the cytoplasm of non-permissive cells. This interaction results in caspase-1 activation, which in turns induce pyroptosis, promoting the structural and functional deterioration of GALT [44, 45]

To our knowledge, this is the first report indicating a higher production of IL-1β through the NLRC4 and NLRP1 inflammasomes during HIV infection, mainly in HIV-progressors. However, it has been demonstrated that the activation of both inflammasomes can result from the interaction with the bacterial proteins flagellina and T3SS for the NLRC4 inflammasome [48], and MDP for the NLRP1 inflammasome [49]. Particularly, flagellina, T3SS and MDP are proteins found in the gut microbiote [50, 51] that could be translocated to systemic circulation during HIV infection because of the increased permeability of the GALT mucosa, previously reported in infected patients [52]. In the opposite side, in HIV-controllers, the low levels of IL-1β and IL-18 could be associated with the effectiveness of regulatory mechanisms, such as post-translation modifications of inflammasome proteins, including deubiquitination of NLRP3 or phosphorylation of ASC proteins [53, 54].

In relation with the NLRP3 inflammasome, no differences were observed between HIV groups. These results might correspond to the fact that the NRLP3 inflammasome has multiple agonists, such as ROS and ATP that could induce its activation through different intracellular pathways. In this sense, the physiopathology experienced by HIV-infected individuals, independent of their ability to control viral replication, induces the production of several DAMPs responsible for the activation of the NLRP3 inflammasome.

Finally, the results suggest that during HIV-infection the required signals to induce the relative expression of different components of the inflammasome are produced, both in GALT and in periphery. The activation of the molecular complexes increased the number of target cells, favoring HIV replication and cell death (by different mechanisms, including pyroptosis), promoting the development of disease.

Supporting information

S1 Table. Primers list.

Sequences used for mRNA amplification of inflammasome-related genes.

https://doi.org/10.1371/journal.pone.0192845.s001

(DOCX)

Acknowledgments

The authors thank the patients and volunteers who kindly participated in this study. The authors also acknowledge the health personnel of the Clínica Bolivariana, Medellin, who carried out the rectosigmoidoscopies. We thank Dr. Luis Alfonso Correa for his support in pathological evaluation and to health institutions who support patient recruitment.

References

  1. 1. Lee S, Tato C, Joyce-Shaikh B. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity. 2015;43(4):727–38. pmid:26431948
  2. 2. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, Collins M, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203(10):2271–9. pmid:16982811
  3. 3. Christensen-Quick A, Lafferty M, Sun L, Marchionni L, DeVico A, Garzino-Demo A. Human Th17 Cells Lack HIV-Inhibitory RNases and Are Highly Permissive to Productive HIV Infection. J Virol. 2016;90(17):7833–47. pmid:27334595
  4. 4. Alvarez Y, Tuen M, Shen G, Nawaz F, Arthos J, Wolff MJ, et al. Preferential HIV infection of CCR6+ Th17 cells is associated with higher levels of virus receptor expression and lack of CCR5 ligands. J Virol. 2013;87(19):10843–54. pmid:23903844
  5. 5. Maresca M, Mahfoud R, Garmy N, Kotler DP, Fantini J, Clayton F. The virotoxin model of HIV-1 enteropathy: involvement of GPR15/Bob and galactosylceramide in the cytopathic effects induced by HIV-1 gp120 in the HT-29-D4 intestinal cell line. J Biomed Sci. 2003;10(1):156–66. pmid:12566994
  6. 6. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71. pmid:17115046
  7. 7. Hazenberg MD, Otto SA, van Benthem BH, Roos MT, Coutinho RA, Lange JM, et al. Persistent immune activation in HIV-1 infection is associated with progression to AIDS. Aids. 2003;17(13):1881–8. pmid:12960820
  8. 8. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84. pmid:20404851
  9. 9. Kanneganti TD, Lamkanfi M, Nunez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007;27(4):549–59. pmid:17967410
  10. 10. Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303(5663):1526–9. pmid:14976262
  11. 11. Doyle T, Goujon C, Malim MH. HIV-1 and interferons: who's interfering with whom? Nat Rev Microbiol. 2015;13(7):403–13. pmid:25915633
  12. 12. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8(9):942–9. pmid:17676045
  13. 13. Tominaga K, Yoshimoto T, Torigoe K, Kurimoto M, Matsui K, Hada T, et al. IL-12 synergizes with IL-18 or IL-1beta for IFN-gamma production from human T cells. Int Immunol. 2000;12(2):151–60. pmid:10653850
  14. 14. Yoshimoto T, Takeda K, Tanaka T, Ohkusu K, Kashiwamura S, Okamura H, et al. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-gamma production. J Immunol. 1998;161(7):3400–7. pmid:9759857
  15. 15. Hernandez JC, Latz E, Urcuqui-Inchima S. HIV-1 induces the first signal to activate the NLRP3 inflammasome in monocyte-derived macrophages. Intervirology. 2014;57(1):36–42. pmid:24008203
  16. 16. Veenhuis RT, Astemborski J, Chattergoon MA, Greenwood P, Jarosinski M, Moore RD, et al. Systemic Elevation of Proinflammatory Interleukin-18 in Human Immunodeficiency Virus (HIV)/Hepatitis C Virus (HCV) Coinfection Versus HIV or HCV Monoinfection. Clin Infect Dis. 2017.
  17. 17. Hernandez J, Sirois C, Latz E. Activation and Regulation of the NLRP3 Inflammasome. The Inflammasomes: Springer Basel; 2011. p. 197–208.
  18. 18. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16(7):407–20. pmid:27291964
  19. 19. Masters SL, Latz E, O’Neill LAJ. The Inflammasome in Atherosclerosis and Type 2 Diabetes. Sci Transl Med. 2011;2(81):1–7.
  20. 20. Lee TH, Song HJ, Park CS. Role of inflammasome activation in development and exacerbation of asthma. Asia Pac Allergy. 2014;4(4):187–96. pmid:25379478
  21. 21. Pontillo A, Silva LT, Oshiro TM, Finazzo C, Crovella S, Duarte AJ. HIV-1 induces NALP3-inflammasome expression and interleukin-1beta secretion in dendritic cells from healthy individuals but not from HIV-positive patients. Aids. 2012;26(1):11–8. pmid:21971358
  22. 22. Pontillo A, Santillo BT, Duarte AJ, Oshiro TM. Differential inflammasome expression and IL-1β secretion in monocyte-derived dendritic cells differentiated with IL-4 or IFN-α. AIDS Res Ther. 2013;10(35):3–7.
  23. 23. Taborda NA, Rugeles MT, Montoya CJ. Spontaneous control of HIV replication, but not HAART-induced viral suppression, is associated with lower activation of immune cells. J Acquir Immune Defic Syndr. 2014;66(4):365–9. pmid:24732877
  24. 24. Taborda NA, Hernandez JC, Lajoie J, Juno JA, Kimani J, Rugeles MT, et al. Short Communication: Low Expression of Activation and Inhibitory Molecules on NK Cells and CD4(+) T Cells Is Associated with Viral Control. AIDS Res Hum Retroviruses. 2015;31(6):636–40. pmid:25738606
  25. 25. Gonzalez SM, Taborda NA, Correa LA, Castro GA, Hernandez JC, Montoya CJ, et al. Particular activation phenotype of T cells expressing HLA-DR but not CD38 in GALT from HIV-controllers is associated with immune regulation and delayed progression to AIDS. Immunol Res. 2016;64(3):765–74. pmid:26724942
  26. 26. Hernandez JC, St Laurent G, Urcuqui-Inchima S. HIV-1-exposed seronegative individuals show alteration in TLR expression and pro-inflammatory cytokine production ex vivo: An innate immune quiescence status? Immunol Res. 2016;64(1):280–90. pmid:26616295
  27. 27. Hunt PW. HIV and inflammation: mechanisms and consequences. Curr HIV/AIDS Rep. 2012;9(2):139–47. pmid:22528766
  28. 28. Stylianou E, Bjerkeli V, Yndestad A, Heggelund L, WÆHre T, DamÅS JK, et al. Raised serum levels of interleukin-18 is associated with disease progression and may contribute to virological treatment failure in HIV-1-infected patients. Clin Exp Immunol. 2003;132(3):462–6. pmid:12780693
  29. 29. Masumoto J, Taniguchi S, Nakayama J, Shiohara M, Hidaka E, Katsuyama T, et al. Expression of apoptosis-associated speck-like protein containing a caspase recruitment domain, a pyrin N-terminal homology domain-containing protein, in normal human tissues. J Histochem Cytochem. 2001;49(10):1269–75. pmid:11561011
  30. 30. Chivero ET, Guo ML, Periyasamy P, Liao K, Callen SE, Buch S. HIV-1 Tat primes and activates microglial NLRP3 inflammasome-mediated neuroinflammation. J Neurosci. 2017.
  31. 31. Walsh JG, Reinke SN, Mamik MK, McKenzie BA, Maingat F, Branton WG, et al. Rapid inflammasome activation in microglia contributes to brain disease in HIV/AIDS. Retrovirology. 2014;11(35):1–18.
  32. 32. 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(4):234–48. pmid:26965674
  33. 33. Atluri VSR, Pilakka-Kanthikeel S, Garcia G, Jayant RD, Sagar V, Samikkannu T, et al. Effect of Cocaine on HIV Infection and Inflammasome Gene Expression Profile in HIV Infected Macrophages. Sci Rep. 2016;6(1):1–12.
  34. 34. Guo H, Gao J, Taxman DJ, Ting JPY, Su L. HIV-1 Infection Induces Interleukin-1β Production via TLR8 Protein-dependent and NLRP3 Inflammasome Mechanisms in Human Monocytes. J Biol Chem. 2014;289(31):21716–26. pmid:24939850
  35. 35. Mamik MK, Hui E, Branton WG, McKenzie BA, Chisholm J, Cohen EA, et al. HIV-1 Viral Protein R Activates NLRP3 Inflammasome in Microglia: implications for HIV-1 Associated Neuroinflammation. J Neuroimmune Pharmacol. 2016;Epub ahead of prin:1–16.
  36. 36. French MA, King MS, Tschampa JM, da Silva BA, Landay AL. Serum immune activation markers are persistently increased in patients with HIV infection after 6 years of antiretroviral therapy despite suppression of viral replication and reconstitution of CD4+ T cells. J Infect Dis. 2009;200(8):1212–5. pmid:19728788
  37. 37. Mehandru S. The Gastrointestinal Tract in HIV-1 Infection: Questions, Answers, and More Questions! PRN. 2007;12:1–10.
  38. 38. Barouch DH, Ghneim K, Bosche WJ, Li Y, Berkemeier B, Hull M, et al. Rapid Inflammasome Activation following Mucosal SIV Infection of Rhesus Monkeys. Cell. 2016;165(3):656–67. pmid:27085913
  39. 39. Ahmad R, Sindhu ST, Toma E, Morisset R, Ahmad A. Elevated levels of circulating interleukin-18 in human immunodeficiency virus-infected individuals: role of peripheral blood mononuclear cells and implications for AIDS pathogenesis. J Virol. 2002;76(24):12448–56. pmid:12438570
  40. 40. Iannello A, Boulassel MR, Samarani S, Tremblay C, Toma E, Routy JP, et al. HIV-1 causes an imbalance in the production of interleukin-18 and its natural antagonist in HIV-infected individuals: implications for enhanced viral replication. J Infect Dis. 2010;201(4):608–17. pmid:20078197
  41. 41. Lim HX, Hong HJ, Jung MY, Cho D, Kim TS. Principal role of IL-12p40 in the decreased Th1 and Th17 responses driven by dendritic cells of mice lacking IL-12 and IL-18. Cytokine. 2013;63(2):179–86. pmid:23697689
  42. 42. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677–88. pmid:16782025
  43. 43. Neumann D, Kollewe C, Martin MU, Boraschi D. The membrane form of the type II IL-1 receptor accounts for inhibitory function. J Immunol. 2000;165(6):3350–7. pmid:10975853
  44. 44. Monroe KM, Yang Z, Johnson JR, Geng X, Doitsh G, Krogan NJ, et al. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science. 2014;343(6169):428–32. pmid:24356113
  45. 45. Chattergoon MA, Latanich R, Quinn J, Winter ME, Buckheit RW, Blankson JN, et al. HIV and HCV Activate the Inflammasome in Monocytes and Macrophages via Endosomal Toll-Like Receptors without Induction of Type 1 Interferon. PLoS Pathog. 2014;10(5).
  46. 46. Gracie JA, Robertson SE, McInnes IB. Interleukin-18. J Leukoc Biol. 2003;73(2):213–24. pmid:12554798
  47. 47. Duewell P, Kisser U, Heckelsmiller K, Hoves S, Stoitzner P, Koernig S, et al. ISCOMATRIX Adjuvant Combines Immune Activation with Antigen Delivery to Dendritic Cells In Vivo Leading to Effective Cross-Priming of CD8+ T Cells. J Immunol. 2011;187(1):55–63. pmid:21613613
  48. 48. Zhao Y, Shao F. The NAIP-NLRC4 inflammasome in innate immune detection of bacterial flagellin and type III secretion apparatus. Immunol Rev. 2015;265(1):85–102. pmid:25879286
  49. 49. Chavarria-Smith J, Vance RE. The NLRP1 inflammasomes. Immunol Rev. 2015;265(1):22–34. pmid:25879281
  50. 50. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148(6):1258–70. pmid:22424233
  51. 51. Faustin B, Lartigue L, Bruey JM, Luciano F, Sergienko E, Bailly-Maitre B, et al. Reconstituted NALP1 inflammasome reveals two-step mechanism of caspase-1 activation. Mol Cell. 2007;25(5):713–24. pmid:17349957
  52. 52. Taborda NA, Gonzalez SM, Correa LA, Montoya CJ, Rugeles MT. Spontaneous HIV Controllers Exhib it Preserved Immune Parameters in Peripheral Blood and Gastrointestinal Mucosa. J Acquir Immune Defic Syndr. 2015;70(2):115–21. pmid:26102449
  53. 53. Py BF, Kim MS, Vakifahmetoglu-Norberg H, Yuan J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol Cell. 2013;49(2):331–8. pmid:23246432
  54. 54. Neumann K, Ruland J. Kinases conquer the inflammasomes. Nat Immunol. 2013;14(12):1207–8. pmid:24240152