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

Intracellular Overexpression of HIV-1 Nef Impairs Differentiation and Maturation of Monocytic Precursors towards Dendritic Cells

  • Yan Guo,

    Affiliation Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China

  • Wen-Wen Xu,

    Affiliations Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China, Graduate School of the Chinese Academy of Sciences, Beijing, China

  • Jie Song,

    Affiliations Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China, Graduate School of the Chinese Academy of Sciences, Beijing, China

  • Wen Deng,

    Affiliation Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China

  • Di-Qiu Liu ,

    Contributed equally to this work with: Di-Qiu Liu, Hua-Tang Zhang

    liudiqiu@mail.kiz.ac.cn

    Affiliation Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China

  • Hua-Tang Zhang

    Contributed equally to this work with: Di-Qiu Liu, Hua-Tang Zhang

    Affiliation Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China

Abstract

Nef functions as an immunosuppressive factor critical for HIV-1 replication, survival and development of AIDS following HIV-1 infection. What effects Nef exerts on differentiation and maturation of monocytes towards dendritic cells (DCs) remains greatly controversial. In this study, we used THP-1 (human monocytic leukemia cell line) as monocytic DC precursors to investigate how overexpression of HIV-1 Nef influences the processes of differentiation and maturation of dendritic cells. In striking contrast to negative controls, our results showed that morphological and phenotypical changes (CD11c, CD14, CD40, CD80, CD83, CD86, and HLA-DR) occurred on recombinant THP-1 expressing HIV-1 Nef (short for Nef) upon co-stimulation of GM-CSF/IL-4 or GM-CSF/IL-4/TNF-α/ionomycin. Moreover, CD4, CCR5, and CXCR4 were also down-regulated on Nef. It might be hypothesized that Nef prevents superinfection and signal transduction in HIV-1 infected monocytes. Collectively, our study demonstrates that long-lasting expression of Nef at high levels indeed retards differentiation and maturation of dendritic cells in terms of phenotype and morphology. We are hopeful that potentially, stable expression of intracellular Nef in vivo may function as a subtle mode to support long-lasting HIV-1 existence.

Introduction

Nef, as one of the accessory proteins of HIV-1, has been illustrated in a number of studies on human HIV-1 patients and animal models to function as a major determinant of the pathogenicity of HIV-1 [1], [2]. Monocytes/macrophages are among the principal target cells of HIV-1, and functions of Nef in target cells are generally accepted to account for many aspects of viral pathogenesis [3], [4], [5]. As a regulatory protein expressed earliest and most abundantly in the viral infection cycle, Nef (27kDa) is expressed in the cytoplasm and membrane of infected cells [6], [7], [8]. Although HIV-1 Nef is composed of only 206 amino acids, it is functionally complex, structurally reflected by overlapping effector domains that interact with multiple cellular proteins [9]. The best-characterized function of Nef is its ability to significantly reduce the HIV-1 receptor CD4. Nef removes CD4 from the cell surface by enhancing its endocytosis via recruitment to AP-2 adapter complexes and directing the receptor to lysosomes for degradation [1], [10], [11]. Another well-conserved and defined property of Nef is its ability to down-regulate the cell surface expression of MHC class I (MHC-I) on different cells. In addition, Nef reduces MHC-I expression through the recruitment of AP-1 to the MHC-I cytoplasmic tail to re-route MHC-I from the trans-Golgi network to lysosomes [1], [11], [12].

Nef is involved in the impairment of immune responses in several ways [13], [14]. Nef has strong regulatory power to exert suppressing or enhancing effects on cells’ activation and differentiation. HIV-1 impairs the functions of CD4+ T cells, CD8+T cells, B cells and natural killer cells [15]. However, how HIV-1 Nef influences dendritic cells (DCs) responsible for initiating and modulating immunity remains unclear [16], [17], [18], [19], [20]. Our recent studies have indicated that intracellular over-expression of Rev, Tat, Vif, Vpr and Vpu could induce apoptosis of THP-1 except for Nef.

CD14+ monocytes belong to the mononuclear phagocytes system, and initially described as circulating precursors for tissue macrophages. Monocytes have been generally confirmed to act as precursors with the capacity to differentiate into distinct DCs’ subsets under various physiological conditions [21], [22], [23], displaying remarkably diverse functions allowing them to perform multiple defensive functions, from eliminating pathogen by phagocytosis to stimulating antigen-specific T cells’ responses [24]. Nef protects human monocyte-derived macrophages from HIV-1-induced apoptosis, which is one mechanism of forming HIV-1 reservoirs in vivo [25]. A large number of studies focus on the well-differentiated DCs interaction with HIV-1 [26], [27], [28]. Moreover, the primary site for HIV-1 entry into body is either blood or mucosal surfaces where circulating monocytes readily encounter HIV-1. Located at the boundary between the inside and outside environments, DCs provide one bridge between innate and adaptive immunity [21]. As sentinels throughout the body, DCs capture, process antigen and undergo maturation, and subsequently migrate to the secondary lymphoid tissues, where they present the processed antigens to naive T cells, and initiate adaptive immune responses. Taken together, DCs derived from either human CD14+ monocytes or CD34+ hematopoietic progenitor cells (HPCs) must be mature to be capable of strongly stimulating allogeneic T cells’ responses, but immature ones do not as well as precursors. However, effects exerted by Nef on maturation and differentiation of DCs from monocytes are less well-understood, prompting us to examine how HIV-1 Nef affects differentiation of DCs from monocytes. DCs, as professional antigen presenting cells (APCs) arising from either CD34+ bone marrow progenitor cells or CD14+ monocytes, fulfill critical roles in regulating the immune system [29].

thumbnail
Figure 1. Establishment of stable cell lines Nef and N2.

The THP-1 cell was transfected with pEGFP-nef and pEGFP-N2 respectively, giving rise to Nef and N2 following the limiting dilution in the presence of 550 µg/ml of G418 within 4 weeks. (A): The visualized PCR product on an ethidium bromide stained 1.2% agarose gel, without any template as controls; (B): a, THP-1 in light background; b, THP-1 in black background; c, EGFP expression by N2 in light background; d, EGFP expression by N2 in black background; e, Nef-EGFP expression by Nef in light background; f, Nef-EGFP expression by Nef in black background. Microscopy of cells in culture at 400X.magnification.

https://doi.org/10.1371/journal.pone.0040179.g001

Human monocytic leukemia cell line THP-1 has been identified as a highly reproducible model for the differentiation of immature DCs (iDCs) and mature DCs (mDCs) in terms of phenotypic, morphologic and functional properties of DCs generated from CD14+ monocytes or CD34+ HPCs [30], [31] and it has been prove to function as an extremely powerful tool to investigate human DCs’ differentiation and maturation processes. In this paper, we demonstrate for the first time how intracellular over-expression of HIV-1 Nef is able to retard differentiation and maturation of monocytic precursors towards DCs [32], [33].

thumbnail
Figure 2. Direct comparisons between phenotype of THP-1, N2 and Nef.

Surface expressions of CD11c, CD14, CD40,CD80,CD83,CD86,HLA-DR of stable cell lines–Nef and N2 (control) within 4 weeks were determined via flow cytometry following establishment. The histograms showed a direct comparison of CD11c,CD14, CD40,CD80,CD83,CD86,HLA-DR levels for Nef vs N2 and THP-1 Blue line, Nef; green line, N2; red line, THP-1. One representative of three performed experiments was presented.

https://doi.org/10.1371/journal.pone.0040179.g002

Results

Recombinant THP-1 Stably Expressed Nef

The plasmid pEGFP-N2 or pEGFP-nef was transfected into THP-1 and then subjected to the first round selection under 100 µg/ml G418 within 4 days and second selection under 550 µg/ml G418 within 4 weeks, finally giving rise to recombinant cell lines pEGFP-N2 (short for N2) and pEGFP-Nef (short for Nef) and stably expressing HIV-1 Nef. No viable cell was observed in the untransfected cells (short for THP-1) under the same experimental conditions. The gene nef was successfully transcribed in recombinant cell line Nef, but not in THP-1 and N2 (Fig.1A). Strong green fluorescence was continuously observed in long-lasting drug-resistant cell lines N2 and Nef by fluorescence microscopy (Fig.1B). On the same conditions, we tried to construct recombinant THP-1 expressing Vpr, Vpu, Vif, Tat and Rev, but each attempt proved abortive (data not shown).

thumbnail
Figure 3. Comparative morphology and phenotype between THP-1, N2 and Nef upon co-stimulation with GM-CSF and IL-4 (5-day culture based on “5+0” protocol).

(A): morphology of THP-1, N2 and Nef was observed in light background (upper panel) and black one (lower panel) at magnification of 400×; (B): The histograms showed a direct comparison of CD11c,CD14, CD40,CD80,CD83,CD86,HLA-DR levels for Nef vs N2 and THP-1. Blue line, Nef; green line, N2; red line, THP-1. One representative of three performed experiments was presented.

https://doi.org/10.1371/journal.pone.0040179.g003

Nef Induced Different Phenotypes in THP-1 Compared with the Untransfected and N2 Prior to Co-stimulation

We analyzed phenotype (CD11c, CD14, CD40, CD80, CD83, CD86 and HLA-DR) changes to THP-1,N2 and Nef prior to stimulation with cytokines. As shown in Fig.2, there was very little difference between THP-1 and N2 in the context of these surface markers. It could be hypothesized that GFP hardly interferes with Nef’s biological functions. The levels of CD83 and HLA-DR for Nef vs THP-1 and N2 had nearly no difference. There was a fine distinction between Nef and THP-1/N2 in terms of CD80 or CD40. By contrast, one sharp difference exists between Nef and the latter two upon CD11c, CD14 and CD86. Thus, we initially concluded that Nef expressed in THP-1monocytic precursor for DCs played a certain role.

thumbnail
Figure 4. Comparative morphology and phenotype between THP-1, N2 and Nef upon co-stimulation with GM-CSF,IL-4,TNFα and ionomycin.

(A): morphology of the above cell types was observed in light background (upper panel) and black one (lower panel) at magnification of 400X; (B): The histograms showed a direct comparison of CD11c, CD14, CD40,CD80,CD83,CD86,HLA-DR levels for Nef vs N2 and THP-1,(7-day culture based on “5+2” protocol ); (C): The histograms showed a direct comparison of above surface markers expression levels for Nef vs N2 and THP-1 (2-day culture based on “0+2” protocol). One representative of three performed experiments was presented.

https://doi.org/10.1371/journal.pone.0040179.g004

thumbnail
Figure 5. Nef down-regulated CD4, CCR5 and CXCR4 on Nef vs THP-1 and N2.

The histogram shows a direct comparison. Blue line, Nef; green line, N2; red line, THP-1. (A), unstimulated; (B), 5-day culture; (C), 7-day culture;(D): 2-day culture. One representative of three performed experiments was presented.

https://doi.org/10.1371/journal.pone.0040179.g005

Nef Impaired THP-1 Differentiation towards iDCs

On the basis of stable cell lines, rhIL-4 (100 ng = 1500 IU/ml) and rhGM-CSF (100 ng = 1500 IU/ml) were added into three cultures for 5 days (5-day culture), and subsequently flow cytometry and fluorescence microscopy was used to analyze alterations in phenotype and morphology. After 5-day culture in the presence of GM-CSF/IL-4, THP-1 and N2 appeared to have characteristic stellate morphology, but Nef did not (Fig.3A). As shown in (Fig.3B), the levels of CD14, CD83, CD86 were supposed to be almost indistinguishable between Nef, N2 and THP-1; fine differences existed between Nef and the latter two upon the level of CD80; furthermore, the level of CD11c, CD40 and HLA-DR of Nef were significantly lower than N2 and THP-1. From this, Nef impaired THP-1 differentiation towards immature DCs.

Nef Retarded DCs’ Maturation

TNF-a/ionomycin, as differentiating inducer of mature DCs from THP-1, was supplemented into the above 5-day culture for 2 days (7-day culture). Post co-stimulation with GM-CSF/IL-4/TNF-α/ionomycin, THP-1 and N2 displayed typical stellate morphology, but Nef still kept its original shape without any changes (Fig.4A). Nef was significantly lower in the levels of CD11c, CD14, CD40, CD83, CD86 and HLA-DR than THP-1 and N2, and appeared slightly higher in the level of CD80 than the other two (Fig.4B), consistent with the results derived from 2-day culture based on “0+2” protocol (Fig.4C). Thus, Nef prevents DCs’ maturation from iDCs or THP-1.

Nef Down-regulated HIV-1 Receptors on THP-1

Based on unstimulation, GM-CSF/IL-4 stimulation (immature, 5-day culture), GM-CSF/IL-4/TNF-a/ionomycin (mature, 7-day and 2-day culture), Nef displayed significantly lower levels of CD4, CCR5 and CXCR4 just equivalent to background levels of isotypic antibody staining (data not shown) in striking contrast to THP-1and N2 (Fig.5).

Discussion

Numerous studies have identified and adopted THP-1 (human monocytic leukemia cell line) in vitro modeling CD14+ monocytes or CD34+ HPCs that readily differentiate into iDCs and mDCs. THP-1-derived monotypic DCs appear highly pure and displayed the morphologic, phenotypic and functional properties of human CD14+ monocytes- or CD34+ HPCs-derived DCs, also including endocytotic activity, differentiation associated and IFN-γ enhanceable expression of immunoproteasome subunits and proteasome activator PA28α, and strong T-cell stimulatory capacity [31], [33], [34]. Thus, THP-1 represents a robust and reproducible model to study the effects of Nef on monocyte/DCs biology. Moreover, differentiated THP-1 reflects significant differences between the immature and mature stages in the phenotype in vitro [30]. We believe the implications of our results for THP-1 function can be extrapolated to naturally isolated CD14+ monocytes, although we clearly cannot rule out the possibility that the effects of Nef expression might be subtly different in natural monocytes.

Over past several years, a large amount of studies focused on Nef’s effects on iDCs developed into mDCs [26], [28], [33], [35], [36]. iDCs have been shown to be immunosuppressive due to their ability to selectively induce the generation and expansion of regulatory T cells (Tregs).Thus, to acquire functional immunostimulating capacity, DCs need to be mature. DCs’ abilities to capture, process and present antigens, activate and modulate T cells, B cells and NK responses, position them at the forefront of therapeutic applications against HIV-1 infection, which has been well documented [19], [37]. However, there is little information available about the effects exerted by Nef on monocytic precursors’ differentiation and maturation towards DCs. Our experimental results revealed that Nef impaired differentiation and maturation from monocytic precursors into DCs in terms of morphological and phenotypic alterations.

On the same conditions, we tried to construct stable cell lines expressing vif, vpu, vpr, rev and tat, respectively, which ended in failure. These accessory proteins caused THP-1 apoptosis, reflected by Annexin V and PI via flow cytometry (data not shown). This finding also confirms that in advanced stages of HIV-1 infection, a number of DCs diminished with reduced antigen-presenting ability, and per se circulating DCs are rare (<1%) in peripheral blood mononuclear cells (PBMCs) [21], [29]. However, it remains debatable that Nef is associated with a decreased number and dysfunction of DCs during HIV infection. Our results show that expression of nef did not result in THP-1 apoptosis. As might be expected, Nef down-regulated the CD4/CCR5/CXCR4 on THP-1, which similarly happened to CD4+ T cells in a previous study [10]. Moreover, the cell line Nef completely lost phagocytic capacity in presence of GM-CSF/IL-4 for 5 days in our previous works (unpublished results). These suggest that HIV-1 replicates and latently exists in targets cells for a long time, most likely by restricting superinfection or avoiding apoptosis and necrosis, and then is well-shielded from immune attack in the advanced stages of HIV-1 infection. Intracellular over-expression of Nef has been hypothesized to function as a subtle approach to support HIV-1 replication, survival, latency and escaping immune along with progression of HIV-1 infection. Despite circulating monocytes resistant to productive HIV-1 infection ex vivo prior to differentiation into macrophages, HIV-1-infected monocytes have been identified in the peripheral blood of viremic and highly active antiretroviral therapy (HAART)-treated patients, and are considered contributors of viral persistence [38], [39].

Granelli-Piperno et al (2004) reported that HIV-1-infected monocytes-derived DCs do not undergo maturation [40]. Nevertheless, another report draws an opposite conclusion, that HIV-1 can induce maturation of monocyte-derived DCs to facilitate subsequent delivery to T cells. This contradiction might result from different stages of HIV infection. Therefore, it remains to be further investigated whether or not Nef plays a decisive role in impairing monocytes’ differentiation and maturation towards mDCs. Interestingly, in this study Nef-expressing THP-1 displayed lower levels of CD14 as compared to N2 and THP-1 prior to and post-co-stimulation possibly because THP-1 originated from neoplastically transformed cell lines, and are unable to activate signal transduction pathways involved in up-regulation of CD14 production [41], [42]. CD14 is the first described pattern recognition receptor which also recognizes other pathogen-associated molecular patterns besides lipopolysaccharide (LPS).

In summary, it is reported for the first time that intracellular expression of HIV-1 Nef impairs differentiation and maturation from THP-1 to DCs. Extrapolating our conclusions into CD14+ monocytes and CD34+ HPCs could also be very worthwhile. Our study offers further insights to help settle the arguments raised by numerous studies on how HIV-1 interferes with immunological functions of DCs. Although we only used a single cell line as a research model throughout the study, it provided the basis for further study in to DCs’ immunological functions in HIV-1 infectious situations. Certainly THP-1 can function as stable and suitable model for HIV-1 infected monocytes/DCs, which need to be further and systematically verified and re-examined using PBMCs as model. We have reasons to speculate that Nef most likely interrupted crosstalk between innate and adaptive immunity during HIV-1 infection via inhibiting monocytes’ differentiation into DCs, especially given the resulting drastic decline in the number of DCs during later stages of HIV-1 infection [43], [44]. Meanwhile, HIV-1 builds up more reservoirs for itself in vivo. The different stimuli might confer specific effects on monocyte-derived DCs, reflecting the differentiating plasticity of monocytes. Precisely how Nef interplays with monocytes in vitro or in vivo under various conditions therefore remains to be explained.

Materials and Methods

DNA Procedure

Nef gene, derived from SF2 strain of HIV-1, was obtained from the plasmid pHXBnPLAP-IRES-N+(a gift from Prof. Xiaoning Xu, Oxford University, UK) via PCR reaction with specific primers (forward:CGCGGATCCATGGGTGGACAAGTGGTCA, underlined BamH Isite; reverse: CGCAAGCTTTCAGCAGTCTTTGTAGTACTCCGGAT, underlined Hind III site ) performed over 30 cycles at 95°C (1 min), 50°C (1 min), 72°C (1 min), followed by 72°C for 10 min. The PCR product was visualized on an ethidium bromide stained 1.2% agarose gel, and it was recovered and purified with TIANgel Mini Purification Kit (Tiangen, Beijing, China). Subsequently, this pre-treated product with BamH I/Hind III underwent another round of purification, and were inserted into the corresponding sites of pEGFP-N2 in frame giving rise to pEGFP-Nef and sequenced prior to and after introducing into THP-1.

RT-PCR

Total RNA extraction and first-strand cDNA synthesis were performed as previously mentioned [45]. The sequences of PCR primer pairs for Nef as above, and for GAPDH as positive control were as follows: forward, 5′-CCATCACCATCTTCCAGGAGCGAG-3′; reverse, 5′CAAAGGTG GAGGAAGTGGGTGTCG-3′. The cDNA template was used in PCR reaction performed over 5 min at 95°C, subsequently 30 cycles at 95°C (1 min), 55°C (1 min), 72°C (1 min), followed by 72°C for 10 min. The PCR product was visualized on an ethidium bromide stained 1.2% agarose gel.

Construction of Recombinant THP-1

THP-1 was obtained from American Type Culture Collection and cultured in complete RPMI medium (RPMI-1640 with 25 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, 100 µg/ml streptomycin and 10% heat-inactivated fetal calf serum). In this study, we established a THP-1 clone expressing a fusion protein composed of Nef and EGFP by introducing pEGFP-Nef plasmid using Cell Line Nucleofector Kit V(Amaxa/Lonza, US) [34] for THP-1 cells according to manufacturer’s recommendations, and cells harboring pEGFP-N2 and without any treatment as controls.

For drug selection of transfected THP-1, the cell culture was first incubated at 37°C for 48 h. The non-adherent cells were decanted and the culture replenished daily with fresh medium containing 100 µg/ml G418 (Sigma) for 4 days. The culture was then replenished every other day with a fresh medium containing 550 µg/ml of G418. Stable drug-resistant cell lines THP-1-EGFP (N2) or THP-1-Nef-EGFP (Nef) clones were established within 4 weeks by limiting dilution in the presence of 550 µg/ml of G418. These recombinant cells were maintained in exponential growth by serial passage in tissue culture flasks in complete RPMI in a humidified incubator at 37°C and 5% CO2.

Detection by Fluorescence Microscopy

After 16 h post-incubation, cultured cells were examined under fluorescence microscopy (Olympus BX51, Tokyo, Japan). Then, the observation of transient EGFP cell was taken once every 12 h till no green fluorescence was detected. Cultured transfected cells were examined once every 12 h till the transgenic cell lines were obtained. Prior to observation, cultures in capped glass culture tubes were first incubated at 4°C for an hour to allow EGFP maturation. Cells were then washed (centrifuged for 10 min at 2,000×g) in ice-cold PBS and fixed in cold 2% formaldehyde. In order to calculate the transfection efficiency, the number of fluorescent THP-1 was counted using a hemocytometer. Where large numbers of cells were present, further dilutions were required.

Cell Count and Flow Cytometry

Cell counting was performed using a Neubauer counting chamber (Hawksely, UK) with at least 200 cells being counted per sample. Cell viability was assessed using trypan blue dye exclusion.

For surface staining of CD11c, CD14, CD40, CD80, CD83, CD86, HLA-DR, CD4, CCR5 and CXCR4, respectively, cultured cells were washed, suspended at 2×105 cells in 200-µl cold FACS solution and incubated with PE-conjugated mAbs or appropriate isotypic controls for 30 min. All antibodies were obtained from BD Biosciences (San Jose, CA, USA). Cells were then washed twice and resuspended in 300 µl of cold FACS solution. Stained cells were analyzed for two-color immunofluorescence and EGFP expression with a FACSCalibur flow cytometer (BD Immunocytometry Systems, San Jose, CA, USA) assisted by Cell Quest software (BD Pharmingen). A minimum of 104 cells were analyzed for each sample. Cell debris was excluded from the analysis by setting a gate on forward and side scatter to include only cells which displayed the light-scattering properties of viable cells. List mode data were analyzed using FlowJo 8 (Treestar, Ashland, OR, USA).

Inducing Differentiation of Immature DCs

THP-1, THP-1-EGFP (N2) and THP-1-Nef-EGFP (Nef) cells were harvested by centrifugation, resuspended in complete RPMI at a concentration of 2×105 cells/ml, and transferred in a final volume of 18 ml into 6-well culture plates. To induce differentiation, rhIL-4 (100 ng = 1500 IU/ml) and rhGM-CSF (100 ng = 1500 IU/ml) were added. Cells were cultured for 5 days to acquire the properties of immature DCs (5-day culture). Medium exchange was performed every 2 days with fresh cytokine-supplemented medium in a humidified incubator at 37°C and 5% CO2, which meant “5+0” protocol [29].

Inducing Differentiation of Mature DCs

Mature DCs were generated from immature DCs (5-day culture) by addition of rhTNF-a (20 ng/ml  = 2000 IU/ml) and 200 ng/ml ionomycin into the medium for 2 days, which named 7-day culture based on “5+2” protocol [29].

Alternatively, THP-1, N2 and Nef cells were harvested by centrifugation, resuspended in serum-free culture medium (without 10% FCS) at a concentration of 2×106 cells/ml, and transferred in a final volume of 18 ml into 6-well culture plates. The following optimized cytokine cocktails were added: rhIL-4 (200 ng/ml  = 3000 IU/ml), rhGM-CSF (100 ng/ml  = 1500 IU/ml), rhTNF-a (20 ng/ml  = 2000 IU/ml), and 200 ng/ml ionomycin. Cells were cultured for 2 days (“0+2” protocol) in a humidified incubator at 37°C and 5% CO2 and used for experiments, which named 2-day culture [29].

Acknowledgments

We wish to thank Prof. Xiaoning Xu for providing the plasmid pHXBnPLAP-IRES-N+ harbouring HIV-1 nef of strain SF2.

Author Contributions

Conceived and designed the experiments: HTZ. Performed the experiments: YG DQL WWX JS. Analyzed the data: DQL HTZ YG. Contributed reagents/materials/analysis tools: JS WD. Wrote the paper: DQL HTZ.

References

  1. 1. Foster JL, Denial SJ, Temple BR, Garcia JV (2011) Mechanisms of HIV-1 Nef function and intracellular signaling. J Neuroimmune Pharmacol 6: 230–246.
  2. 2. de Ronde A, Klaver B, Keulen W, Smit L, Goudsmit J (1992) Natural HIV-1 NEF accelerates virus replication in primary human lymphocytes. Virology 188: 391–395.
  3. 3. Tangsinmankong N, Day NK, Good RA, Haraguchi S (2000) Monocytes are target cells for IL-10 induction by HIV-1 Nef protein. Cytokine 12: 1506–1511.
  4. 4. Carreer R, Groux H, Ameisen JC, Capron A (1994) Role of HIV-1 Nef expression in activation pathways in CD4+ T cells. AIDS Res Hum Retroviruses 10: 523–527.
  5. 5. Lehmann MH, Walter S, Ylisastigui L, Striebel F, Ovod V, et al. (2006) Extracellular HIV-1 Nef increases migration of monocytes. Exp Cell Res 312: 3659–3668.
  6. 6. Tokarev A, Guatelli J (2011) Misdirection of membrane trafficking by HIV-1 Vpu and Nef: Keys to viral virulence and persistence. Cell Logist 1: 90–102.
  7. 7. Sugiyama R, Naganuma H, Nishitsuji H, Takaku H (2011) Human immunodeficiency virus-1 Nef suppresses Hsp70-mediated Tat activation. FEBS Lett 585: 3367–3371.
  8. 8. Sowrirajan B, Barker E (2011) The Natural Killer Cell Cytotoxic Function Is Modulated by HIV-1 Accessory Proteins. Viruses 3: 1091–1111.
  9. 9. Morgan CR, Miglionico BV, Engen JR (2011) Effects of HIV-1 Nef on human N-myristoyltransferase 1. Biochemistry 50: 3394–3403.
  10. 10. Sloan RD, Donahue DA, Kuhl BD, Bar-Magen T, Wainberg MA (2010) Expression of Nef from unintegrated HIV-1 DNA downregulates cell surface CXCR4 and CCR5 on T-lymphocytes. Retrovirology 7: 44.
  11. 11. Gray LR, Gabuzda D, Cowley D, Ellett A, Chiavaroli L, et al. (2011) CD4 and MHC class 1 down-modulation activities of nef alleles from brain- and lymphoid tissue-derived primary HIV-1 isolates. J Neurovirol 17: 82–91.
  12. 12. Lubben NB, Sahlender DA, Motley AM, Lehner PJ, Benaroch P, et al. (2007) HIV-1 Nef-induced down-regulation of MHC class I requires AP-1 and clathrin but not PACS-1 and is impeded by AP-2. Mol Biol Cell 18: 3351–3365.
  13. 13. Hiyoshi M, Takahashi-Makise N, Yoshidomi Y, Chutiwitoonchai N, Chihara T, et al. (2012) HIV-1 Nef perturbs the function, structure, and signaling of the golgi through the Src kinase Hck. J Cell Physiol 227(3): 1090–1097.
  14. 14. Breuer S, Schievink SI, Schulte A, Blankenfeldt W, Fackler OT, et al. (2011) Molecular design, functional characterization and structural basis of a protein inhibitor against the HIV-1 pathogenicity factor Nef. PLoS One 6: e20033.
  15. 15. Chen X, Wang B, Chang LJ (2006) Induction of primary anti-HIV CD4 and CD8 T cell responses by dendritic cells transduced with self-inactivating lentiviral vectors. Cell Immunol 243: 10–18.
  16. 16. van der Vlist M, van der Aar AM, Gringhuis SI, Geijtenbeek TB (2011) Innate signaling in HIV-1 infection of dendritic cells. Curr Opin HIV AIDS 6: 348–352.
  17. 17. Smed-Sorensen A, Lore K (2011) Dendritic cells at the interface of innate and adaptive immunity to HIV-1. Curr Opin HIV AIDS 6: 405–410.
  18. 18. Poudrier J, Roger M (2011) Dendritic cell status modulates the outcome of HIV-related B cell disease progression. PLoS Pathog 7: e1002154.
  19. 19. Patham B, Simmons GL, Subramanya S (2011) Advances in Dendritic Cell-Based Vaccines for HIV. Curr Med Chem 18: 3987–3994.
  20. 20. Huang J, Yang Y, Al-Mozaini M, Burke PS, Beamon J, et al. (2011) Dendritic cell dysfunction during primary HIV-1 infection. J Infect Dis 204: 1557–1562.
  21. 21. Leon B, Lopez-Bravo M, Ardavín C (2005) Monocyte-derived dendritic cells. Waltham, MA: Elsevier. pp. 313–318.
  22. 22. Schmid MA, Kingston D, Boddupalli S, Manz MG (2010) Instructive cytokine signals in dendritic cell lineage commitment. Immunol Rev 234: 32–44.
  23. 23. Ginhoux F, Tacke F, Angeli V, Bogunovic M, Loubeau M, et al. (2006) Langerhans cells arise from monocytes in vivo. Nat Immunol 7: 265–273.
  24. 24. Langenkamp A, Messi M, Lanzavecchia A, Sallusto F (2000) Kinetics of dendritic cell activation: impact on priming of TH1, TH2 and nonpolarized T cells. Nature immunology 1: 311–316.
  25. 25. Olivetta E, Federico M (2006) HIV-1 Nef protects human-monocyte-derived macrophages from HIV-1-induced apoptosis. Exp Cell Res 312: 890–900.
  26. 26. Messmer D, Bromberg J, Devgan G, Jacque JM, Granelli-Piperno A, et al. (2002) Human immunodeficiency virus type 1 Nef mediates activation of STAT3 in immature dendritic cells. AIDS Res Hum Retroviruses 18: 1043–1050.
  27. 27. Quaranta MG, Mattioli B, Spadaro F, Straface E, Giordani L, et al. (2003) HIV-1 Nef triggers Vav-mediated signaling pathway leading to functional and morphological differentiation of dendritic cells. FASEB J 17: 2025–2036.
  28. 28. Quaranta MG, Mattioli B, Giordani L, Viora M (2006) The immunoregulatory effects of HIV-1 Nef on dendritic cells and the pathogenesis of AIDS. FASEB J 20: 2198–2208.
  29. 29. Takeuchi S, Furue M (2007) Dendritic cells: ontogeny. Allergol Int 56: 215–223.
  30. 30. Berges C, Naujokat C, Tinapp S, Wieczorek H, Hoh A, et al. (2005) A cell line model for the differentiation of human dendritic cells. Biochem Biophys Res Commun 333: 896–907.
  31. 31. Cho MH, Ahn HJ, Ha HJ, Park J, Chun JH, et al. (2010) Bacillus anthracis capsule activates caspase-1 and induces interleukin-1beta release from differentiated THP-1 and human monocyte-derived dendritic cells. Infect Immun 78: 387–392.
  32. 32. Ogasawara N, Kojima T, Go M, Fuchimoto J, Kamekura R, et al. (2009) Induction of JAM-A during differentiation of human THP-1 dendritic cells. Biochem Biophys Res Commun 389: 543–549.
  33. 33. Lambrechts N, Verstraelen S, Lodewyckx H, Felicio A, Hooyberghs J, et al. (2009) THP-1 monocytes but not macrophages as a potential alternative for CD34+ dendritic cells to identify chemical skin sensitizers. Toxicol Appl Pharmacol 236: 221–230.
  34. 34. Maess MB, Buers I, Robenek H, Lorkowski S (2011) Improved protocol for efficient nonviral transfection of premature THP-1 macrophages. Cold Spring Harb Protoc 2011. https://doi.org/10.1101/pdb.prot5612
  35. 35. Wang X, Ye L, Zhou Y, Liu MQ, Zhou DJ, et al. (2011) Inhibition of anti-HIV microRNA expression: a mechanism for opioid-mediated enhancement of HIV infection of monocytes. Am J Pathol 178: 41–47.
  36. 36. Tippett E, Cheng WJ, Westhorpe C, Cameron PU, Brew BJ, et al. (2011) Differential expression of CD163 on monocyte subsets in healthy and HIV-1 infected individuals. PLoS One 6: e19968.
  37. 37. Niu L, Termini JM, Kanagavelu SK, Gupta S, Rolland MM, et al. (2011) Preclinical evaluation of HIV-1 therapeutic ex vivo dendritic cell vaccines expressing consensus Gag antigens and conserved Gag epitopes. Vaccine 29: 2110–2119.
  38. 38. Coquillard G, Patterson BK (2009) Determination of hepatitis C virus-infected, monocyte lineage reservoirs in individuals with or without HIV coinfection. J Infect Dis 200: 947–954.
  39. 39. Spivak AM, Salgado M, Rabi SA, O’Connell KA, Blankson JN (2011) Circulating monocytes are not a major reservoir of HIV-1 in elite suppressors. J Virol 85: 10399–10403.
  40. 40. Granelli-Piperno A, Golebiowska A, Trumpfheller C, Siegal FP, Steinman RM (2004) HIV-1-infected monocyte-derived dendritic cells do not undergo maturation but can elicit IL-10 production and T cell regulation. Proc Natl Acad Sci U S A 101: 7669–7674.
  41. 41. Creery D, Angel JB, Aucoin S, Weiss W, Cameron WD, et al. (2002) Nef protein of human immunodeficiency virus and lipopolysaccharide induce expression of CD14 on human monocytes through differential utilization of interleukin-10. Clin Diagn Lab Immunol 9: 1212–1221.
  42. 42. Nockher WA, Bergmann L, Scherberich JE (1994) Increased soluble CD14 serum levels and altered CD14 expression of peripheral blood monocytes in HIV-infected patients. Clin Exp Immunol 98: 369–374.
  43. 43. Martinez-Pomar N, Raga S, Ferrer J, Pons J, Munoz-Saa I, et al. (2006) Elevated serum interleukin (IL)-12p40 levels in common variable immunodeficiency disease and decreased peripheral blood dendritic cells: analysis of IL-12p40 and interferon-γ gene. Clinical & Experimental Immunology 144: 233–238.
  44. 44. Bergamaschi A, Pancino G (2010) Host hindrance to HIV-1 replication in monocytes and macrophages. Retrovirology 7: 31.
  45. 45. Hayakawa E, Yoshimoto T, Sekizawa N, Sugiyama T, Hirata Y (2012) Overexpression of receptor for advanced glycation end products induces monocyte chemoattractant protein-1 expression in rat vascular smooth muscle cell line. J Atheroscler Thromb 19: 13–22.