8
HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells Susana Álvarez a , Almudena Blanco b , Florian Kern b , Manuel Fresno a ,M a Ángeles Muñoz-Fernández b, a Centro de Biología Molecular, Consejo Superior de Investigaciones Cientícas-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain b Lab. Inmuno-Biología Molecular, Hospital General Universitario Gregorio Marañón, 28007, Madrid, Spain abstract article info Article history: Received 9 June 2008 Returned to author for revision 2 July 2008 Accepted 10 July 2008 Available online 27 August 2008 Keywords: HIV-2 Human astrocytes Cyclooxygenase NF-κB HIV-2 invades CNS and causes neurological disease as well as HIV-1 does. Induction of COX-2 in CNS of HIV-1 infected people has been proposed as a cause of cognitive impairment, so we tested whether HIV-2 may cause damage by a similar mechanism. COX-2 mRNA and protein expression were induced in human astrocytes upon interaction with HIV-2, being this induction abrogated by CXCR4 antagonists. HIV-2 induced COX-2 promoter transcription and deletion of the two NF-κB binding sites of the promoter abrogated this induction. Neither AP-1 nor NFATseem to be involved in COX-2 transcriptional activation. Overexpression of IκBα completely abrogated COX-2 induction, and transfection of p65/relA NF-κB induced COX-2 transcription. Interestingly, HIV-2 activated NF-κB by inducing p65/relA transactivating activity through Ser536 phosphorylation. Moreover, the astrocyte activation induced by HIV-2 was abrogated by different COX inhibitors. In summary, HIV-2 induces COX-2 in human astrocytes depending on CXCR4 coreceptor. © 2008 Elsevier Inc. All rights reserved. Introduction Both HIV-1 and HIV-2 belong to the primate lentivirussubgenus, and although they present a similar genomic organization, there are important differences between them that provide insights into virus evolution, tropism and pathogenesis. Major differences include reduced pathogenicity of HIV-2 relative to HIV-1, enhanced immune control of HIV-2 infection and often some degree of CD4-indepen- dence for HIV-2 infection (Reeves and Doms, 2002). Despite this lower pathogenicity, HIV encephalitis (HIVE) has been reported as more frequent in brains of HIV-2 infected patients than HIV-1 (Lucas et al., 1993). In addition, the reported independency from CD4 (Endres et al., 1996) to establish a productive infection could account for the higher neuropathogenicity of HIV-2. Recent studies showed that a subset of HIV-1 and HIV-2 isolates were able to replicate in primary adult astrocytes, brain microvascular endothelial cells, and macrophages using an alternative coreceptor(s) to enter the cell (Clapham and McKnight, 2002; Willey et al., 2003). However, since no productive neuronal infection is evident in the brain of HIVE patients, other indirect mechanisms must be responsible for the neuropathological damage observed in the brain of HIV-infected people (Reeves and Doms, 2002). In this regard, recent ndings support the role of cyclooxygenase- 2 (COX-2) in the neuropathogenicity of HIV-1 (Alvarez et al., 2005; Flora et al., 2006). COX is the rate-limiting enzyme in the production of prostaglandins (PGs) from arachidonic acid (AA). Two isoforms have been identied, the constitutively expressed COX-1 and COX-2 that is induced by different stimuli (Smith, DeWitt, and Garavito, 2000; Vane, Bakhle, and Botting, 1998). Human COX-2 promoter contains a classical TATA box and multiple regulatory elements, including two putative nuclear factor (NF)-κB-binding sites, one NF- interleukin (IL)6/CCAAT-enhancer binding protein (cEBP), one acti- vator protein (AP)-1, one cyclic AMP-response element and two binding sites of NF of activated T-cells (NFAT). These sequences have been shown to act as positive regulatory elements for the COX gene transcription in different cell types (Allport et al., 2000; Alvarez et al., 2005; Alvarez et al., 2007; Iniguez et al., 2000). NF-κB is a collective term referring to a class of dimeric trans- cription factors belonging to the rel family. In resting cells, NF-κB exists in the cytoplasm as an inactive complex bound to inhibitory proteins of the IκB family. In response to a variety of stimuli, IκB proteins undergo phosphorylation of Ser32 and Ser36 followed by ubiquitination and subsequent degradation, allowing translocation of active NF-κB to the nucleus. However, there is accumulating evidence suggesting that another level of NF-κB regulation independent on IκB degradation exists. This second level of regulation relies in the activation of the transcriptional activity of p65/relA and c-rel NF-κB members (reviewed in Schmitz, Bacher, and Kracht, 2001; Viatour et al., 2005). Therefore, phosphorylation (Anrather et al., 1999; Sizemore, Leung, and Stark, 1999) and acetylation (Deng, Zhu, and Wu, 2003) of NF-κB represent another important level of regulation. After strong activation, astrocytes secrete various neurotoxic subs- tances and express an enhanced level of glial brillary acidic protein (GFAP), which is considered a marker for astrogliosis (Eng and Virology 380 (2008) 144151 Corresponding author. Fax: +34 91 5868018. E-mail address: [email protected] (M.Á. Muñoz-Fernández). 0042-6822/$ see front matter © 2008 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2008.07.008 Contents lists available at ScienceDirect Virology journal homepage: www.elsevier.com/locate/yviro

HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

Embed Size (px)

Citation preview

Page 1: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

Virology 380 (2008) 144–151

Contents lists available at ScienceDirect

Virology

j ourna l homepage: www.e lsev ie r.com/ locate /yv i ro

HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in humanastroglial cells

Susana Álvarez a, Almudena Blanco b, Florian Kern b, Manuel Fresno a, Ma Ángeles Muñoz-Fernández b,⁎a Centro de Biología Molecular, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spainb Lab. Inmuno-Biología Molecular, Hospital General Universitario Gregorio Marañón, 28007, Madrid, Spain

⁎ Corresponding author. Fax: +34 91 5868018.E-mail address: [email protected] (

0042-6822/$ – see front matter © 2008 Elsevier Inc. Aldoi:10.1016/j.virol.2008.07.008

a b s t r a c t

a r t i c l e i n f o

Article history:

HIV-2 invades CNS and caus Received 9 June 2008Returned to author for revision 2 July 2008Accepted 10 July 2008Available online 27 August 2008

Keywords:HIV-2Human astrocytesCyclooxygenaseNF-κB

es neurological disease as well as HIV-1 does. Induction of COX-2 in CNS of HIV-1infected people has been proposed as a cause of cognitive impairment, so we tested whether HIV-2 maycause damage by a similar mechanism. COX-2 mRNA and protein expression were induced in humanastrocytes upon interaction with HIV-2, being this induction abrogated by CXCR4 antagonists. HIV-2 inducedCOX-2 promoter transcription and deletion of the two NF-κB binding sites of the promoter abrogated thisinduction. Neither AP-1 nor NFAT seem to be involved in COX-2 transcriptional activation. Overexpression ofIκBα completely abrogated COX-2 induction, and transfection of p65/relA NF-κB induced COX-2transcription. Interestingly, HIV-2 activated NF-κB by inducing p65/relA transactivating activity throughSer536 phosphorylation. Moreover, the astrocyte activation induced by HIV-2 was abrogated by differentCOX inhibitors. In summary, HIV-2 induces COX-2 in human astrocytes depending on CXCR4 coreceptor.

© 2008 Elsevier Inc. All rights reserved.

Introduction

Both HIV-1 and HIV-2 belong to the “primate lentivirus” subgenus,and although they present a similar genomic organization, there areimportant differences between them that provide insights into virusevolution, tropism and pathogenesis. Major differences includereduced pathogenicity of HIV-2 relative to HIV-1, enhanced immunecontrol of HIV-2 infection and often some degree of CD4-indepen-dence for HIV-2 infection (Reeves and Doms, 2002). Despite this lowerpathogenicity, HIV encephalitis (HIVE) has been reported as morefrequent in brains of HIV-2 infected patients than HIV-1 (Lucas et al.,1993). In addition, the reported independency from CD4 (Endres et al.,1996) to establish a productive infection could account for the higherneuropathogenicity of HIV-2. Recent studies showed that a subset ofHIV-1 and HIV-2 isolates were able to replicate in primary adultastrocytes, brain microvascular endothelial cells, and macrophagesusing an alternative coreceptor(s) to enter the cell (Clapham andMcKnight, 2002; Willey et al., 2003). However, since no productiveneuronal infection is evident in the brain of HIVE patients, otherindirect mechanisms must be responsible for the neuropathologicaldamage observed in the brain of HIV-infected people (Reeves andDoms, 2002).

In this regard, recent findings support the role of cyclooxygenase-2 (COX-2) in the neuropathogenicity of HIV-1 (Alvarez et al., 2005;Flora et al., 2006). COX is the rate-limiting enzyme in the production

M.Á. Muñoz-Fernández).

l rights reserved.

of prostaglandins (PGs) from arachidonic acid (AA). Two isoformshave been identified, the constitutively expressed COX-1 and COX-2that is induced by different stimuli (Smith, DeWitt, and Garavito,2000; Vane, Bakhle, and Botting, 1998). Human COX-2 promotercontains a classical TATA box and multiple regulatory elements,including two putative nuclear factor (NF)-κB-binding sites, one NF-interleukin (IL)6/CCAAT-enhancer binding protein (cEBP), one acti-vator protein (AP)-1, one cyclic AMP-response element and twobinding sites of NF of activated T-cells (NFAT). These sequences havebeen shown to act as positive regulatory elements for the COX genetranscription in different cell types (Allport et al., 2000; Alvarez et al.,2005; Alvarez et al., 2007; Iniguez et al., 2000).

NF-κB is a collective term referring to a class of dimeric trans-cription factors belonging to the rel family. In resting cells, NF-κBexists in the cytoplasm as an inactive complex bound to inhibitoryproteins of the IκB family. In response to a variety of stimuli, IκBproteins undergo phosphorylation of Ser32 and Ser36 followed byubiquitination and subsequent degradation, allowing translocation ofactive NF-κB to the nucleus. However, there is accumulating evidencesuggesting that another level of NF-κB regulation independent on IκBdegradation exists. This second level of regulation relies in theactivation of the transcriptional activity of p65/relA and c-rel NF-κBmembers (reviewed in Schmitz, Bacher, and Kracht, 2001; Viatouret al., 2005). Therefore, phosphorylation (Anrather et al., 1999;Sizemore, Leung, and Stark, 1999) and acetylation (Deng, Zhu, andWu, 2003) of NF-κB represent another important level of regulation.

After strong activation, astrocytes secrete various neurotoxic subs-tances and express an enhanced level of glial fibrillary acidic protein(GFAP), which is considered a marker for astrogliosis (Eng and

Page 2: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

145S. Álvarez et al. / Virology 380 (2008) 144–151

Ghirnikar, 1994). However, the exact mechanism by which astroglialexpression of GFAP is increased in neurodegenerative CNS remainsunclear.

In this work, we have found that although HIV-2CBL23 does notinfect U-87 cells, it is able to induce COX-2 expression at the trans-criptional level by activating NF-κB. This activation seems to involveSer536 phosphorylation of p65/relA subunit and not IκBα degrada-tion. Finally, we demonstrate that HIV-2 induces an increase in GFAPexpression in U-87 cells, being this effect reverted by COX-2 inhibitors.

Results

HIV-2 induces COX-2 expression in human astrocytes

Most inflammatorymediators have relatively fewactions in healthyCNS tissue and are expressed at very low or undetectable levels.However, they are induced rapidly in response to tissue injury orinfection, and exert diverse actions. In this regard, it has been pre-viously described that HIV-1 induces COX-2 expression in brain cells(Alvarez et al., 2005; Alvarez et al., 2007). So, we wanted to determinewhether HIV-2 virus similarly induced COX-2 synthesis in astroglialcells. To test whether HIV-2CBL23 isolate was able to infect humanastrocytes, first of all we confirmed the presence of different cellularreceptors on U-87 cells by RT-PCR. As expected, CXCR4-mRNA but notCD4-mRNA was detected in these cells (Fig. 1A). Those results wereconfirmed at the cell surface by flow cytometry (Fig. 1A). To determinewhether HIV-2 virus could infect human astrocytes, we cultured thosecells in the presence of HIV-2CBL23 isolate at different MOI and after 3days, viral production was measured by ELISA. Although U-87 cellswere unable to produce new virus (Fig. 1B), HIV-2 induced COX-2mRNA expression analyzed by real-time RT-PCR, in a dose-dependentmanner (Fig. 1C), suggesting that COX-2 induction was mediatedmerely by binding of the envelope glycoprotein to CXCR4 and not byviral infection per se. This result was confirmed by using monoclonalantibodies against CXCR4 as well as the CXCR4 antagonist, AMD3100and measuring COX-2mRNA induction by HIV-2. In both cases, wefound a decrease in COX-2mRNA induction (Fig. 1C). Moreover, HIV-2CBL23was able to induce COX-2 protein expression inU-87 cells aswellas in human primary astrocytes, analyzed by Western blot. AlthoughCOX-2 protein was already detectable in unstimulated cells (Fig. 1D),after culture with HIV-2, a heavily increase in protein levels at 6 h wasobserved, which subsequently decreased after 24 and 48 h of culture.Accordingly, we found PGE2 production increased over basal levelsafter 16 h of treatment so much at U-87 cells as in primary astrocytes(Fig. 1E). The selective inhibitor of COX-2, NS-398, totally abrogatedPGE2 production in U-87 cells confirming the main role of COX-2isoform on PGE2 synthesis.

HIV-2 induces COX-2 promoter activity in U-87 cells

The above results suggested that HIV-2 interaction with glial cellswas able to induce COX-2 at the transcriptional level. To furtheraddress this, we transfected glial cells with the COX-2 promotercontaining a region spanning from −1796 to +104 bp relative to thetranscription start site of the human COX-2 gene in the luciferasereporter plasmid pXP2 (P2-1900). The transfected cells were culturedin the presence of HIV-2CBL23 (MOI 1), and 16 h later, luciferase activitywas measured. P2-1900 COX-2 promoter activity was up-regulated(more than 2 fold) in the presence of HIV-2 (Fig. 2A). To investigatewhether CXCR4 receptor was required for this induction, U-87transfected cells were pretreated with AMD3100 (50 μM) during30min at 37 °C, before HIV-2 addition to the cells for 16 h. As shown inFig. 2A, the HIV-2-induced COX-2 transcriptional activity wasabsolutely abrogated in the presence of AMD3100, indicating againthat HIV-2-dependent COX-2 induction occurred through its interac-tion with CXCR4 receptor.

To gain some insight into the pathways involved in HIV-2CBL23induction of COX-2 transcription, we first determined the regionwithin the COX-2 promoterwhich responded toHIV-2 byusing a seriesof deletion mutants derived from the COX-2 5′upstream regulatorysequence through transfection assays. The P2-431 plasmid has a dele-tion of a distal NF-κB site, whereas the P2-274 presents a deletion ofboth NF-κB sites in the COX-2 promoter, keeping intact NFAT, AP-1 orCRE sites (Iniguez et al., 2000). As shown in Fig. 2B, deletion up to −327(P2-431) strongly abrogated HIV-2 inducibility, and further deletionsup to−170 (P2-274) increased this effect. Those results suggested that 2regions between −1786 and −327 were necessary for COX-2 induction.

NF-κB is required for COX-2 induction by HIV-2 in human astrocytes

Although the above results indicated that NF-κB activation wasrequired for HIV-2 induction of COX-2 in U-87 cells, we wanted todiscardwhether HIV-2 induced NFAT, or AP-1 activity.We observed noincrease in the transcriptional activity of reporter genes under controlof AP-1 or NFAT sites in U-87 cells upon culture with HIV-2 (Fig. 3A)discarding any contribution of any of these factors in COX-2 inductionin glial cells by HIV-2CBL23. In contrast, as shown in Fig. 3A, HIV-2CBL23caused a marked increase of around 3–4 fold on average of a NF-κB-dependent luciferase reporter plasmid in HIV-2-U-87 cells comparedto control cells.

To further investigate this activity, U-87 cells were transiently co-transfected with the active NF-κB member p65/relA or the inhibitorysubunit IκBα and P2-1900 COX-2-luc reporter plasmid. Overexpres-sion of p65/relA was able, to increase transcriptional activity of COX-2promoter in those cells (Fig. 3B) (an average of 4.1 fold) over controlcells transfected with the empty vector. Besides, p65/relA over-expression synergistically cooperated with HIV-2 in inducing COX-2promoter activity (around 16 fold). Interestingly, transfection of IκBαcompletely abrogated HIV-2-induced COX-2 promoter activity. Takentogether the above results suggest that NF-κB is themain transcriptionfactor induced by HIV-2 involved in COX-2 transcription in U-87 cells.To check this fact in primary cultures, we pretreatedNHA cellswith theagent PDTC before contact with HIV-2 and after 16 h of culture, PGE2productionwasmeasured in the supernatants. As you can see in Fig. 3C,it is totally clear that NF-κB is really involved in PGE2 production notonly in established cell lines but also in primary astrocytes.

Activation of NF-κB by HIV-2CBL23 is associated with increases in thetransactivation function of p65/relA

The activation of NF-κB usually involves the degradation of IκBαsubunit bound to NF-κB dimer allowing its translocation to thenucleus where it binds and activates NF-κB-dependent genes. Thus, toinvestigate the mechanisms by which HIV-2 induces NF-κB activation,levels of NF-κB/p65/relA and IκBα were studied by Western blot. Asshown in Fig. 4A, p65/relA was present in nuclear extracts of controlU-87 cells. This fact was not due to cytoplasmic contamination in thenuclear fraction (data not shown). More surprisingly, upon HIV-2interaction there was no change neither in cytoplasmic nor in nuclearlevels of p65/relA, as it would be expected if an increase in p65/relAtranslocation had taken place. At the same time similar total levels ofIκBα were observed at any time tested after HIV-2 culture, indicatingthat IκBα degradation is not taking place upon HIV-2 contact in thesecells.

All data indicated that HIV-2 was inducing the NF-κB activitywithout altering p65/relA nuclear translocation or IκBα degradation.However, the second mechanism to regulate NF-κB activity, withoutIκBα degradation, involving Ser536 and/or Ser529 phosphorylation inthe transactivation domain of p65/relA has been described (Schmitz,Bacher, and Kracht, 2001). This pathway involves the phosphorylationof p65/relA or c-rel in several sites of their transactivation domains,leading to an increase in the transactivation activity. To confirm this

Page 3: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

146 S. Álvarez et al. / Virology 380 (2008) 144–151

possibility, we used the Gal4 transcription system, which allows thestudy of transcription factor activities independently of nucleartranslocation. U-87 cells were transfected with a reporter construct

in which luciferase expression is driven by four tandem copies of theresponsive element for the yeast transcription factor Gal4 (pGal4-Luc)along with expression vectors containing fusion of the DNA binding

Page 4: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

Fig. 2. Analysis of COX-2 transcriptional activity induced by HIV-2CBL23. (A) HIV-2 induces COX-2 promoter activity. U-87 cells were transfected with COX-2 promoter construct andcultured in the presence of HIV-2CBL23 (MOI 1) alone or in combinationwith AMD3100. (B) U-87 cells were transfected with the indicated COX-2 promoter constructs and cultured inthe presence of HIV-2CBL23 isolate for 16 h and assayed for luciferase activity. Assays were performed in triplicate and are expressed as fold induction about basal values of untreatedcells. Differences from HIV-2CBL23 induction of full promoter (P2-1900): ⁎p= 0.02; –, non-significant.

147S. Álvarez et al. / Virology 380 (2008) 144–151

domain of Gal4 linked to the transcription activation domain of p65/relA, either in the wild type version or in a form mutated in Ser529and Ser536. HIV-2 increased the transactivation a Gal4-luc reportermediated by Gal4-p65 in U-87 cells, by around 2 fold on average, butnot by the Ser529 and Ser536 mutant Gal4-p65 form (Fig. 4B). Theabove results indicated that phosphorylation of p65/relA at Ser536,which is a key modification that potentiates p65/relA transactivationfunction, hence activation (Schmitz, Bacher, and Kracht, 2001;Sizemore, Leung, and Stark, 1999; Viatour et al., 2005), was involvedin HIV-2 induction of NF-κB. Thus, we tested whether HIV-2 affectedphosphorylation of endogenous p65/relA at Ser536 through Westernblot. As shown in Fig. 4C, culture of U-87 cells in the presence of HIV-2CBL23 led to a significantly level of p65/relA phosphorylation at theSer536 position higher than in control cells.

HIV-2 increases GFAP expression

It is well established that GFAP is increased during activation ofastrocytes and astrogliosis. To determine the effect of HIV-2 on GFAPexpression, we cultured U-87 cells in contact with HIV-2CBL23 and after16 h we examined its expression by immunofluorescence assay. The

Fig.1. COX-2 expression is induced in human astrocytes after culturing with HIV-2. (A) Expresamplified with CD4, and CXCR4-specific primers. (A2) Flow cytometry analysis of CD4 and CXU-87 cells were infected at MOI 1, and Agp24 was quantified in the supernatants of cultures 3(C) COX-2 mRNA induction detected by real-time PCR. Cells were cultured with HIV-2CBL23 (Mwere collected. RT-PCR amplification was performed using 200 ng of total RNA using Lighdetected by Western blot analysis. (E) PGE2 release in the supernatant of those cells after 16from HIV-2CBL23-production of PGE2 in human astrocytes: ⁎pb0.01; ⁎⁎p=0.02.

results showed a significantly higher level of GFAP expression instimulated cells compared to control (Fig. 5).

To investigate whether COX-2 induction was involved in thisactivation, U-87 cells were cultured with HIV-2CBL2 in the presence ofseveral COX-2 inhibitors. NS-398 and ibuprofen were able to decreaseGFAP expression in HIV-2-U-87 cells suggesting a role of COX-2protein in astroglial activation (Fig. 5). We discarded that the decreasein GFAP expression was due to toxicity of these inhibitors on cellcultures since any of them did not produce cell death measured byMTT assay (data not shown).

Discussion

There are very few studies concerning HIV-2 and even less aboutits neuropathogenicity, nonetheless the fact is that HIV-2 wouldcertainly provide a good model for HIV-1 related neurotoxicity.Although the incidence of HAD has markedly decreased since it hasbecome possible to effectively control viral replication by administer-ing HAART, a less severe form of HAD, comprising a milder cognitiveand motor disorder, is now potentially a serious problem (Gonzalez-Scarano and Martin-Garcia, 2005).

sion of CD4 and CXCR4 in U-87 cells. (A1) Purified total RNAwas reverse-transcribed andCR4 expression in U-87 and MT-2 cells. (B) Infection of U-87 cells by HIV-2CBL23 isolate.after infection. HIV-1NL4.3 infection (MOI 1) was the positive control of the experiment.OI 1 or 2) in the presence or absence of anti-CXCR4 or AMD3100 during 16 h and then

t-Cycler RT-PCR kit. (D) COX-2 protein expression in HIV-2-U-87 and HIV-2-NHA cellsh of culture. Data are representative of at least 3 independent experiments. Differences

Page 5: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

Fig. 3. Role of NFAT, AP-1, and NF-κB transcription factors in COX-2 transcriptional activity induced by HIV-2. (A) U-87 cells were transfected with the −73col-luc reporter plasmid(AP-1-dependent), the NFAT-luciferase reporter plasmid, or with the κB-luciferase reporter plasmid and cultured in the presence of the isolate HIV-2CBL23 (MOI 1) during 16 h. (B) U-87cells were cotransfected with the P2-1900 COX-2 promoter together with an expression vectors for the p65/relA, and IκBα subunits or an empty vector. U-87 cells were cultured inpresence or absence (control) of HIV-2CBL23 isolate (MOI 1) during 16 h and assayed for luciferase activity. Data shown are expressed as fold induction about basal values of untreatedcells. (C) NHA cells were treated with HIV-2CBL23 isolate alone or in combination with the NF-κB inhibitor PDTC (10 μM). PGE2 release was measured in the supernatants after 16 h ofculture. Differences fromHIV-2CBL23-induction of COX-2 promoter (P2-1900) in empty vector-transfected cells: ⁎p=0.01. Differences fromHIV-2CBL23-production of PGE2 in NHA cells:⁎⁎pb0.01.

148 S. Álvarez et al. / Virology 380 (2008) 144–151

On the other hand, COX-2 has been involved in the pathology ofseveral neurodegenerative diseases as well as in the cell damageassociated with neuroinflammation. Thus, elevated expression ofCOX-2 mRNA and protein has been reported in Alzheimer's disease(Pasinetti, 1998), and recently in AIDS infection (Alvarez et al., 2005;Flora et al., 2006).

Since HIV-2CBL23 does not infect U-87 cells, the whole resultspresented here show that COX-2 is induced in human astroglial cells

Fig. 4. HIV-2 induces p65/relA transactivation but not IκBα degradation. (A) HIV-2 does not2CBL23 (MOI 1) for the indicated time points (0–120 min). The cell lysates were blotted withindependent experiments. (B) HIV-2 induces activation p65/relA transactivation. Cells wer(Ser529/536 to Ala) constructs and 100 ng of 5XGal4 luciferase reporter and cultured withexpressed as fold induction above control. (C) HIV-2CBL23 induces p65/relA phosphorylationtimes. Whole cell lysates were prepared and blotted with p65/relA phospho-specific antibo

by HIV-2 virus, being dependent on the presence of the viral core-ceptor CXCR4. In addition, the transcriptional regulation of COX-2 byHIV-2 involves NF-κB activation. Thus, we found that deletion of thedistal NF-κB element in the COX-2 promoter did reduce stronglyHIV-2-induced COX-2 promoter activity. Further deletion of a pro-ximal NF-κB site completely abrogated this induction, indicatingthat both NF-κB sites are responsible for COX-2 induction after HIV-2 stimulation. The main role of NF-κB on COX-2 induction by HIV-2

induce p65/relA translocation or IκBα degradation. U-87 cells were cultured with HIV-antibodies specific for the p65/relA and IκBα. Western blot data are representative of 3e cotransfected with 10 ng of the Gal4DBD-p65 wild type and Gal4DBD-p65 mutantmedium in basal condition or cultured with HIV-2CBL23 (MOI 1). Reporter activity isin Ser536. U-87 cells were cultured with HIV-2CBL23 isolate (MOI 1) for the indicated

dy that detects phosphorylation at Ser536 position (indicated as P-p65 (Ser-536)).

Page 6: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

Fig. 5. HIV-2 increases GFAP expression in U-87 cells. U-87 cells were cultured in the presence or absence of HIV-2CBL23 alone or in combination with the COX inhibitors, NS-398(5 μM) and ibuprofen (10 μM). After 16 h, cells on coverslips were fixed and stained with antibodies against the cell specific marker, GFAP. Original magnification ×40. Backgroundstaining was observed using irrelevant antibodies.

149S. Álvarez et al. / Virology 380 (2008) 144–151

was further supported by the fact that p65/relA overexpression wasenough to induce COX-2 promoter activity and synergized with HIV-2 in this induction. More importantly, cotransfection experimentswith a plasmid encoding for the NF-κB subunit inhibitory IκBα plusthe COX-2 promoter abrogated COX-2 transcriptional activity byHIV-2. All our data indicate that NF-κB activation is required andsufficient for the upregulation of glial COX-2 expression by HIV-2.Interestingly, HIV-2 induces NF-κB activation by a mechanism notinvolving IκBα degradation or p65/relA translocation. Regulation ofthe NF-κB pathway is also brought about through multiple post-translational modifications that control the activity of the corecomponents of NF-κB signalling: the IKK complex, the IκB proteinsand the NF-κB subunits themselves. These regulatory modificationscan vary depending on the nature of the NF-κB-inducing stimulus.Phosphorylation and degradation of IκB and the consequentliberation of NF-κB do not seem to be sufficient to activate NF-κB-dependent transcription in some instances (Ghosh and Karin, 2002;Viatour et al., 2005). NF-κB transactivation in the absence ofincreased NF-κB nuclear translocation or induced IκBα degradationwas also reported in several cell types (Doerre and Corley, 1999;Ghosh and Karin, 2002). Mutation of Ser529 and Ser536 eliminatedHIV-2-induced Gal-4 transactivation. This result fits nicely with thefact that HIV-2 induces Ser536 phosphorylation and suggests thatHIV-2 induces NF-κB activation through increases in the transacti-vation potential of p65/rel due to phosphorylation in Ser536. Theinhibitory effect of IκBα overexpression on COX-2 induction by HIV-2 may simply reflect that it sequesters nuclear p65/relA preventingp65 phosphorylation. However, future work is needed to addressthe exact mechanism by which HIV-2 achieves this effect on NF-κBtransactivation.

Reactive astrocytosis or gliosis, defined mainly as an increase insize and number of GFAP-positive cells, is a prominent feature of HIV-1 infected persons (Sharer, 1992). We could demonstrate for the firsttime that HIV-2 modulates the state of astrocytic cells by upregulatingGFAP expression which is a marker for activated astrocytes in similarway than HIV-1 does (Kohleisen et al., 1999). Interestingly, this effectof HIV-2 could be blocked by COX-2 inhibitors. Along with theseresults, previous data described that P2Y receptor-mediated reactiveastrogliosis occurs via induction of COX-2 derived PGE2 (Brambilla etal., 1999).

In summary, we demonstrate here that HIV-2 efficiently inducesCOX-2 transcription in human astrocytes through a novel pathwaydepending on NF-κB response elements. To our knowledge thisrepresents the first example of the existence of a new HIV-2 viralmechanism of COX-2 induction in brain cells through regulation of NF-κB p65/relA phosphorylation and transactivation. We also demon-strate that, as a consequence of COX-2 induction, astrocytic activationis increased, supporting an undescribed role of astrocytes in HIV-2-associated neuropathogenesis. Further studies are required to effec-tively evaluate the effects of COX-2 inhibitors in the pathogenesis ofthe HIV-2 infection.

Materials and methods

Cell culture and reagents

Normal human astrocytes (NHA) isolated from the cerebrum of 5-month-old human fetuses were purchased from Cambrex (CC-2565,NHA-Normal Human Astrocytes; Walkersville, MD, USA). NHA cellswere cultured by plating the cells at a density of 15,000 cells ⁄cm2 inexpansion media (AGM Bullet Kit media, Cambrex) according to themanufacturer's protocol. The cells were incubated at 37 °C with 5%humidified CO2 and allowed to reach about 95% confluency. After this,they were harvested using 0.25% trypsin and 1 mm EDTA for 5 min at37 °C. The astrocytoma human cell line U-87, was routinely grown inDMEM (Gibco) containing 10% heat-inactivated fetal calf serum, 1%penicillin/streptomycin, and 2 mM L-glutamine (ICN Pharmaceuticals,CostaMesa, CA,USA) at 37 °C in ahumidified atmosphere of 5%CO2.MT-2 cells (T cell leukemia) were routinely grown in RPMI 1640 (BiochromKG Seromed), containing 10% heat-inactivated fetal calf serum, 1%penicillin/streptomycin, and 2 mM L-glutamine (ICN Pharmaceuticals,Costa Mesa, CA, USA) at 37 °C in a humidified atmosphere of 5% CO2.

The HIV-2CBL23 viral isolate (generously donated by Dr. BalbinoAlarcón, Centro de Biologia Molecular, Madrid, Spain), and a Tlymphocyte-adapted strain, HIV-1NL4.3 (X4) were used for infectivityassays. Virus stocks were prepared by expanding viral isolates in PBLand were titrated using the end point dilution method. The virusstocks gave 104 to 105 TCID50 when PBL were inoculated on micro-dilution plates and read 10 days after with Agp24 assay (INNOTESTHIV antigen mAb, Innogenetics N.V., Zwijndrecht, Belgium) accordingmanufacturer's instructions. The sensitivity of the assay was 10 pg/ml.To assess the purity of the virus stocks thawed aliquots were filteredthrough 0.22-μm filters before use.

AMD3100 was from NIH (AIDS Research and Reference ReagentProgram, Rockville, USA), and anti-CXCR4 monoclonal antibody waspurchased from R&D Systems (Europe, Abingdon, UK). Ibuprofen, andLPS (lipopolysaccharide) were purchased from Sigma (St. Louis, MO).The selective inhibitor of COX-2, NS-398, was from Cayman ChemicalCompany (MI, USA). The agent PDTC was obtained from Santa CruzBiotechnology, Inc. Anti-human CD4-phycoeritrine (PE), and anti-human CXCR4 antibodies were from Beckman Coulter (Marseille,France).

HIV-2 infection of U-87 cells

U-87 cells were exposed to HIV-2CBL23 isolate, at different multi-plicity of infection (MOI) for 2 h at 37 °C. At the end of this period, theculture medium was removed, cells were extensively washed withPBS, and complete medium was added to each well. Virus titers wereevaluated in the last washing buffer (time 0). Cell supernatants wereharvested 3 days post-infection to monitor p24 viral core antigenproduction by ELISA. HIV-1NL4.3 infection was used as a positive con-trol of the experiment.

Page 7: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

150 S. Álvarez et al. / Virology 380 (2008) 144–151

Plasmid constructs

Human COX-2 promoter constructs were generated as previouslydescribed (Iniguez et al., 2000). The pNF-κB-luc reporter plasmid con-tains a trimer of the NF-κB-bindingmotif of the H-2k gene upstream ofthe TK (thymidine kinase) minimal promoter and the luciferasereporter gene. The pNFAT-luc reporter plasmid was a gift from Dr. G.Crabtree (Department of Pathology, Howard Hughes Medical Institute,Stanford University Medical School, Stanford, CA 94305, USA). Itcontains three tandem copies of the distal NFAT site of the human IL-2promoter fused to the minimal human IL-2 promoter. The p-73col-lucreporter plasmid contains AP-1-binding region (-73/+63pb) of humancollagenase promoter. The p65/relA, and IκBα expression plasmidswere also generously provided by Dr. G. Crabtree. The reporter Gal-4Luc contains five tandem repeats of the Gal4 element upstream theluciferase reporter gene (Martin and Fresno, 2000). Thewild typeGal4-p65 ΔN plasmid active in binding to Gal4-binding sites as well as intransactivation, containing amino acids 386-551 of p65/relA and theGal4 Ser529 and Ser536 to Ala mutant were a generous gift of Dr. L.Schmitz (Schmitz and Baeuerle, 1991).

CD4 and CXCR4 detection

The expression of CD4 and CXCR4 was evaluated by reverse trans-criptase-polymerase chain reaction (RT-PCR). The mRNA from 105

cells was isolated using SV Total RNA Isolation System (PromegaCorporation, Madison), according to the manufacturer's instructionsand cDNA synthesis with AccessQuick RT-PCR System (PromegaCorporation, Madison). The polymerase chain reaction was carriedout in an automatic Thermal Cycler (Perkin-Elmer GeneAmp PCRsystem 9600). Amplified cDNAswere separated by agarose gel electro-phoresis and bands visualized by ethidium bromide staining.

Flow cytometry

The expression of CD4 and CXCR4 cell surface antigens wasdetermined by flow cytometry by using a FAC Sort (Epics XL-MCLBeckman Coulter).

COX-2 mRNA quantification by real-time PCR

Real-time PCR using 200 ng of total RNA was performed using aRoche Light-Cycler PCR instrument and Light-Cycler RT-PCR kit (RochePerkin-Elmer, Foster City, California, USA). The standard used to makethe control curve was the commercial plasmid pCRTM II (InvitrogenCorporation, Life Technologies, Rockville, MD, USA) where a fragmentof the COX-2 promoter was inserted. The number of molecules of theplasmid was quantified measuring the quantity of DNA in a spectrum-photometer. As the weight of the standard (plasmid+sequence ofCOX-2) was known the exact number ofmolecules can be extrapolated.

PGE2 determination

Supernatants of U-87 andNHA cells after different treatmentsweretested for PGE2 production using a competitive enzyme immunoassayusing the PGE2 EIA kit (Cayman Chemical Company, MI, USA).

Transfection and luciferase assays

Transcriptional activity was measured using luciferase reportergene assays in transiently transfectedU-87 cells. Cellswere transfectedby Lipofectin reagent as recommended by the manufacturer (LifeTechnologies, Rockville, MD, USA). Briefly, exponential growing cells(1.5×106) were incubated for 8 h at 37 °Cwith amixture of 1.5 μg/ml ofthe corresponding reporter plasmid and 3 μg/ml lipofectin in 1 ml ofOpti-MEM Reduced Serum Medium modification of MEM (Eagle's)

(GIBCO-BRL, Invitrogen Corporation). In cotransfection experiments,0.15–0.5 μg/ml of the correspondent expression plasmid was included.The total amount of DNA in each transfection was kept constant byusing the corresponding empty expression vectors. Cells were thenresuspended in completemediumand incubated at 37 °C for additional16 h. Transfected cells were exposed to different stimuli as indicated.Then, cells were harvested and lysed. Luciferase activity was deter-mined by using a luciferase assay kit (Promega Corporation, Madison,USA) with a luminometer 1450 Microbeta Luminiscence Counter(Walax, Trilux). Protein contents were measured using the bicincho-ninic acid method (BCA protein assay kit from Pierce, Rockford, USA)according to the manufacturer's instructions. The data presented areexpressed as fold induction respect untreated cells of at least 3 inde-pendent experiments.

Western blot analysis

Human primary astrocytes and U-87 cells were exposed to HIV-2CBL23 isolate for different times. Protein contents were measuredusing BCA protein assay according to the manufacturer's instructions.ForWestern blotting, 20 μg of protein from each sample was subjectedto SDS-polyacrylamide gel electrophoresis (PAGE) on a 7.5% gel.Proteins were then transferred onto a PVDF fluoride membrane(Millipore, Bedford, MA, USA) by humidified transference blotting.The membrane was blocked overnight at 4 °C using Rotiblock (Roth,Karlsruhe) and for another hour at room temperature before incuba-tionwith the primaryantibodies: amouse anti-humanCOX-2 (CaymanChemical Company, MI, USA), a mouse anti-human p65/relA NF-κB(Chemicon International, USA), a mouse anti-IκBα subunit, a rabbitanti-Ser 536 phosphorylated p65/relA (Santa Cruz Biotechnology, Inc)or a mouse anti-humanα-tubulin (Sigma, St. Louis, MO). Visualizationof protein bands was performed using enhanced chemiluminescence(ECL) reagents (Amersham Biosciences, UK). All Western blot experi-ments were carried out at least three times. In some cases as loadingcontrol, blots were probed for tubulin using a specific antibody.

Indirect immunofluorescence assays

U-87 cells were pretreated or not with different COX-2 inhibitorsfor 2 h beforeHIV-2CBL23 culture during 16 h, and left to adhere to slidesduring 2 h at 37 °C. Monolayers were fixed for 20 min in 4% para-formaldehyde. Cultures were incubated for 1 h with primarymonoclonal antibody anti-human GFAP 1:100 (Chemicon Interna-tional, MI, USA). Following washing with PBS, the monolayers wereincubated in the dark for half an hour with goat anti-human fluo-rescein isothiocyanate (FITC) conjugated (Jackson ImmunoResearch,Baltimore, USA). Following a final PBS wash, the slides were mountedwith DAKO® Ultramount, Aqueous Permanent Mounting Medium(DAKO Corporation, CA, USA).

Statistical analysis

The data were expressed as the mean±S.E. from three to six inde-pendent experiments. Differences were analyzed using non-para-metric tests (Mann–Whitney “U”).

Acknowledgments

This work was supported by grants from Fondos de InvestigaciónSanitaria (FIS PI061479) Red Temática de Investigación CooperativaSanitaria ISCIII (RETIC RD06/0006/0035), Fundación para la Investiga-ción y Prevención del SIDA en España, FIPSE (36514/05, 36536/05) andFundación Caja Navarra to MAMF. From RED RICET (RD06/0021),Programa Nacional de Salud of Spain (SAF2005-02220), Laboratoriosdel Dr. ESTEVE, the 6th EU Framework Programme European Com-mission (Integrated project EICOSANOX, LSH-CT-2004-005033 and

Page 8: HIV-2 induces NF-κB activation and cyclooxygenase-2 expression in human astroglial cells

151S. Álvarez et al. / Virology 380 (2008) 144–151

MAIN network of excellence) and the Fundación Ramón Areces toMF. From Fondo de Investigación Sanitaria (PI040883) and Comu-nidad Autónoma de Madrid (SAL/2001/2004) to both MAMF andMF. Susana Álvarez is supported by a fellowship of FIS (CD06/00321) and Almudena Blanco is supported by a fellowship of RETICRD06/0006/0035).

References

Allport, V.C., Slater, D.M., Newton, R., Bennett, P.R., 2000. NF-kappaB and AP-1 arerequired for cyclo-oxygenase 2 gene expression in amnion epithelial cell line(WISH). Mol. Hum. Reprod. 6 (6), 561–565.

Alvarez, S., Serramia, M.J., Fresno, M., Munoz-Fernandez, M., 2005. Human immuno-deficiency virus type 1 envelope glycoprotein 120 induces cyclooxygenase-2expression in neuroblastoma cells through a nuclear factor-kappaB and activatingprotein-1 mediated mechanism. J. Neurochem. 94 (3), 850–861.

Alvarez, S., Serramia, M.J., Fresno, M., Munoz-Fernandez, M.A., 2007. HIV-1 envelopeglycoprotein 120 induces cyclooxygenase-2 expression in astrocytoma cellsthrough a nuclear factor-kappaB-dependent mechanism. Neuromol. Med. 9 (2),179–193.

Anrather, J., Csizmadia, V., Soares, M.P., Winkler, H., 1999. Regulation of NF-kappaB RelAphosphorylation and transcriptional activity by p21(ras) and protein kinase Czeta inprimary endothelial cells. J. Biol. Chem. 274 (19), 13594–13603.

Brambilla, R., Burnstock, G., Bonazzi, A., Ceruti, S., Cattabeni, F., Abbracchio, M.P., 1999.Cyclo-oxygenase-2 mediates P2Y receptor-induced reactive astrogliosis. Br. J.Pharmacol. 126 (3), 563–567.

Clapham, P.R., McKnight, A., 2002. Cell surface receptors, virus entry and tropism ofprimate lentiviruses. J. Gen. Virol. 83 (Pt 8), 1809–1829.

Deng, W.G., Zhu, Y., Wu, K.K., 2003. Up-regulation of p300 binding and p50acetylation in tumor necrosis factor-alpha-induced cyclooxygenase-2 promoteractivation. J. Biol. Chem. 278 (7), 4770–4777.

Doerre, S., Corley, R.B., 1999. Constitutive nuclear translocation of NF-kappa B in B cellsin the absence of I kappa B degradation. J. Immunol. 163 (1), 269–277.

Endres, M.J., Clapham, P.R., Marsh, M., Ahuja, M., Turner, J.D., McKnight, A., Thomas, J.F.,Stoebenau-Haggarty, B., Choe, S., Vance, P.J., Wells, T.N., Power, C.A., Sutterwala, S.S.,Doms, R.W., Landau, N.R., Hoxie, J.A., 1996. CD4-independent infection by HIV-2 ismediated by fusin/CXCR4. Cell 87 (4), 745–756.

Eng, L.F., Ghirnikar, R.S., 1994. GFAP and astrogliosis. Brain Pathol. 4 (3), 229–237.Flora, G., Pu, H., Hennig, B., Toborek, M., 2006. Cyclooxygenase-2 is involved in HIV-1

Tat-induced inflammatory responses in the brain. Neuromol. Med. 8 (3), 337–352.

Ghosh, S., Karin, M., 2002. Missing pieces in the NF-kappaB puzzle. Cell 109 (Suppl),S81–S96.

Gonzalez-Scarano, F., Martin-Garcia, J., 2005. The neuropathogenesis of AIDS. Nat. Rev.,Immunol. 5 (1), 69–81.

Iniguez, M.A., Martinez-Martinez, S., Punzon, C., Redondo, J.M., Fresno, M., 2000. Anessential role of the nuclear factor of activated T cells in the regulation of theexpression of the cyclooxygenase-2 gene in human T lymphocytes. J. Biol. Chem.275 (31), 23627–23635.

Kohleisen, B., Shumay, E., Sutter, G., Foerster, R., Brack-Werner, R., Nuesse, M., Erfle, V.,1999. Stable expression of HIV-1 Nef induces changes in growth properties andactivation state of human astrocytes. Aids 13 (17), 2331–2341.

Lucas, S.B., Hounnou, A., Peacock, C., Beaumel, A., Djomand, G., N'Gbichi, J.M., Yeboue, K.,Honde, M., Diomande, M., Giordano, C., et al., 1993. The mortality and pathology ofHIV infection in a west African city. Aids 7 (12), 1569–1579.

Martin, A.G., Fresno, M., 2000. Tumor necrosis factor-alpha activation of NF-kappa Brequires the phosphorylation of Ser-471 in the transactivation domain of c-Rel.J. Biol. Chem. 275 (32), 24383–24391.

Pasinetti, G.M., 1998. Cyclooxygenase and inflammation in Alzheimer's disease:experimental approaches and clinical interventions. J. Neurosci. Res. 54 (1),1–6.

Reeves, J.D., Doms, R.W., 2002. Human immunodeficiency virus type 2. J. Gen. Virol. 83(Pt 6), 1253–1265.

Schmitz, M.L., Bacher, S., Kracht, M., 2001. I kappa B-independent control of NF-kappa Bactivity by modulatory phosphorylations. Trends Biochem. Sci. 26 (3), 186–190.

Schmitz, M.L., Baeuerle, P.A., 1991. The p65 subunit is responsible for the strongtranscription activating potential of NF-kappa B. Embo J. 10 (12), 3805–3817.

Sharer, L.R., 1992. Pathology of HIV-1 infection of the central nervous system. A review.J. Neuropathol. Exp. Neurol. 51 (1), 3–11.

Sizemore, N., Leung, S., Stark, G.R., 1999. Activation of phosphatidylinositol 3-kinase inresponse to interleukin-1 leads to phosphorylation and activation of the NF-kappaBp65/RelA subunit. Mol. Cell. Biol. 19 (7), 4798–4805.

Smith, W.L., DeWitt, D.L., Garavito, R.M., 2000. Cyclooxygenases: structural, cellular, andmolecular biology. Annu. Rev. Biochem. 69, 145–182.

Vane, J.R., Bakhle, Y.S., Botting, R.M., 1998. Cyclooxygenases 1 and 2. Annu. Rev.Pharmacol. Toxicol. 38, 97–120.

Viatour, P., Merville, M.P., Bours, V., Chariot, A., 2005. Phosphorylation of NF-kappaB andIkappaB proteins: implications in cancer and inflammation. Trends Biochem. Sci. 30(1), 43–52.

Willey, S.J., Reeves, J.D., Hudson, R., Miyake, K., Dejucq, N., Schols, D., De Clercq, E., Bell, J.,McKnight, A., Clapham, P.R., 2003. Identification of a subset of human immuno-deficiency virus type 1 (HIV-1), HIV-2, and simian immunodeficiency virus strainsable to exploit an alternative coreceptor on untransformed human brain andlymphoid cells. J. Virol. 77 (11), 6138–6152.