Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
FASEB J ; 21(10): 2323-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17431094

ABSTRACT

Dendritic cells (DCs) and natural killer (NK) cells are essential components of the innate immunity and play a critical role in the first phase of host defense against infection. Interactions between DCs and NK cells have been demonstrated in a variety of settings, with evidence emerging of complex bidirectional crosstalk between the two cell types. The accessory HIV-1 Nef protein is a crucial determinant for viral replication and pathogenesis. We previously demonstrated that Nef, hijacking DC functional activity, subverts the DC arm of immune response to escape the adaptive immune attack. Here, we monitor the effect of Nef on the outcome of the innate immune response, focusing on the impact of Nef on DC/NK crosstalk. We demonstrate that Nef up-regulates the ability of DCs to stimulate the immunoregulatory NK cells (CD56(bright)) as assessed by the activated phenotype, up-regulation of their proliferative response and INF-gamma release. On the other hand, Nef-pulsed DCs inhibit cytotoxic NK cells (CD56(dim)), as assessed by the reduced HLA-DR surface expression, reduced proliferation and cytotoxic activity. Moreover, in the presence of Nef-pulsed DCs, we found a significant up-regulation of TNF-alpha secretion and a significant reduction of IL-10, GM-CSF, MIP-1alpha and RANTES secretion. Our findings suggest that the Nef-induced dysregulation in the DC/NK cell crosstalk may represent a potential mechanism through which HIV escapes innate immune surveillance.


Subject(s)
CD56 Antigen/physiology , Dendritic Cells/immunology , Gene Products, nef/pharmacology , Killer Cells, Natural/immunology , Acquired Immunodeficiency Syndrome/immunology , CD56 Antigen/classification , CD56 Antigen/drug effects , Cell Division , Dendritic Cells/drug effects , Dendritic Cells/virology , Flow Cytometry , Gene Products, nef/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-10/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Recombinant Proteins/pharmacology , Virus Replication/physiology , nef Gene Products, Human Immunodeficiency Virus
2.
J Gen Virol ; 88(Pt 1): 242-250, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17170457

ABSTRACT

Natural killer (NK) cells are a major component of the host innate immune defence against various pathogens. Several viruses, including Human immunodeficiency virus 1 (HIV-1), have developed strategies to evade the NK-cell response. This study was designed to evaluate whether HIV-1 could interfere with the expression of NK cell-activating ligands, specifically the human leukocyte antigen (HLA)-I-like MICA and ULBP molecules that bind NKG2D, an activating receptor expressed by all NK cells. Results show that the HIV-1 Nef protein downmodulates cell-surface expression of MICA, ULBP1 and ULBP2, with a stronger effect on the latter molecule. The activity on MICA and ULBP2 is well conserved in Nef protein variants derived from HIV-1-infected patients. In HIV-1-infected cells, cell-surface expression of NKG2D ligands increased to a higher extent with a Nef-deficient virus compared with wild-type virus. Mutational analysis of Nef showed that NKG2D ligand downmodulation has structural requirements that differ from those of other reported Nef activities, including HLA-I downmodulation. Finally, data demonstrate that Nef expression has functional consequences on NK-cell recognition, causing a decreased susceptibility to NK cell-mediated lysis. These findings provide a novel insight into the mechanisms evolved by HIV-1 to escape from the NK-cell response.


Subject(s)
Gene Products, nef/pharmacology , HIV-1/chemistry , Killer Cells, Natural/immunology , Receptors, Immunologic/metabolism , Cell Line , Cytotoxicity, Immunologic , Down-Regulation/drug effects , Down-Regulation/immunology , HIV-1/immunology , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Natural Killer Cell , nef Gene Products, Human Immunodeficiency Virus
3.
J Virol ; 81(6): 2777-91, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182689

ABSTRACT

The viral protein Nef is a virulence factor that plays multiple roles during the early and late phases of human immunodeficiency virus (HIV) replication. Nef regulates the cell surface expression of critical proteins (including down-regulation of CD4 and major histocompatibility complex class I), T-cell receptor signaling, and apoptosis, inducing proapoptotic effects in uninfected bystander cells and antiapoptotic effects in infected cells. It has been proposed that Nef intersects the CD40 ligand signaling pathway in macrophages, leading to modification in the pattern of secreted factors that appear able to recruit and activate T lymphocytes, rendering them susceptible to HIV infection. There is also increasing evidence that in vitro cell treatment with Nef induces signaling effects. Exogenous Nef treatment is able to induce apoptosis in uninfected T cells, maturation in dendritic cells, and suppression of CD40-dependent immunoglobulin class switching in B cells. Previously, we reported that Nef treatment of primary human monocyte-derived macrophages (MDMs) induces a cycloheximide-independent activation of NF-kappaB and the synthesis and secretion of a set of chemokines/cytokines that activate STAT1 and STAT3. Here, we show that Nef treatment is capable of hijacking cellular signaling pathways, inducing a very rapid regulatory response in MDMs that is characterized by the rapid and transient phosphorylation of the alpha and beta subunits of the IkappaB kinase complex and of JNK, ERK1/2, and p38 mitogen-activated protein kinase family members. In addition, we have observed the activation of interferon regulatory factor 3, leading to the synthesis of beta interferon mRNA and protein, which in turn induces STAT2 phosphorylation. All of these effects require Nef myristoylation.


Subject(s)
Gene Products, nef/pharmacology , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Enzyme Activation/drug effects , Gene Products, nef/genetics , Gene Products, nef/physiology , HIV-1/chemistry , HIV-1/genetics , Humans , In Vitro Techniques , Models, Biological , Monocytes/metabolism , Monocytes/virology , Myristic Acid/metabolism , Recombinant Fusion Proteins/pharmacology , nef Gene Products, Human Immunodeficiency Virus
4.
Exp Cell Res ; 312(6): 890-900, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16445909

ABSTRACT

HIV-1 Nef is the regulatory protein expressed earliest and most abundantly in the infection cycle. Its expression has been correlated with a plethora of effects detectable either in producer, target, and bystander cells, as well as in the viral particles. Even if the relationship between Nef expression and apoptosis has been already matter of investigation in infected lymphocytes, whose resistance to HIV infection is however limited to few days, this remains to be investigated in cells that in vivo well resist the HIV cytopathic effect. In such an instance, we were interested in establishing whether Nef influences the apoptotic processes in primary human-monocyte-derived macrophages (MDM). High efficiency HIV-1 infection of MDM allowed us to establish that virus-expressed Nef strongly counteracts the HIV-1-induced apoptosis. The Nef mutant analysis suggested that this effect relies on the interaction with different protein partners and cell compartments. We also observed that the Nef protection to the HIV-1-induced apoptosis correlated with the hyper-phosphorylation and consequent inactivation of the pro-apoptotic Bad protein. On the basis of these results, we propose the Nef anti-apoptotic effect as a relevant part of the mechanism of the in vivo establishment of the HIV macrophage reservoirs.


Subject(s)
Gene Products, nef/pharmacology , HIV-1/physiology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Macrophages/cytology , Macrophages/virology , Adult , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Gene Products, nef/genetics , Gene Products, nef/metabolism , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Leukocytes, Mononuclear/drug effects , Macrophages/drug effects , Male , Mutation , Tumor Necrosis Factor-alpha/metabolism , nef Gene Products, Human Immunodeficiency Virus
5.
Eur Cytokine Netw ; 16(3): 186-90, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16266857

ABSTRACT

Interleukin (IL)-18 is a proinflammatory cytokine that plays an important role in both innate and adaptive immune responses against several infectious pathogens. Relatively little is known about its production in HIV-1 infection, and there are controversial data on the influence of IL-18 on HIV-1 replication in vitro. In this study, we investigated the effect of HIV-1 infection, and challenge with recombinant HIV-1 proteins, on IL-18 production by THP-1 cells. This is a monocytoid cell line spontaneously producing IL-18, and consequently is particularly suitable for the study of HIV-1 effects on this type of cytokine regulation. The results reported here demonstrate a significant reduction in IL-18 secretion during HIV-infection. In fact, low levels of IL-18 were released until 120 h from viral challenge (15 +/- 11 pg/mL at 24 h and 17 +/- 13 at 96 h and < 12.5 at 120 h), whereas IL-18 production by uninfected control cells was 193 +/- 104 pg/mL and 214 +/- 114 pg/mL at 24 h and 120 h respectively. At 168 h of incubation, IL-18 production by infected and uninfected cells was found to be 164 +/- 88 pg/mL and 325 +/- 101 pg/mL respectively (p = 0.001). Of the following viral proteins: gp 120, p24 and Nef, only the last one induced decreased IL-18 secretion in the supernatants of THP-1 cells. This effect is more evident with the concentrations of 5 -1.25 microg/mL of Nef protein (p < 0.0001). In conclusion, our data show that HIV-1 and its regulatory protein, Nef, are able to down-regulate the release of IL-18, in vitro. These results confirm that a variety of modulating effects on the immune response, induced by HIV-infection, may facilitate progression of HIV-1 infection.


Subject(s)
Gene Products, nef/pharmacology , HIV-1/physiology , Interleukin-18/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Monocytes/drug effects , Monocytes/immunology , Monocytes/virology , nef Gene Products, Human Immunodeficiency Virus
6.
Cell Death Differ ; 12 Suppl 1: 932-41, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15818415

ABSTRACT

Cytoskeletal components play a major role in the human immunodeficiency virus-1 (HIV-1) infection. A wide variety of molecules belonging to the microfilament system, including actin filaments and actin binding proteins, as well as microtubules have a key role in regulating both cell life and death. Cell shape maintenance, cell polarity and cell movements as well as cytoplasmic trafficking of molecules determining cell fate, including apoptosis, are in fact instructed by the cytoskeleton components. HIV infection and viral particle production seem to be controlled by cytoskeleton as well. Furthermore, HIV-associated apoptosis failure can also be regulated by the actin network function. In fact, HIV protein gp120 is able to induce cytoskeleton-driven polarization, thus sensitizing T cells to CD95/Fas-mediated apoptosis. The microfilament system seems thus to be a sort of cytoplasmic supervisor of the viral particle, the host cell and the bystander cell's very fate.


Subject(s)
Actin Cytoskeleton/virology , Apoptosis , Gene Products, nef/physiology , Gene Products, tat/physiology , Gene Products, vpr/physiology , HIV Envelope Protein gp120/physiology , HIV-1/physiology , T-Lymphocytes/virology , Viral Proteins/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Gene Products, nef/pharmacology , Gene Products, tat/pharmacology , Gene Products, vpr/pharmacology , HIV Envelope Protein gp120/pharmacology , Humans , Microfilament Proteins/metabolism , Microtubules/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/ultrastructure , Viral Proteins/pharmacology , nef Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus
7.
J Leukoc Biol ; 77(4): 522-34, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15637102

ABSTRACT

Human immunodeficiency virus (HIV) has derived a variety of means to evade the host immune response. HIV-derived proteins, including Tat, Nef, and Env, have all been reported to decrease expression of host molecules such as CD4 and major histocompatibility complex I, which would assist in limiting viral replication. The mannose receptor (MR) on the surface of macrophages and dendritic cells (DC) has been proposed to function as an effective antigen-capture molecule, as well as a receptor for entering pathogens such as Mycobacterium tuberculosis and Pneumocystis carinii. Regulation of this receptor would therefore benefit HIV in removing an additional arm of the innate immune system. Previous work has shown that MR function is reduced in alveolar macrophages from HIV-infected patients and that surface MR levels are decreased by the HIV-derived protein Nef in DC. In addition, several laboratories have shown that CD4 is removed from the surface of T cells in a manner that might be applicable to decreased MR surface expression in macrophages. In the current study, we have investigated the role of Nef in removing MR from the cell surface. We have used a human macrophage cell line stably expressing the MR as well as human epithelial cells transiently expressing CD4 and a unique CD4/MR chimeric molecule constructed from the extracellular and transmembrane domains of CD4 and the cytoplasmic tail portion of the MR. We show that the MR is reduced on the cell surface by approximately 50% in the presence of Nef and that the MR cytoplasmic tail can confer susceptibility to Nef in the CD4/MR chimera. These data suggest that the MR is a potential intracellular target of Nef and that this regulation may represent a mechanism to further cripple the host innate immune system.


Subject(s)
Gene Products, nef/pharmacology , HIV-1/physiology , Lectins, C-Type/metabolism , Macrophages/physiology , Mannose-Binding Lectins/metabolism , Protein Processing, Post-Translational , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , Flow Cytometry , HeLa Cells , Humans , Lectins, C-Type/genetics , Mannose Receptor , Mannose-Binding Lectins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , nef Gene Products, Human Immunodeficiency Virus
8.
Ann N Y Acad Sci ; 1056: 279-92, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16387695

ABSTRACT

Evidence is presented to suggest that HIV-1 accessory protein Nef could be involved in AIDS pathogenesis. When present in extracellular medium, Nef causes the death of a wide variety of cells in vitro and may therefore be responsible for the depletion of bystander cells in lymphoid tissues during HIV infection. When present inside the cell, Nef could prevent the death of infected cells and thereby contribute to increased viral load. Intracellular Nef does this by preventing apoptosis of infected cells by either inhibiting proteins involved in apoptosis or preventing the infected cells from being recognized by CTLs. Neutralization of extracellular Nef could prevent the death of uninfected immune cells and thereby the destruction of the immune system. Neutralization of intracellular Nef could hasten the death of infected cells and help reduce the viral load. Nef is therefore a very important molecular target for developing therapeutics that slow progression to AIDS. The N-terminal region of Nef and the naturally occurring bee venom mellitin have very similar primary and tertiary structures, and they both act by destroying membranes. Chemical analogs of a mellitin inhibitor prevent Nef-mediated cell death and inhibit the interaction of Nef with cellular proteins involved in apoptosis. Naturally occurring bee propolis also contains substances that prevent Nef-mediated cell lysis and increases proliferation of CD4 cells in HIV-infected cultures. These chemical compounds and natural products are water soluble and nontoxic and are therefore potentially very useful candidate drugs.


Subject(s)
AIDS Vaccines , Acquired Immunodeficiency Syndrome/immunology , Gene Products, nef/pharmacology , Acquired Immunodeficiency Syndrome/physiopathology , Amino Acid Sequence , Gene Products, nef/chemistry , Gene Products, nef/immunology , Humans , Melitten/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/immunology , Viral Regulatory and Accessory Proteins/pharmacology , nef Gene Products, Human Immunodeficiency Virus
9.
FASEB J ; 18(12): 1459-61, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15240562

ABSTRACT

The accessory HIV-1 Nef protein is a crucial determinant for viral replication and pathogenesis. During HIV infection, loss of immune control in the setting of a strong and broad HIV-specific T-lymphocyte response, leads to a lethal outcome through AIDS. Moreover, dysfunction of dendritic cells (DCs) may contribute to the immune suppression associated with AIDS progression. We recently demonstrated that exogenous Nef selectively activates immature DCs manipulating their phenotypical, morphological, and functional developmental program. Here, we tracked whether Nef, targeting DCs, could be involved in the dysregulation of CD8+ T cell responses. We found that Nef inhibits the capacity of DCs to prime alloreactive CD8+ T cell responses down-regulating their proliferation and functional competence. This coincides with the induction of CD8+ T cell apoptosis. Nef oversees apoptotic killing of CD8+ T cells up-regulating TNF-alpha and FasL production by DCs and interfering with the death receptor pathway in CD8+ T cells and thus activating caspase 8. Our findings suggest that Nef may contribute to the immune evasion associated with HIV-1 infection, subverting DC biology. This may help explain the pleiotropic function that Nef plays during infection and makes this protein an attractive target for preventive and therapeutic intervention.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gene Products, nef/pharmacology , HIV-1/pathogenicity , Immune Tolerance/drug effects , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Caspase 8 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cross-Priming/drug effects , Dendritic Cells/metabolism , Enzyme Activation/drug effects , Fas Ligand Protein , Gene Products, nef/immunology , HIV-1/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immune Tolerance/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Isoantigens/immunology , Lymphocyte Activation/drug effects , Membrane Glycoproteins/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , nef Gene Products, Human Immunodeficiency Virus
10.
J Virol ; 78(12): 6287-96, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15163722

ABSTRACT

The Nef protein enhances human immunodeficiency virus type 1 (HIV-1) replication through an unknown mechanism. We and others have previously reported that efficient HIV-1 replication in activated primary CD4(+) T cells depends on the ability of Nef to downregulate CD4 from the cell surface. Here we demonstrate that Nef greatly enhances the infectivity of HIV-1 particles produced in primary T cells. Nef-defective HIV-1 particles contained significantly reduced quantities of gp120 on their surface; however, Nef did not affect the levels of virion-associated gp41, indicating that Nef indirectly stabilizes the association of gp120 with gp41. Surprisingly, Nef was not required for efficient replication of viruses that use CCR5 for entry, nor did Nef influence the infectivity or gp120 content of these virions. Nef also inhibited the incorporation of CD4 into HIV-1 particles released from primary T cells. We propose that Nef, by downregulating cell surface CD4, enhances HIV-1 replication by inhibiting CD4-induced dissociation of gp120 from gp41. The preferential requirement for Nef in the replication of X4-tropic HIV-1 suggests that the ability of Nef to downregulate CD4 may be most important at later stages of disease when X4-tropic viruses emerge.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Gene Products, nef/pharmacology , HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Receptors, CXCR4/metabolism , Virus Replication/drug effects , CD4 Antigens/metabolism , Cells, Cultured , Down-Regulation , Gene Products, nef/metabolism , HIV Envelope Protein gp41/metabolism , HIV-1/pathogenicity , Humans , Virion/metabolism , nef Gene Products, Human Immunodeficiency Virus
11.
J Virol ; 78(6): 3099-109, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990729

ABSTRACT

The effects of soluble Nef protein on CD4(+) T cells were examined. CD4(+)-T-cell cultures exposed to soluble Nef were analyzed for apoptosis by terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling and hallmarks of apoptosis including cytoplasmic shrinkage, nuclear fragmentation, DNA laddering, and caspase activation. We observed dose- and time-dependent inductions of apoptosis. DNA laddering and activated caspase 3 were also evident. Cells treated with Nef/protein kinase inhibitor complexes were protected from Nef-induced apoptosis, suggesting possible roles for protein kinases in the apoptosis pathway. Similarly, cells treated with Nef/anti-Nef antibody complexes were protected from Nef-induced apoptosis. The cellular receptor responsible for Nef-induced apoptosis was identified through antibody- and ligand-blocking experiments as a receptor commonly involved in viral entry. CXCR4 antibodies, as well as the endogenous ligand SDF-1alpha, were effective in blocking Nef-induced apoptosis, while CCR5 and CD4 antibodies were ineffective. Moreover, a CXCR4-deficient cell line, MDA-MB-468, which was resistant to Nef-induced apoptosis, became sensitive upon transfection with a CXCR4-expressing vector. This study suggests that extracellular Nef protein could contribute to the decline of CD4 counts prior to and during the onset of AIDS in patients with human immunodeficiency virus type 1 infections.


Subject(s)
Apoptosis , Gene Products, nef/metabolism , Gene Products, nef/pharmacology , HIV-1/pathogenicity , Receptors, CXCR4/metabolism , Apoptosis/drug effects , CD4-Positive T-Lymphocytes , Cell Line , Gene Products, nef/genetics , HIV-1/metabolism , Humans , Jurkat Cells , Signal Transduction , Transfection , nef Gene Products, Human Immunodeficiency Virus
12.
J Leukoc Biol ; 74(5): 821-32, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12960275

ABSTRACT

Increasing evidence indicates that the expression of the human immunodeficiency virus-1 (HIV-1) Nef protein significantly influences the activation state of the host cell. Here we report that Nef specifically activates STAT3 in primary human monocyte-derived macrophages (MDM). This was demonstrated by both single-cycle infection experiments driven by Vesicular Stomatitis virus glycoprotein (VSV-G) pseudotyped HIV-1 and treatment with exogenous recombinant Nef. The analysis of the effects of Nef mutants revealed that domains of the C-terminal flexible loop interacting with the cell endocytotic machinery are involved in the STAT3 activation. In particular, our data suggest that the Nef-dependent STAT3 activation relies on the targeting of Nef to the late endosome/lysosome compartment. In addition, we found that Nef activates STAT3 through a mechanism mediated by the release of soluble factor(s), including MIP-1alpha, that requires de novo protein synthesis but appears independent from the activation of src tyrosine kinases. The results presented here support the idea that the first intervention of Nef in the intracellular signaling of monocyte-macrophages could generate, by means of the release of soluble factor(s), a secondary wave of activation that could be of a potential pathogenetic significance.


Subject(s)
DNA-Binding Proteins/metabolism , Endocytosis/physiology , Gene Products, nef/pharmacology , HIV-1/physiology , Macrophage Activation/drug effects , Macrophages/physiology , Monocytes/physiology , Trans-Activators/metabolism , Acute-Phase Proteins/metabolism , Cells, Cultured , Endocytosis/drug effects , Gene Products, nef/chemistry , Gene Products, nef/genetics , HIV-1/genetics , Humans , Macrophage Activation/physiology , Macrophages/drug effects , Models, Biological , Monocytes/drug effects , Phosphorylation , Recombinant Proteins/pharmacology , STAT3 Transcription Factor , nef Gene Products, Human Immunodeficiency Virus
13.
Clin Diagn Lab Immunol ; 9(6): 1212-21, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12414752

ABSTRACT

We investigated the expression of membrane-bound CD14 (mCD14) on monocytes and soluble CD14 (sCD14) released into the culture supernatants of peripheral blood lymphocytes (PBMC) from human immunodeficiency virus (HIV)-infected individuals. Monocytes from HIV-positive individuals exhibited both enhanced mCD14 expression and sCD14 production in the PBMC culture supernatants compared to the levels of mCD14 and sCD14 in HIV-negative individuals. This enhanced mCD14 expression and sCD14 production in HIV-infected individuals may be due to the effects of cytokines, the bacterial product lipopolysaccharide (LPS), and/or the HIV regulatory antigens Tat and Nef. Interleukin-10 (IL-10), an immunoregulatory cytokine, as well as LPS enhanced mCD14 expression and the release of sCD14 in the culture supernatants. HIV-Nef, unlike Tat, enhanced mCD14 expression on monocytes but did not induce the release of sCD14 into the culture supernatants. Studies conducted to investigate the mechanism underlying HIV-Nef-induced mCD14 expression revealed that HIV-Nef upregulated mCD14 expression via a mechanism that does not involve endogenously produced IL-10. In contrast, LPS upregulated the expression of mCD14 and increased the release of sCD14 via a mechanism that involves, at least in part, endogenously produced IL-10. Furthermore, dexamethasone, an anti-inflammatory and immunosuppressive agent, inhibited HIV-Nef-induced CD14 expression in an IL-10-independent manner. In contrast, dexamethasone inhibited IL-10-dependent LPS-induced CD14 expression by interfering with IL-10-induced signals but not by blocking IL-10 production. These results suggest that HIV-Nef and IL-10 constitute biologically important modulators of CD14 expression which may influence immunobiological responses to bacterial infections in HIV disease.


Subject(s)
Gene Products, nef/pharmacology , HIV/chemistry , Interleukin-10/physiology , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Adult , Dexamethasone/pharmacology , Humans , nef Gene Products, Human Immunodeficiency Virus
14.
Immunol Lett ; 84(2): 97-101, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12270545

ABSTRACT

Interleukin-10 (IL-10) plays an important immunopathogenic role in immunologic diseases, especially in HIV infection and atopic dermatitis. The control and regulatory mechanisms of IL-10 production have not been described in these diseases. Recently, we demonstrated that HIV-1 Nef induces IL-10 production in monocytes and that staphylococcal enterotoxin A (SEA) induces IL-10 production in T-lymphocytes. Here we show that Nef-induced IL-10 production and mRNA expression are strongly blocked by rapamycin, but are not blocked by cyclosporin (CsA) or FK506. Conversely, we show that CsA and FK506 completely inhibit SEA-induced IL-10 protein production and mRNA expression. The results of this study demonstrate that IL-10 production by Nef and SEA is controlled and regulated by different mechanisms.


Subject(s)
Enterotoxins/immunology , Gene Products, nef/immunology , HIV-1/immunology , Interleukin-10/metabolism , Leukocytes, Mononuclear/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Enterotoxins/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Products, nef/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Interleukin-10/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , RNA, Messenger/analysis , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , Tacrolimus/pharmacology , nef Gene Products, Human Immunodeficiency Virus
15.
Clin Diagn Lab Immunol ; 9(5): 983-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12204947

ABSTRACT

Dysfunction of neutrophils (polymorphonuclear leukocytes [PMNL]) and macrophagic cells occurs as a consequence of human immunodeficiency virus type 1 (HIV-1) infection. Macrophages contribute to the resolution of early inflammation ingesting PMNL apoptotic bodies. This study investigated macrophage ability to phagocytose PMNL apoptotic bodies in patients with HIV-1 infection in comparison with uninfected individuals and the effect of HIV Nef protein on apoptotic body phagocytosis to determine if phagocytic activity is impaired by HIV infection. Monocytes/macrophages were isolated from 10 HIV-1-infected patients and from five healthy volunteers, whereas PMNL were isolated from healthy volunteers. Macrophage phagocytosis of apoptotic PMNL was determined by staining of apoptotic bodies with fluorescein-conjugated concanavalin A or with fluorescein-labeled phalloidin. Our data show significant impairment of PMNL apoptotic body macrophage phagocytosis in subjects with HIV-1 infection presenting a concentration of CD4(+) T lymphocytes of >200/mm(3) and in particular in those with <200 CD4(+) T lymphocyte cells/mm(3). In addition, HIV-1 recombinant Nef protein is able to decrease phagocytosis of apoptotic PMNL from normal human macrophages in a dose-dependent manner. The results of our study suggest that impaired macrophage phagocytosis of PMNL apoptotic bodies may contribute to the persistence of the inflammatory state in HIV-infected subjects, especially during opportunistic infections that are often favored by defective phagocytic activity.


Subject(s)
Apoptosis/immunology , HIV Infections/immunology , HIV-1 , Macrophages/immunology , Neutrophils/immunology , Phagocytosis/immunology , Adult , Gene Products, nef/immunology , Gene Products, nef/pharmacology , Humans , Macrophages/metabolism , Macrophages/virology , Neutrophils/virology , Oxidation-Reduction , Phagocytosis/drug effects , nef Gene Products, Human Immunodeficiency Virus
16.
Exp Cell Res ; 275(2): 243-54, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11969293

ABSTRACT

Human immunodeficiency virus (HIV)-1 Nef protein is an essential modulator of AIDS pathogenesis and we have previously demonstrated that rNef enters uninfected human monocytes and induces T cells bystander activation, up-regulating IL-15 production. Since dendritic cells (DCs) play a central role in HIV-1 primary infection we investigated whether rNef affects DCs phenotypic and functional maturation in order to define its role in the immunopathogenesis of AIDS. We found that rNef up-regulates the expression on immature DCs of surface molecules known to be critical for their APC function. These molecules include CD1a, HLA-DR, CD40, CD83, CXCR4, and to a lower extent CD80 and CD86. On the other hand, rNef down-regulates surface expression of HLA-ABC and mannose receptor. The functional consequence of rNef treatment of immature DCs is a decrease in their endocytic and phagocytic activities and an increase in cytokine (IL-1beta, IL-12, IL-15, TNF-alpha) and chemokine (MIP-1alpha, MIP-1beta, IL-8) production as well as in their stimulatory capacity. These results indicate that rNef induces a coordinate series of phenotypic and functional changes promoting DC differentiation and making them more competent APCs. Indeed, Nef induces CD4(+) T cell bystander activation by a novel mechanism involving DCs, thus promoting virus dissemination.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Products, nef/pharmacology , HIV-1/pathogenicity , Lectins, C-Type , Lymphocyte Activation , Mannose-Binding Lectins , Antigen Presentation , Antigens, CD/biosynthesis , Cell Differentiation , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/drug effects , Endocytosis , HLA-DR Antigens/biosynthesis , Humans , Kinetics , Mannose Receptor , Phagocytosis , Receptors, Cell Surface/biosynthesis , Up-Regulation , nef Gene Products, Human Immunodeficiency Virus
17.
J Virol ; 76(8): 3587-95, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11907198

ABSTRACT

Human and simian immunodeficiency virus (HIV and SIV, respectively) infections are characterized by gradual depletion of CD4+ T cells. The underlying mechanisms of CD4+ T-cell depletion and HIV and SIV persistence are not fully determined. The Nef protein is expressed early in infection and is necessary for pathogenesis. Nef can cause T-cell activation and downmodulates cell surface signaling molecules. However, the effect of Nef on the cell cycle has not been well characterized. To determine the role of Nef in the cell cycle, we investigated whether the SIV Nef protein can modulate cell proliferation and apoptosis in CD4+ Jurkat T cells. We developed a CD4+ Jurkat T-cell line that stably expresses SIV Nef under the control of an inducible promoter. Alterations in cell proliferation were determined by flow cytometry using stable intracytoplasmic fluorescent dye 5- and 6-carboxyfluorescein diacetate succinimidyl ester and bromodeoxyuridine incorporation. Apoptotic cell death was measured by annexin V and propidium iodide staining. Our results demonstrated that SIV Nef inhibited Fas-induced apoptosis in these cells and that the mechanism involved upregulation of the Bcl-2 protein. SIV Nef suppressed CD4+ T-cell proliferation by inhibiting the progression of cells into S phase of the cell cycle. Suppression involved an upregulation of cyclin-dependent kinase inhibitors p21 and p27 and the downregulation of cyclin D1 and cyclin A. In summary, inhibition of apoptosis by Nef can lead to persistence of infected cells and can support viral replication. In addition, a Nef-mediated delay in cell cycle progression may contribute to CD4+ T-cell anergy/depletion seen in HIV and SIV disease.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , G1 Phase/drug effects , Gene Products, nef/pharmacology , S Phase/drug effects , Animals , Apoptosis , CD4-Positive T-Lymphocytes/virology , Cell Division/drug effects , Cell Line , Gene Products, nef/genetics , Gene Products, nef/metabolism , Humans , Jurkat Cells , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Transfection
18.
J Virol ; 76(7): 3179-88, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884542

ABSTRACT

The cerebral complement system is hypothesized to contribute to neurodegeneration in the pathogenesis of AIDS-associated neurological disorders. Our former results have shown that the human immunodeficiency virus (HIV) strongly induces the synthesis of complement factor C3 in astrocytes. This upregulation explains in vivo data showing elevated complement levels in the cerebrospinal fluid of patients with AIDS-associated neurological symptoms. Since inhibition of complement synthesis and activation in the brain may represent a putative therapeutic goal to prevent virus-induced damage, we analyzed in detail the mechanisms of HIV-induced modulation of C3 expression. HIV-1 increased the C3 levels in astrocyte culture supernatants from 30 to up to 400 ng/ml; signal transduction studies revealed that adenylate cyclase activation with upregulation of cyclic AMP is the central signaling pathway to mediate that increase. Furthermore, activity of protein kinase C is necessary for HIV induction of C3, since inhibition of protein kinase C by prolonged exposure to the phorbol ester tetradecanoyl phorbol acetate partly abolished the HIV effect. The cytokines tumor necrosis factor alpha and gamma interferon were not involved in mediating the HIV-induced C3 upregulation, since neutralizing antibodies had no effect. Besides whole HIV virions, the purified viral proteins Nef and gp41 are biologically active in upregulating C3, whereas Tat, gp120, and gp160 were not able to modulate C3 synthesis. Further experiments revealed that neurons were also able to respond on incubation with HIV with increased C3 synthesis, although the precise pattern was slightly different from that in astrocytes. This strengthens the hypothesis that HIV-induced complement synthesis represents an important mechanism for the pathogenesis of AIDS in the brain.


Subject(s)
Astrocytes/immunology , Complement C3/biosynthesis , HIV-1/immunology , Neurons/immunology , Adenylyl Cyclases/pharmacology , Astrocytes/virology , Cell Line , Cyclic AMP/metabolism , Gene Products, nef/pharmacology , HIV Envelope Protein gp41/pharmacology , HIV Infections/immunology , HIV Infections/virology , Humans , Neurons/virology , Signal Transduction/drug effects , Up-Regulation , nef Gene Products, Human Immunodeficiency Virus
19.
J Biomed Sci ; 9(1): 82-96, 2002.
Article in English | MEDLINE | ID: mdl-11810028

ABSTRACT

The early human immunodeficiency virus (HIV) accessory protein Nef makes an important contribution to virulence, but the mechanisms by which Nef influences pathogenesis remain unclear. Many well-studied effects of Nef, like CD4 and class I MHC downregulation, occur posttranslationally. However, Nef has the potential to affect gene expression by interfering with cell signaling pathways and by virtue of structural features such as the Pro-X-X-Pro motif, which may interact with src homology region-3 domains of src-like kinases. We used a cDNA microarray screening strategy to identify cellular genes whose steady state transcriptional levels may be affected by Nef. We generated HeLa cell lines expressing wild-type or mutant HIV-1 nef protein sequences. Using cDNA microarray technology, we compared the patterns of cellular gene expression in the various cell lines to the pattern in non-Nef-expressing HeLa cells. By matching the patterns of cellular gene expression in HeLa cell lines expressing various Nefs with that of parental HeLa cells, we identified several cellular genes whose expression was modulated differentially by Nef and its mutants. We confirmed the differential expression of selected genes by RNA filter blotting. Genes expressed at higher levels included proteases, transcription factors, protein kinases, nuclear import/export proteins, adaptor molecules and cyclins, some of which have previously been implicated as being important for HIV replication and pathogenesis. The results indicate that Nef expression can alter the expression of cellular genes and suggest that this alteration in cellular gene expression may serve to optimize the cell to support the subsequent stages of viral replication.


Subject(s)
Gene Expression Profiling , Gene Products, nef/genetics , Gene Products, nef/pharmacology , Gene Expression/drug effects , HIV Infections/etiology , HIV Infections/metabolism , HIV-1/chemistry , HeLa Cells , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Transcription, Genetic/drug effects , Transfection , nef Gene Products, Human Immunodeficiency Virus
20.
Blood ; 98(9): 2752-61, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675348

ABSTRACT

Monocytes/macrophages play a predominant role in the immunologic network by secreting and reacting to a wide range of soluble factors. Human immunodeficiency virus (HIV) infection leads to deep immunologic dysfunctions, also as a consequence of alterations in the pattern of cytokine release. Recent studies on in vivo models demonstrated that the expression of HIV Nef alone mimics many pathogenetic effects of HIV infection. In particular, Nef expression in monocytes/macrophages has been correlated with remarkable modifications in the pattern of secreted soluble factors, suggesting that the interaction of Nef with monocytes/macrophages plays a role in the pathogenesis of acquired immunodeficiency syndrome (AIDS). This study sought to define possible alterations in intracellular signaling induced by Nef in monocytes/macrophages. Results demonstrate that HIV-1 Nef specifically activates both alpha and beta isoforms of the signal transducer and activator of transcription 1 (STAT1). This was observed both by infecting human monocyte-derived macrophages (MDMs) with HIV-1 deletion mutants, and by exploiting the ability of MDMs to internalize soluble, recombinant Nef protein (rNef). STAT1-alpha activation occurs on phosphorylation of both C-terminal Tyr701 and Ser727 and leads to a strong binding activity. Nef-dependent STAT1 activation is followed by increased expression of both STAT1 and interferon regulatory factor-1, a transcription factor transcriptionally regulated by STAT1 activation. It was also established that Nef-induced STAT1- alpha/beta activation occurs through the secretion of soluble factors. Taken together, the results indicate that HIV-1 Nef could interfere with STAT1-governed intracellular signaling in human monocytes/macrophages.


Subject(s)
Cytokines/metabolism , DNA-Binding Proteins/drug effects , Gene Products, nef/pharmacology , Macrophages/drug effects , Trans-Activators/drug effects , Acquired Immunodeficiency Syndrome/blood , Acquired Immunodeficiency Syndrome/etiology , Adult , Cytokines/drug effects , Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Gene Products, nef/genetics , Gene Products, nef/physiology , HIV-1/chemistry , HIV-1/genetics , Humans , Interferon Regulatory Factor-1 , Macrophages/metabolism , Macrophages/virology , Male , Monocytes/drug effects , Monocytes/metabolism , Monocytes/virology , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Isoforms/drug effects , Protein Isoforms/metabolism , STAT1 Transcription Factor , Sequence Deletion , Signal Transduction/drug effects , Trans-Activators/metabolism , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...