Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Allergy ; 68(6): 732-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23621380

ABSTRACT

BACKGROUND: It is commonly acknowledged that asthma is exacerbated by viral infections. On the other hand, basophil infiltration of lung tissues has been evidenced postmortem in cases of fatal disease, raising the question of a possible link between these two observations. OBJECTIVES: Herein, we addressed the relationship between asthma exacerbation by viral infection and basophil activation and expansion by investigating how stimulation with the dsRNA polyadenylic/polyuridylic acid [poly(A:U)] affected basophil activities and recruitment in an allergic airway inflammation model. METHODS: The effect of dsRNA on basophils was assessed by measuring the cytokine levels produced upon stimulation. We used an OVA-induced experimental model of allergic asthma. Airway hyperreactivity, recruitment of infiltrating cells, and cytokine production were determined in the lung of mice having received poly(A:U), as compared with untreated controls. The exacerbating effect of basophils was assessed both by adoptive transfer of poly(A:U)-treated basophils and by their in vivo depletion with Ba103 antibody. RESULTS: We found that in vitro treatment with poly(A:U) increased basophil functions by inducing TH 2-type cytokine and histamine production, whereas in vivo treatment increased peripheral basophil recruitment. Furthermore, we provide the first demonstration for increased infiltration of basophils in the lung of mice suffering from airway inflammation. In this model, disease symptoms were clearly exacerbated upon adoptive transfer of basophils exposed to poly(A:U), relative to their unstimulated counterpart. Conversely, in vivo basophil depletion alleviated disease syndromes, thus validating the transfer data. CONCLUSIONS: Our findings provide the first evidence for airway inflammation exacerbation by basophils following dsRNA stimulation.


Subject(s)
Asthma/immunology , Basophils/metabolism , Lung/immunology , Poly A-U/metabolism , RNA, Double-Stranded/metabolism , Animals , Asthma/virology , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Progression , Female , Flow Cytometry , Histamine/metabolism , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Poly A-U/administration & dosage , Th2 Cells/metabolism
2.
Atherosclerosis ; 184(2): 330-41, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16051252

ABSTRACT

Strategies aimed at treating atherosclerosis by immunization protocols are emerging. Such protocols commonly use adjuvants as non-specific stimulators of immune responses. However, adjuvants are known to modify various disease processes. The aim of this study was to determine whether adjuvants alter the development of atherosclerosis. We performed immunization protocols in apolipoprotein E knockout mice (E degrees ) following chronic administration schedules commonly employed in experimental atherosclerosis. Our results point out a dramatic effect of several adjuvants on the development of atherosclerosis; three of the four adjuvants tested reduced lesion size. The Alum adjuvant, which is the adjuvant currently used in most vaccination protocols in humans, displayed a strong atheroprotective effect. Mechanisms accounting for atheroprotective effect of Freund's adjuvants included their capacity to increase both Th2 responses and anti-MDA-LDL IgM titers, and/or to impose atheroprotective lipoprotein profiles. The present study indicates that adjuvants have potent atheromodulating capabilities, and thus, implies that the choice of adjuvant is crucial in long-term immunization protocols in experimental atherosclerosis.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alum Compounds/therapeutic use , Atherosclerosis/drug therapy , Freund's Adjuvant/therapeutic use , Immunization/methods , Animals , Antibodies, Anti-Idiotypic/immunology , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/immunology , Cytokines/blood , Disease Models, Animal , Follow-Up Studies , Gas Chromatography-Mass Spectrometry , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Mice , Mice, Knockout , Time Factors , Treatment Outcome
3.
J Immunol ; 166(3): 1755-62, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160221

ABSTRACT

The NK1.1(+)TCRalphabeta(int) CD4(+), or double negative T cells (NK T cells) consist of a mixture of CD1d-restricted and CD1d-unrestricted cells. The relationships between CD4(+)NK1.1(+) T cells and conventional T cells are not understood. To compare their respective TCR repertoires, NK1.1(+)TCRalphabeta(int), CD4(+) T cells have been sorted out of the thymus, liver, spleen, and bone marrow of C57BL/6 mice. Molecular analysis showed that thymus and liver used predominantly the Valpha14-Jalpha281 and Vbeta 2, 7, and 8 segments. These cells are CD1d restricted and obey the original definition of NK T cells. The complementarity-determining region 3 (CDR3) sequences of the TCR Vbeta8.2-Jbeta2.5 chain of liver and thymus CD4(+) NK T cells were determined and compared with those of the same rearrangements of conventional CD4(+) T cells. No amino acid sequence or usage characteristic of NK T cells could be evidenced: the Vbeta8.2-Jbeta2.5 diversity regions being primarily the same in NK T and in T cells. No clonal expansion of the beta-chains was observed in thymus and liver CD1d-restricted CD4(+)NK T cells, suggesting the absence of acute or chronic Ag-driven stimulation. Molecular analysis of the TCR used by Valpha14-Jalpha281 transgenic mice on a Calpha(-/-) background showed that the alpha-chain can associate with beta-chains using any Vbeta segment, except in NK T cells in which it paired predominately with Vbeta 2, 7, and 8(+) beta-chains. The structure of the TCR of NK T cells thus reflects the affinity for the CD1d molecule rather than a structural constraint leading to the association of the invariant alpha-chain with a distinctive subset of Vbeta segment.


Subject(s)
CD4 Antigens/biosynthesis , Killer Cells, Natural/metabolism , Proteins , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Antigens/biosynthesis , Antigens, Ly , Antigens, Surface , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Separation , Clone Cells , Gene Expression Regulation/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Genes, T-Cell Receptor alpha/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lectins, C-Type , Liver/cytology , Liver/immunology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B , Organ Specificity/immunology , Protein Biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
4.
Infect Immun ; 67(9): 4485-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456890

ABSTRACT

Macrophages are among the most sensitive targets of bacterial endotoxin (LPS), responding to minute amounts of LPS by releasing a battery of inflammatory mediators. Transfection of macrophages with secretory leukocyte protease inhibitor (SLPI) renders these cells refractory to LPS stimulation. Here we show that uptake of LPS from soluble CD14 (sCD14)-LPS complexes by SLPI-overexpressing cells was only 50% of that seen in control cells. SLPI transfectants and mock transfectants did not differ in the surface expression of CD14 or CD18. We show, in addition, that recombinant human SLPI can bind to purified endotoxin in vitro. SLPI caused a decrease in the binding of LPS to sCD14 as assessed both by fluorescence quenching of labeled LPS and by nondenaturing polyacrylamide gel electrophoresis. These results suggest that the inhibitory effect of SLPI on macrophage responses to LPS may, in part, be due to its blockade of LPS transfer to soluble CD14 and its interference with uptake of LPS from LPS-sCD14 complexes by macrophages.


Subject(s)
Lipopolysaccharides/metabolism , Macrophages/metabolism , Proteins/metabolism , Serine Proteinase Inhibitors/metabolism , CD18 Antigens/biosynthesis , Cell Line, Transformed , Humans , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Nitrites/metabolism , Proteinase Inhibitory Proteins, Secretory , Proteins/genetics , Secretory Leukocyte Peptidase Inhibitor , Tumor Necrosis Factor-alpha/biosynthesis
5.
J Exp Med ; 190(4): 523-34, 1999 Aug 16.
Article in English | MEDLINE | ID: mdl-10449523

ABSTRACT

Addition of lipopolysaccharide (LPS) to cells in the form of LPS-soluble (s)CD14 complexes induces strong cellular responses. During this process, LPS is delivered from sCD14 to the plasma membrane, and the cell-associated LPS is then rapidly transported to an intracellular site. This transport appears to be important for certain cellular responses to LPS, as drugs that block transport also inhibit signaling and cells from LPS-hyporesponsive C3H/HeJ mice fail to exhibit this transport. To identify the intracellular destination of fluorescently labeled LPS after its delivery from sCD14 into cells, we have made simultaneous observations of different organelles using fluorescent vital dyes or probes. Endosomes, lysosomes, the endoplasmic reticulum, and the Golgi apparatus were labeled using Texas red (TR)-dextran, LysoTrackertrade mark Red DND-99, DiOC6(3), and boron dipyrromethane (BODIPY)-ceramide, respectively. After 30 min, LPS did not colocalize with endosomes, lysosomes, or endoplasmic reticulum in polymorphonuclear leukocytes, although some LPS-positive vesicles overlapped with the endosomal marker, fluorescent dextran. On the other hand, LPS did appear to colocalize with two markers of the Golgi apparatus, BODIPY-ceramide and TRITC (tetramethylrhodamine isothiocyanate)-labeled cholera toxin B subunit. We further confirmed the localization of LPS in the Golgi apparatus using an epithelial cell line, HeLa, which responds to LPS-sCD14 complexes in a CD14-dependent fashion: BODIPY-LPS was internalized and colocalized with fluorescently labeled Golgi apparatus probes in live HeLa cells. Morphological disruption of the Golgi apparatus in brefeldin A-treated HeLa cells caused intracellular redistribution of fluorescent LPS. These results are consistent with the Golgi apparatus being the primary delivery site of monomeric LPS.


Subject(s)
Golgi Apparatus/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Biological Transport , Biomarkers , Boron Compounds , Cell Compartmentation , Cytoplasmic Granules , Dose-Response Relationship, Drug , Endoplasmic Reticulum , Fluorescent Dyes , HeLa Cells , Humans , Interleukin-8/metabolism , Lysosomes , Microscopy, Confocal , Molecular Probes , Neutrophils , Signal Transduction
6.
J Leukoc Biol ; 65(6): 750-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10380895

ABSTRACT

Secretory nonpancreatic group IIA phospholipase A2 (sPLA2), a lipolytic enzyme found in plasma, is thought to play an important role in inflammation. In patients with sepsis, a strong positive correlation is observed between the plasma level of sPLA2 and poor clinical outcome in sepsis. We have thus asked whether sPLA2 could play a role in enabling responses of cells to bacterial lipopolysaccharide (LPS), a key contributor to sepsis. In the presence of sPLA2, cellular responses to LPS were significantly increased. This was demonstrated in assays of LPS-stimulated interleukin-6 (IL-6) production in whole blood and binding of freshly isolated human polymorphonuclear neutrophils (PMN) to fibrinogen-coated surfaces. We further found that sPLA2 enhanced binding of labeled LPS to PMN, and that the sPLA2-mediated cell responses to LPS were all blocked by monoclonal antibodies directed against membrane CD14. Two properties ofsPLA2 may contribute to its activity to mediate responses to LPS. sPLA2 appears to bind LPS because pre-exposure of sPLA2 to LPS led to a dose-dependent increase in its ability to hydrolyze phospholid substrate, and incubation of sPLA2 with BODIPY-LPS micelles resulted in enhanced fluorescence, presumably from the disaggregation of the LPS aggregates. Additional studies demonstrated that the esterolytic function of sPLA2 is also needed both for the disaggregation of LPS and CD14-dependent cell stimulation. The precise mechanisms by which LPS-binding and esterolytic activity contribute to sPLA2 activity are not clear but our data strongly suggest that these activities result in interaction of LPS with CD14 and subsequent cell activation.


Subject(s)
Leukocytes/physiology , Lipopolysaccharides/pharmacology , Phospholipases A/pharmacology , Boron Compounds/blood , Fluorescent Dyes/metabolism , Humans , Interleukin-6/blood , Leukocytes/drug effects , Neutrophils/drug effects , Phospholipases A2 , Shock, Septic/enzymology
7.
Immunity ; 8(6): 771-7, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9655491

ABSTRACT

Lipopolysaccharide ([LPS], an endotoxin) from most bacterial species provokes a strong inflammatory response in naive animals. LPS from Rhodobacter sphaeroides (RsLPS) has a relatively small hydrophobic region and does not stimulate cells or animals but instead acts as antagonist of LPS action. Here, we show that the activity of RsLPS is transformed from antagonist to full agonist by the addition of chlorpromazine (CPZ) and other cationic membrane-active agents. In addition, while LPS is rapidly transported from the plasma membrane to an intracellular site, we find that RsLPS is not transported but instead remains in the cell periphery. Addition of CPZ also reverses this behavior, causing RsLPS to be transported to a perinuclear site. The data suggest that the interaction of LPS with membrane lipids is influenced by membrane-modifying agents such as CPZ, and these interactions dictate both its intracellular transport and its ability to stimulate cellular responses.


Subject(s)
Endocytosis/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Lipopolysaccharides/agonists , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/immunology , Signal Transduction/immunology , Animals , Biological Transport/immunology , Humans , Lipopolysaccharides/chemistry , Rhodobacter sphaeroides
8.
J Exp Med ; 185(12): 2095-100, 1997 Jun 16.
Article in English | MEDLINE | ID: mdl-9182681

ABSTRACT

We have recently shown that monomeric bacterial LPS is rapidly delivered from the plasma membrane to an intracellular site and that agents that block vesicular transport block responses of neutrophils to lipopolysaccharide (LPS) (Detmers, P.A., N. Thiéblemont, T. Vasselon, R. Pironkova, D.S. Miller, and S.D. Wright. 1996. J. Immunol. 157:5589-5596). To examine further the connection between intracellular transport of LPS and signaling, we observed internalization of fluorescently labeled LPS in cells from LPS-hyporesponsive (Lpsd) mice. Binding of fluorescent LPS from LPS-soluble CD14 (sCD14) complexes by peritoneal macrophages from Lpsd and control (Lpsn) mice was quantitatively similar, and confocal images obtained from these cells exhibited an identical appearance immediately after labeling. Incubation of labeled Lpsn macrophages at 37 degrees C caused movement of the fluorescence from the cell perimeter in one or two spots in the perinuclear region. However, in Lpsd cells the fluorescence remained dispersed, suggesting a defect in vesicular transport. LPS resembles ceramide, and Lpsd mice fail to respond to ceramide. As with LPS, we found that binding of fluorescent ceramide by Lpsd and Lpsn macrophages was quantitatively similar, and the label moved rapidly to one to two spots in the perinuclear region in Lpsn mice. However, in Lpsd macrophages the fluorescence remained dispersed. These results show that cells deficient in responses to LPS exhibit defective vesicular transport of LPS and ceramide and point to a role for vesicular transport in responses to these mediators.


Subject(s)
Ceramides/pharmacokinetics , Endocytosis , Lipopolysaccharides/pharmacology , Lipopolysaccharides/pharmacokinetics , Animals , Biological Transport , Cells, Cultured , Macrophages/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
9.
Clin Immunol Immunopathol ; 83(1): 60-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9073537

ABSTRACT

We have used two-color flow cytometry to study in vivo monocytic cytokine production at the single-cell level in HIV-infected patients. We demonstrated the presence of intracellular IL-1 alpha, IL-1 beta, IL-1ra, and TNF alpha in circulating CD14+ monocytes from HIV-infected patients. The specificity of intracellular staining with anti-cytokine antibodies was demonstrated by the suppression of the fluorescent signal when staining was performed in the presence of recombinant cytokines. We did not detect any specific intracellular staining when anti-IL-4 antibodies were used since monocytes do not produce IL-4. In vivo intracellular cytokine production of IL-1 alpha, IL-1 beta, IL-1ra, and TNF alpha was higher in monocytes from HIV-infected individuals compared to monocytes from healthy controls; however, only the data concerning IL-1 alpha reached statistical significance. Monocytic cytokines are involved in the regulation of HIV gene expression and may participate in the modulation of the Th1/Th2 balance. The ability to follow the production of a wide range of cytokines by circulating monocytes of HIV-infected patients should allow one to better analyze the role of monocytic cytokines in the pathogenesis of HIV disease.


Subject(s)
Cytokines/biosynthesis , HIV Seropositivity/blood , Monocytes/metabolism , Flow Cytometry , HIV Seronegativity/immunology , Humans , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/blood , Lipopolysaccharide Receptors/blood , Monocytes/immunology , Receptors, Interleukin-1/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Sialoglycoproteins/blood , Tumor Necrosis Factor-alpha/metabolism
10.
J Immunol ; 158(3): 1310-4, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9013974

ABSTRACT

Several studies have pointed to a link between immune and endocrine systems, including a regulatory function of GH on monocyte activation. The present study demonstrates that human THP-1 promonocytic cells, engineered by gene transfer to constitutively produce human growth hormone (hGH), secreted depressed amounts of TNF-alpha in response to challenge by LPS. The effect of GH appears to occur in an autocrine fashion, since the inhibitory effect on TNF-alpha secretion by constitutive GH production could be abolished in the presence of anti-hGH mAb. The GH-induced inhibitory effect was also observed using normal human monocytes and monocyte-derived macrophages. Inhibition of TNF-alpha production by THP-1-hGH-transfected cells cultured in the presence of LPS is dependent on a selective pathway, since no inhibition of TNF-alpha production was observed when cells were cultured in the presence of PMA. Inhibition of TNF-alpha secretion by LPS-stimulated THP-1-hGH cells was associated with a decrease in nuclear translocation of nuclear factor-kappaB. The capacity of GH to inhibit LPS-induced TNF-alpha production by monocytes without altering other pathways leading to TNF-alpha production may be of potential relevance in septic shock, since GH is available for clinical use.


Subject(s)
Human Growth Hormone/physiology , Monocytes/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , CD4 Antigens/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Human Growth Hormone/pharmacology , Humans , Lipopolysaccharides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transfection
11.
J Immunol ; 157(12): 5589-96, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8955211

ABSTRACT

Human polymorphonuclear leukocytes (PMN) respond to LPS with strongly increased integrin-mediated adhesion. While the first step of this process has been identified as the interaction of LPS with CD14 on the cell surface, subsequent steps remain to be elucidated. The experiments presented here suggest that monomeric LPS is internalized in vesicles, and uptake may be required for signaling. Fluorescently labeled LPS presented as monomeric complexes with soluble CD14 appeared in the plasma membrane of PMN by 5 min and was concentrated in cytoplasmic vesicles by 20 min. Adhesion in response to LPS/soluble CD14 occurred only after a 15- to 20-min lag period, consistent with endocytosis occurring before signal generation. In contrast, there was no time lag for adhesion in response to the formyl peptide formyl-norleucyl-leucyl-phenylalanine (fNLLP). Adhesion in response to LPS, but not fNLLP, was completely blocked by lowering the temperature to 19 degrees C, a procedure that prevents vesicle fusion. These studies indicated that an event with the time and temperature dependence of endocytosis precedes signaling by LPS. Cytochalasin D, an inhibitor of phagocytosis, and wortmannin, an inhibitor of phosphatidylinositol 3-kinase that blocks vesicle fusion and phagocytosis, both completely blocked adhesion in response to LPS but not in response to fNLLP. These results support the idea that LPS internalization and early endosomal fusion may be required for signal transduction. Parallel studies showed that the adhesion response to TNF had time, temperature, and inhibitor sensitivities nearly identical with those of LPS, suggesting that responses to TNF may also include an obligate vesicle fusion step.


Subject(s)
Lipopolysaccharides/pharmacology , Neutrophils/physiology , Tumor Necrosis Factor-alpha/pharmacology , Cell Adhesion/drug effects , Cell Membrane/physiology , Cells, Cultured , Cytochalasin D/pharmacology , Endocytosis , Enzyme Activation/drug effects , Humans , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Temperature
12.
Eur J Immunol ; 25(12): 3418-24, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8566032

ABSTRACT

Infection with the human immunodeficiency virus HIV-1 is associated with the expansion of a CD14lowCD16high monocyte subset in peripheral blood. This subset, which represents a minor subpopulation of monocytes in healthy individuals, increases during HIV infection and, in patients with AIDS, may represent up to 40% of the total circulating monocyte cell population. The CD14lowCD16high circulating monocytes co-express MAX.1, p150,95 and HLA-DR which are typical of tissue macrophage markers. These cells also express higher levels of intracellular interleukin (IL)-1 alpha and tumor necrosis factor (TNF)-alpha than the CD14highCD16low monocyte population from the same patients. The CD14lowCD16high cells also express low levels of CD35, CD11a and CD4 in common with normal monocytes. When cultured in vitro, monocytes from HIV-seropositive individuals differentiated within a few hours into an elongated fibroblastoid shape characteristic of migratory cells. Our results suggest that the expansion of the CD14lowCD16high monocyte subset, which produce high amount sof TNF-alpha and IL-1 alpha, may participate in the immune dysfunction observed during HIV infection.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Lipopolysaccharide Receptors , Monocytes/cytology , Monocytes/metabolism , Receptors, IgG , Cell Division/immunology , HIV Seronegativity/immunology , HIV Seropositivity/immunology , Humans , Immunophenotyping , Interleukin-1/biosynthesis , Lipopolysaccharide Receptors/analysis , Monocytes/classification , Receptors, IgG/analysis , Tumor Necrosis Factor-alpha/biosynthesis
13.
J Immunol ; 155(10): 4861-7, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-7594489

ABSTRACT

Monocyte/macrophages may harbor HIV in a nonproductive fashion for prolonged periods of time. Viral gene expression may be reactivated by stimulation of the cells with LPS or cytokines such as TNF-alpha in vitro. The effect of LPS and TNF-alpha is mediated by their ability to induce nuclear translocation of the DNA-binding heterodimer NF-kappa B (p50/p65), which binds to a specific sequence in the HIV-long terminal repeat. The present study demonstrates that triggering of complement receptors CR1 (CD35) and CR3 (CD11b/CD18) enhances viral replication in HIV-infected human monocytic cells. Monocytic cell lines and normal peripheral blood monocytes were infected with HIV-1 in vitro and cultured in the presence or absence of F(ab')2 fragments of monoclonal anti-CR1 or anti-CR3 Abs or with C3 fragments. Stimulation of CR1 or CR3 induces a two- to fourfold increase in the amount of cell-associated and released p24 Ag in cell cultures that was equivalent to that observed in control cultures triggered with LPS. We further observed that stimulation of CR1 or CR3 induces the nuclear translocation of NF-kappa B p50/p65 in infected cells. Translocation of NF-kappa B p50/p65 was also observed following stimulation of CR1 or CR3 of uninfected peripheral blood monocytes from HIV-seronegative donors. The amount of protein translocated was similar to that observed when cells were stimulated with rhTNF-alpha. TNF-alpha did not mediate the translocation of NF-kappa B p50/p65 induced by triggering of complement receptors. Taken together, these observations suggest that HIV gene expression may be activated in infected monocytes through interaction of the cells with complement-opsonized particles and that enhanced viral replication is associated with C3 receptor-mediated nuclear translocation of the NF-kappa B complex.


Subject(s)
CD11 Antigens/physiology , DNA Replication , HIV Infections/physiopathology , HIV-1/physiology , Monocytes/virology , NF-kappa B/physiology , Receptors, Complement 3b/physiology , Virus Replication , Biological Transport , Cell Nucleus/physiology , Cells, Cultured , HIV Infections/immunology , Humans , Monocytes/physiology
15.
Clin Exp Immunol ; 92(1): 106-13, 1993 Apr.
Article in English | MEDLINE | ID: mdl-7682158

ABSTRACT

Peripheral blood and tissue mononuclear phagocytes serve as major viral reservoirs in HIV-infected individuals. We investigated the role of complement receptors CR1 (CD35) and CR3 (CD11b/CD18) in mediating productive infection with complement-opsonized HIV-1 and HIV-2 of cultured normal human peripheral blood monocytes, the promonocytic cell line THP-1, the monocytic cell line Mono Mac 6 and the glial cell line U251-MG. Cells were infected with the HTLV-IIIB strain of HIV-1 or the LAV-2 strain of HIV-2 that had been preopsonized with fresh human normal HIV seronegative serum. Productive infection was assessed by syncytia formation, the MTT cytotoxicity assay and/or release of p24 antigen in culture supernatants. Using suboptimal amounts of virus to infect the cells, we observed a higher and earlier productive infection of the cells with complement-opsonized HIV than with unopsonized virus. The enhancing effect of complement was totally suppressed by blocking CR1 or CR3 function with F(ab)'2 fragments of anti-receptor MoAbs; while blocking of the LFA-1 antigen had no effect. The infection of monocytic cells with complement-opsonized virus occurred independently of CD4 since it was not inhibited by F(ab)'2 fragments of a MoAb against the gp120 binding site of CD4 and since infection also occurred with Mono Mac 6 and U251-MG cells, which lack expression of the CD4 antigen and of CD4 mRNA. These observations suggest that complement may mediate productive infection of cells of the monocytic lineage with 'lymphocytotropic' HIV strains independently of CD4.


Subject(s)
Complement System Proteins/immunology , HIV/immunology , Macrophage-1 Antigen/immunology , Monocytes/microbiology , Receptors, Complement 3b/immunology , Antigens, CD/biosynthesis , Blotting, Northern , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Line , Cells, Cultured , Clone Cells , Flow Cytometry , HIV/physiology , Humans , Immunophenotyping , Opsonin Proteins/immunology , RNA/analysis
17.
AIDS Res Hum Retroviruses ; 9(3): 229-33, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8471312

ABSTRACT

The ability of the gp160 envelope glycoprotein of HIV-1 to activate human complement and to bind C3 fragments was investigated by incubating mammalian-derived recombinant gp160 with seronegative serum and by quantitating the binding of C3b/iC3b to the protein using a biotinylated monoclonal antibody directed against a neoepitope expressed by cleaved human C3. Recombinant gp160 activated complement in a dose- and time-dependent fashion. Complement activation occurred through the classical pathway, independently of antibodies, and required C1q. Binding of anti-HIV IgG to rgp160 prior to exposure of the envelope glycoprotein to serum resulted in enhanced complement activation. Complexes of rgp120 with anti-HIV IgG also cleaved C3 in serum, resulting in deposition of C3b on gp120. These results provide a basis for C3-mediated facilitation of viral entry into target cells expressing receptors for fragments of human C3.


Subject(s)
Complement Activation , Gene Products, env/immunology , HIV-1/immunology , Protein Precursors/immunology , Antibody Specificity , Complement C3/immunology , Complement Pathway, Classical , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160 , Humans , Kinetics
SELECTION OF CITATIONS
SEARCH DETAIL