Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
AIDS Res Hum Retroviruses ; 17(16): 1489-500, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11709093

ABSTRACT

Cell motility is likely to play a pivotal role in HIV infection by promoting the dissemination of infected cells. On the basis of observations indicating an interaction between HIV-1 Gag and target cell filamentous actin, we hypothesized that these interactions would promote cell motility of HIV-infected cells. Indeed, we have found that HIV-1 infection enhances the chemotactic response of macrophages. To specifically investigate the significance of the interactions between Gag and cellular actin, we transfected NIH 3T3 fibroblasts and HeLa cells with a construct that permits the expression of HIV-1 Gag in the absence of any other viral protein. Fractionation experiments showed that Gag was present in cytoskeletal fraction containing long actin filaments and in a high-speed postcytoskeletal fraction with short actin filaments. We have also localized HIV-1 Gag to the lamellipodia of chemoattractant-stimulated cells. Significantly, the motility of Gag-expressing cells was enhanced in chemotaxis assays. In vitro mutagenesis experiments showed that HIV-1 Gag binds filamentous actin through the nucleocapsid domain (NC). An NC-green fluorescent protein fusion had the same cellular distribution as the complete protein, and its expression increased cell motility. These data suggest that interactions between HIV-1 Gag and actin in infected cells enhance cell motility. Ultimately this enhanced motility of infected cells could promote the dissemination of virus into the brain and other tissues.


Subject(s)
Chemotaxis/physiology , Cytoskeleton/metabolism , Gene Products, gag/metabolism , HIV-1/pathogenicity , Macrophages/physiology , Nucleocapsid/metabolism , 3T3 Cells/physiology , 3T3 Cells/virology , Actins/metabolism , Animals , Gene Products, gag/genetics , HIV-1/physiology , HeLa Cells/physiology , HeLa Cells/virology , Humans , Macrophages/virology , Mice , Monocytes/physiology , Monocytes/virology , Transfection
2.
J Virol ; 74(23): 10994-1000, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11069994

ABSTRACT

CD4(-) epithelial cells covering mucosal surfaces serve as the primary barrier to prevent human immunodeficiency virus type 1 (HIV-1) infection. We used HIV-1 vectors carrying the enhanced green fluorescent protein gene as a reporter gene to demonstrate that HIV-1 can infect some CD4(-) human epithelial cell lines with low but significant efficiencies. Importantly, HIV-1 infection of these cell lines is independent of HIV-1 envelope proteins. The Env-independent infection of CD4(-) cells by HIV-1 suggests an alternative pathway for HIV-1 transmission. Even on virions bearing Env, a neutralizing antibody directed against gp120 is incapable of neutralizing the infection of these cells, thus raising potential implications for HIV-1 vaccine development.


Subject(s)
CD4 Antigens/physiology , Gene Products, env/physiology , HIV-1/physiology , CD4 Antigens/analysis , Cell Line , Epithelial Cells/virology , Humans
3.
J Neuroimmunol ; 83(1-2): 133-8, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9610681

ABSTRACT

The pathogenesis of AIDS is a complex and prolonged process that is affected by a variety of cofactors, including the abuse of both intravenous and smoked (crack) cocaine. The exact mechanisms by which cocaine facilitates this disease are yet to be proven, but likely include a combination of increased risk due to cocaine-related social behaviours, a wide-ranging capacity for cocaine to suppress the immune system, and an effect of cocaine on the infectivity and replication of HIV. While sometimes contradictory, both human and animal studies document that cocaine alters the function of natural killer (NK) cells, T cells, neutrophils and macrophages, and alters the ability of these cells to secrete immunoregulatory cytokines. In addition to these effects on the immune system, cocaine also enhances the infectivity and/or replication of HIV when tested using human cells in vitro.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Crack Cocaine/immunology , Immune System/drug effects , Immune System/virology , Crack Cocaine/adverse effects , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/virology
4.
Am J Respir Crit Care Med ; 156(5): 1606-13, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9372683

ABSTRACT

Use of marijuana and cocaine is on the rise in the United States. Although pulmonary toxicity from these drugs has occasionally been reported, little is known about their effects on the lung microenvironment. We evaluated the function of alveolar macrophages (AMs) recovered from the lungs of nonsmokers and habitual smokers of either tobacco, marijuana, or crack cocaine. AMs recovered from marijuana smokers were deficient in their ability to phagocytose Staphylococcus aureus (p < 0.01). AMs from marijuana smokers and from cocaine users were also severely limited in their ability to kill both bacteria and tumor cells (p < 0.01). Studies using NG-monomethyl-L-arginine monoacetate, an inhibitor of nitric oxide synthase, suggest that AMs from nonsmokers and tobacco smokers were able to use nitric oxide as an antibacterial effector molecule, while AMs from smokers of marijuana and cocaine were not. Finally, AMs from marijuana smokers, but not from smokers of tobacco or cocaine, produced less than normal amounts of tumor necrosis factor-alpha, granulocyte-macrophage colony-stimulating factor, and interleukin-6 when stimulated in culture with lipopolysaccharide. In contrast, the production of transforming growth factor-beta, an immunosuppressive cytokine, was similar in all groups. These findings indicate that habitual exposure of the lung to either marijuana or cocaine impairs the function and/or cytokine production of AMs. The ultimate outcome of these effects may be an enhanced susceptibility to infectious disease, cancer, and AIDS.


Subject(s)
Crack Cocaine , Cytokines/biosynthesis , Macrophages, Alveolar/drug effects , Marijuana Smoking/immunology , Substance-Related Disorders/immunology , Adult , Arginine/analogs & derivatives , Arginine/pharmacology , Cytotoxicity, Immunologic , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Interleukin-6/biosynthesis , Macrophages, Alveolar/immunology , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Phagocytosis , Smoking/immunology , Staphylococcus aureus , Transforming Growth Factor beta/biosynthesis , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
5.
Chest ; 111(3): 698-705, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9118711

ABSTRACT

Crack cocaine has become a major drug of abuse in the United States and its use is associated with a broad spectrum of pulmonary complications. The present study was conducted to determine whether controlled in vivo administration of cocaine (inhaled or IV) alters the function of circulating inflammatory cells in a manner capable of contributing to acute lung injury. Subjects who regularly smoked crack cocaine were asked to abstain from illicit drug use for at least 8 h, and were then administered one of the following treatments on each of 4 study days: inhaled cocaine base (45 mg), inhaled placebo (4.5 mg cocaine base, a subphysiologic dose), IV cocaine HCl (0.35 to 0.50 mg/kg), or IV placebo (saline solution). Samples of blood were obtained from a peripheral venous catheter and blood cells were isolated before and 10 to 45 min after treatment. The administration of either cocaine base or cocaine HCl, but not their corresponding placebos, resulted in the activation of circulating polymorphonuclear neutrophils (PMNs). Exposure to cocaine in vivo enhanced the antibacterial activity of PMNs, as measured by their ability to kill Staphylococcus aureus. Antitumor activity, as measured in an antibody-dependent cell-mediated cytotoxicity assay, also increased following short-term administration of cocaine. Finally, short-term exposure to cocaine enhanced production of interleukin 8, a potent PMN chemoattractant and neutrophil-activating factor associated with both acute and chronic lung injury. These studies demonstrate that short-term in vivo exposure to cocaine activates the effector function and cytokine production of circulating PMNs. Therefore, it is possible that bursts of acute inflammatory activity resulting from crack use could contribute to lung injury.


Subject(s)
Crack Cocaine/adverse effects , Lung Diseases/chemically induced , Neutrophil Activation/drug effects , Administration, Inhalation , Adult , Antibody-Dependent Cell Cytotoxicity , Crack Cocaine/administration & dosage , Female , Humans , Injections, Intravenous , Interleukin-8/biosynthesis , Lung Diseases/immunology , Lung Diseases/physiopathology , Male , Middle Aged , Neutrophils/physiology , Phagocytosis , Substance-Related Disorders/immunology , Substance-Related Disorders/metabolism
6.
Cytometry ; 22(2): 103-10, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7587740

ABSTRACT

We investigated, via multicolor flow cytometry, the in vivo effects of colony-stimulating factors (CSFs) on cell size, frequencies, and expression of surface antigens on peripheral blood monocytes from melanoma patients treated concurrently with CSFs and tumor-specific monoclonal antibody (mAb) R24. Recombinant human macrophage colony-stimulating factor (rhM-CSF) increased cell size, relative percentages of monocytes, percentages of CD14+, HLA-DQ+, CD11b+, and CD16+ monocytes, and cell-surface expressions of HLA-DR and CD11b; rhM-CSF also up-regulated cell-surface expression of CD14 on CD14brightCD16- monocytes. Recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) increased cell size, percentages of CD14+, HLA-DQ+, and CD11b+ monocytes, and cell-surface expressions of HLA-DR, HLA-DQ, CD11b, and CD58. Relative percentages of monocytes and CD16+ cells and cell-surface expression of CD14 on CD14brightCD16- monocytes decreased. In addition, monocytes derived from patients treated with rhM-CSF showed functional activity when assayed in vitro for antibody-dependent cellular cytotoxicity (ADCC). During treatment and coincident with increased CD16 expression, monocytes derived from rhM-CSF patients had enhanced levels of cytotoxicity towards melanoma target cells compared to healthy controls and to patients treated with rhGM-CSF.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antigens, Surface/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Macrophage Colony-Stimulating Factor/therapeutic use , Melanoma/therapy , Monocytes/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/biosynthesis , Cell Size , Combined Modality Therapy , Flow Cytometry , Humans , Immunophenotyping , Melanoma/immunology , Melanoma/pathology , Recombinant Proteins/therapeutic use
7.
Stem Cells ; 12(6): 626-37, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7533581

ABSTRACT

There is increasing clinical interest focused on ex vivo manipulation and expansion of hematopoietic cells. In this study, we demonstrate that a simple combination of growth factors can expand progenitors to yield functional myeloid cells. Furthermore, this system can produce mature, functionally competent cells in the absence of fetal bovine serum (FBS), which will enhance the clinical utility of this approach. Hematopoietic progenitor cells obtained from normal bone marrow and from leukapheresis products were studied. The mononuclear fraction was enriched for CD34 cells using the Ceprate CD34 biotin kit (CellPro #LC34-1 or LC34-2). The selected cells were expanded for two weeks in Iscove's medium supplemented with 20% FBS and various combinations of interleukin-3 (IL-3), granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and interleukin -6 (IL-6) added either simultaneously or sequentially. The optimal combination of these factors identified for myeloid expansion was simultaneous addition of IL-3, SCF and G-CSF (at 50 ng/ml each), resulting in an average 773 +/- 133-fold expansion of nucleated cells (n = 5). When corrected for the purity of CD34 cells in the starting population, the mean fold expansion with IL-3, SCF and G-CSF was 2,265 +/- 729. A mean of 74.7 +/- 10.5% (n = 3) of the expanded cells was positive for CD11b; 86-91% (n = 2) of the cells were promyelocytes or more mature granulocytes. Functional assays demonstrated normal phagocytosis and intracellular killing of Staphylococcus aureus (S. aureus) by the expanded cell population. Studies performed using cells expanded in defined serum-free media demonstrated that fold expansion was decreased and that the cells produced were less mature and functionally less competent than cells expanded with FBS. The decreased expansion could be partially reversed, and the functionality almost completely restored by the addition of autologous plasma.


Subject(s)
Hematopoietic Stem Cells/cytology , Antigens, CD/metabolism , Antigens, CD34 , Cell Separation , Colony-Forming Units Assay , Culture Media , Cytotoxicity, Immunologic , Flow Cytometry , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocytes/cytology , Granulocytes/drug effects , Granulocytes/physiology , Growth Substances/administration & dosage , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/administration & dosage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , In Vitro Techniques , Interleukin-3/administration & dosage , Interleukin-6/administration & dosage , Phagocytosis , Stem Cell Factor
8.
Oral Microbiol Immunol ; 9(1): 55-9, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7478756

ABSTRACT

The effects of infection by human immunodeficiency virus (HIV) on phagocytic function towards Candida pseudotropicalis were tested using monocytotropic and nonmonocytotropic HIV strains on peripheral blood monocytes. There were no differences in either quantitative phagocytic activity or efficacy between the two infected and control mock-infected monocytes. Immunofluorescence for cytoplasmic p24 antigen and in situ hybridization for detection of HIV-specific RNA sequences revealed that very few cells (10(-4)) exhibit productive infection. Occasional cells with active infection as defined by microscopically detectable fluorescence labelling contained phagocytized yeasts.


Subject(s)
Candida/immunology , HIV Infections/immunology , HIV/immunology , Phagocytosis , AIDS-Related Opportunistic Infections/immunology , Candida/isolation & purification , Candidiasis/complications , Candidiasis/immunology , Cells, Cultured , HIV/isolation & purification , HIV Infections/complications , Humans , Macrophages/immunology , Macrophages/microbiology , Monocytes/immunology , Monocytes/microbiology
9.
Blood ; 82(11): 3279-82, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8241500

ABSTRACT

We previously showed the presence of receptors for granulocyte-macrophage colony-stimulating factor (GM-CSF) on tumor tissues and tumor cell lines that are derived from the neural crest. To determine whether normal neural cells express functional GM-CSF receptors, we isolated and analyzed primary rat brain cells, including microglia, astrocytes, and oligodendrocytes. Scatchard analysis of equilibrium binding of 125I-GM-CSF to primary rat oligodendrocytes showed an average of 1,110 GM-CSF binding sites per cell, with a kd of 20 pmol/L. In six separate experiments, no specific binding was detectable on the astrocyte population. Microglia were used in competitive binding experiments with oligodendrocytes, and addition of microglia did not increase the specific binding of labeled ligand to oligodendrocytes. In dose-response assays, we measured 3H-thymidine uptake in rat oligodendrocytes, microglia and control murine 32D cells stimulated with various concentrations of GM-CSF. Over concentration ranges of 0.025 to 1000 pmol/L, cell proliferation and peak 3H-thymidine incorporation was observed at approximately 30 pmol/L for both the control cells and the oligodendrocytes. However, the microglial cells did not proliferate in response to GM-CSF. These data indicate the presence of a functional receptor for GM-CSF on primary rat oligodendrocytes, and suggest that hematopoietic growth factors such as GM-CSF may play a role in nerve cell development, function, or response to injury.


Subject(s)
Oligodendroglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Animals , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis , Rats , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
10.
Blood ; 82(9): 2742-8, 1993 Nov 01.
Article in English | MEDLINE | ID: mdl-8219226

ABSTRACT

Tumor necrosis factor (TNF) may play a central role in proviral activation and release from latency in cells infected with the human immunodeficiency virus (HIV). We studied viral production and its relation to TNF in a HL-60 cell line (J22-HL-60) infected with a monocytotropic strain of HIV-1JR-FL. Viral production was stimulated to similar levels by TNF, 12-O-tetradecanoyl-phorbol-13-acetate (TPA), and 1,25-dihydroxyvitamin D3 (1,25[OH]2D3). Production of the virus was not suppressed by 3'-azido-3'-deoxythymidine (AZT), indicating that viral production was not caused by superinfection. Low concentrations of TNF (0.1 ng/mL) induced viral production with a short lag period of 8 hours, and this proviral activation was specifically suppressed by anti-TNF antibodies. However, induction of virus production by 1,25(OH)2D3 showed an extended lag period of 2 to 3 days. The effect of 1,25(OH)2D3 on virus production was also blocked by anti-TNF antibodies, which suggests the direct participation of TNF in this process. TNF accumulated in the culture supernatant of cells stimulated with 1,25(OH)2D3 with a kinetics consistent with its involvement in the action of 1,25(OH)2D3 on viral production. The J22-HL-60 cell line produced low levels of virus when cultured in the absence of an external stimulus; however, this basal viral production was suppressed greater than 80% in the presence of anti-TNF antibodies. Corresponding low levels of TNF were detected in the culture supernatants. Viral production decreased slowly with increasing passage of the cells, and no virus was detected in the supernatants of cells maintained in culture for several months. Concomitantly, TNF was no longer detected in the supernatant of these cells, which suggests that endogenous autocrine production of TNF drives viral production in the unstimulated cells. However, viral production was stimulated in these cells by low concentrations (0.1 ng/mL) of added TNF. These results argue for a central role for TNF in HIV proviral activation in chronically infected myeloid cells.


Subject(s)
HIV-1/growth & development , Leukemia, Promyelocytic, Acute/microbiology , Tumor Necrosis Factor-alpha/pharmacology , Virus Activation/drug effects , Calcitriol/pharmacology , Humans , Receptors, Tumor Necrosis Factor/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Zidovudine/pharmacology
11.
Br J Haematol ; 83(4): 545-53, 1993 Apr.
Article in English | MEDLINE | ID: mdl-7686031

ABSTRACT

We conducted experiments to determine the optimal conditions for colony-stimulating factor-enhanced neutrophil- and mononuclear phagocyte-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) using monoclonal antibodies to disialogangliosides expressed on neuroectodermal tumour target cells. Neutrophil ADCC was most effective at effector:target ratios of 100:1, with maximal cytotoxic responses to melanoma target cells generated by 3 h. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF) were the most potent stimulators of neutrophil ADCC, and enhanced ADCC activity was inhibited in the presence of antibody to Fc receptor type II (FcRII). GM-CSF and macrophage colony-stimulating factor (M-CSF) treatment of freshly isolated monocytes inhibited antibody-independent cytotoxicity but enhanced antibody-dependent responses. After 3 d in culture with CSF, 3-10-fold enhancement of ADCC against melanoma target cells was observed at effector:target cell ratios of 10:1. Greatest stimulation of macrophage ADCC was obtained when GM-CSF, M-CSF or interleukin 3 (IL-3) were used in conjunction with a secondary stimulus. Although gamma interferon (gamma-IFN) did not augment the cytotoxic capability of GM-CSF- and IL-3-stimulated macrophages, prominent cytotoxic enhancement was seen when M-CSF-stimulated macrophages were exposed to gamma-IFN. A chimaeric mouse/human monoclonal antibody was found to be equivalent in activity to the murine antibody in neutrophil ADCC; however, in macrophage ADCC assays with submaximal effector cell stimulation, the chimaeric antibody was associated with a two-fold greater response. These studies indicate that under specific conditions, CSFs capable of increasing the number and functional activity of mature myeloid effector cells enhance antibody-dependent cytotoxicity to neuroectodermal tumour target cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Colony-Stimulating Factors/immunology , Melanoma/immunology , Neuroblastoma/immunology , Dose-Response Relationship, Immunologic , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-3/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Tumor Cells, Cultured
12.
Dev Biol ; 151(2): 352-67, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1601172

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is one of a family of glycoprotein cytokines that have potent effects in stimulating the proliferation, maturation, and function of hematopoietic cells. Deriving its name from its ability to stimulate the formation of macroscopic colonies containing neutrophils, eosinophils, macrophages, or mixtures of these cell types, GM-CSF stimulates the proliferation and maturation of myeloid progenitors, as well as functionally activating mature neutrophils, eosinophils, and macrophages. As most of the effects observed using GM-CSF in vitro have been shown to occur in vivo either in animal models or in human subjects, it is important to consider that GM-CSF may also exert some biological effects on nonhematopoietic cells. In response to immunologic stimuli, immunologic surveillance cells and cells of the microenvironment are capable of producing GM-CSF. In vitro experiments indicate that GM-CSF production is tightly regulated. In that regard, GM-CSF is not present in measurable quantities in normal serum, but little is known about the in vivo process of GM-CSF production and regulation. The biologic capabilities of GM-CSF have triggered its widespread clinical use in situations where hematopoiesis is compromised. GM-CSF can act as a potent growth factor in vivo, increasing the number and enhancing the function of hematopoietic progenitors and mature cells. However, the precise in vivo effect that GM-CSF may have on normal and neoplastic cells of nonhematopoietic origin remains undefined. The full range of GM-CSF bioactivity is mediated following binding to its receptor. The presence of specific receptors for GM-CSF has been demonstrated in all responsive cells of hematopoietic lineage, as well as in nonhematopoietic cells, both responsive and unresponsive. In conclusion, a large body of work from a number of laboratories has defined the biology of GM-CSF. Currently available reagents and technology will provide additional insights into the biology of this molecule, thereby expanding our present definition and allowing us to explore the mechanisms regulating hematopoiesis.


Subject(s)
Cell Differentiation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Animals , Humans
13.
Blood ; 78(3): 609-15, 1991 Aug 01.
Article in English | MEDLINE | ID: mdl-1830497

ABSTRACT

Hematopoietic growth factor receptors are present on cells of normal nonhematopoietic tissues such as endothelium and placenta. We previously demonstrated functional human granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors on small cell carcinoma of the lung cell lines, and others have reported that certain solid tumor cell lines respond to GM-CSF in clonogenic assays. In the current study, we examine human melanoma cell lines and fresh specimens of melanoma to determine whether they have functional GM-CSF receptors. Scatchard analyses of 125I-GM-CSF equilibrium binding to melanoma cell lines showed a mean of 542 +/- 67 sites per cell with a kd of 0.72 +/- 0.14 nmol/L. Cross-linking studies in the melanoma cell line, M14, showed a major GM-CSF receptor species of 84,000 daltons. Under the conditions tested, the M14 cells did not have a proliferative response to GM-CSF in vitro, nor was any induction of primary response genes detected by Northern analysis in response to GM-CSF. Studies to determine internal translocation of the receptor-ligand complex indicated less than 10% of the 125I-GM-CSF internalized was specifically bound to receptors. Primary melanoma cells from five surgical specimens had GM-CSF receptors; Scatchard analysis was performed on one sample, showing 555 sites/cell with a kd of 0.23 nmol/L. These results indicate that human tumor cells may express a low-affinity GM-CSF receptor protein that localizes to the cell surface and binds ligand, but lacks functional components or accessory factors needed to transduce a signal.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Melanoma/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Cell Line , Cells, Cultured , Gene Expression/drug effects , Humans , Kinetics , Neoplasm Metastasis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Recombinant Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology
14.
Blood ; 77(8): 1699-705, 1991 Apr 15.
Article in English | MEDLINE | ID: mdl-1707694

ABSTRACT

To define the relationship between human immunodeficiency virus type 1 (HIV-1) infection in hematopoietic stem cells and virus production by their progeny, we performed kinetic studies infecting bone marrow (BM) stem cells and culturing them in the presence of hematopoietic growth factors. CD34-positive (CD34+), CD4-negative (CD4-) BM cells were isolated and infected in vitro with the monocytotropic HIV-1JR-FL strain or the laboratory-maintained HTLV-IIIB strain at a high multiplicity of infection. The cells were susceptible to productive infection only with HIV-1JR-FL, and virus production as measured by p24 protein release was markedly increased (more than fivefold) in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 (IL-3). Macrophage CSF (M-CSF) was less stimulatory and granulocyte CSF (G-CSF) had no effect on virus production. Virus production coincided with proliferation of mononuclear phagocytes but was not related to granulocytic proliferation in G-CSF-treated BM cultures. Although peak virus production from GM-CSF-treated macrophages occurred 2 to 3 weeks after infection, peak virus production in infected stem cells was observed 5 to 6 weeks after. Enhancement in virus production had a more rapid onset when CD34+/CD4- cells were cultured in the presence of both GM-CSF and IL-3 for 7 or 14 days. Under these conditions there was a 10-fold enhancement in virus production after 7 days of preincubation and a 50-fold enhancement after 14 days. These data indicate that while the stem cell compartment may be susceptible to infection with a monocytotropic HIV-1 strain, productive and sustained infection is realized only after macrophage differentiation. The lack of effect of G-CSF on virus production is likely because of the limited effect of this hematopoietin on mononuclear phagocyte generation and function.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HIV-1/physiology , Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/pharmacology , Virus Replication/drug effects , Antigens, CD/analysis , Bone Marrow/microbiology , Bone Marrow Cells , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , DNA Replication/drug effects , Drug Synergism , Flow Cytometry , HIV-1/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/microbiology , Humans , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Kinetics
15.
Blood ; 76(10): 1980-8, 1990 Nov 15.
Article in English | MEDLINE | ID: mdl-2173633

ABSTRACT

Monocytotropic human immunodeficiency virus type 1 (HIV-1) isolates from patients with acquired immunodeficiency syndrome (AIDS) infect mononuclear phagocytes as well as activated T cells, but do not usually infect immature human myeloid cell lines in vitro. The HL-60 promyelocytic/myeloblastic cell line and the promonocytic line, U937, were susceptible to productive infection by monocytotropic HIV-1 isolates (HIV-1JR-FL and HTLV-IIIBa-L) after treatment with retinoic acid, dimethyl sulfoxide, dibutyryl cAMP, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), or 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Virus production was only detected when these compounds were added before virus infection. Virus replication did not correlate with CD4 receptor expression because undifferentiated HL-60 cells express CD4 and the level of CD4 expression did not increase after differentiation in the presence of retinoic acid, 1,25(OH)2D3, or TPA. A mature monocytic cell line (THP-1) was capable of infection without pretreatment, and treatment with differentiating agents enhanced virus production. A chronically infected cell line (J-HL-60) was isolated after HIV-1JR-FL infection of HL-60 cells treated with retinoic acid. Virus production in this cell line was enhanced more than 10-fold after differentiation in the presence of 1,25(OH)2D3 or TPA. The majority of virus production by 1,25(OH)2D3-treated J-HL-60 cells was associated with the mature, adherent population. Molecular analysis of a cloned line of J-HL-60 showed integration of a single DNA provirus. These results suggest that cellular factors associated with precursor cell differentiation along the myelomonocytic pathway are required for optimal replication of monocytotropic HIV-1 strains in vitro.


Subject(s)
Bucladesine/pharmacology , Calcitriol/pharmacology , Dimethyl Sulfoxide/pharmacology , HIV-1 , Leukemia, Myeloid/microbiology , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , CD4 Antigens/genetics , CD4 Antigens/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , DNA, Viral/genetics , DNA, Viral/metabolism , Gene Expression , HIV Infections/complications , HIV-1/genetics , HIV-1/growth & development , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Thymidine/metabolism , Tritium , Virus Replication/drug effects , Virus Replication/physiology
16.
Proc Natl Acad Sci U S A ; 87(10): 3933-7, 1990 May.
Article in English | MEDLINE | ID: mdl-2187195

ABSTRACT

There is compelling clinical evidence for dysfunction of the mononuclear phagocyte system in patients with AIDS, which is believed due in part to loss of T-cell cooperativity. The direct consequences of human immunodeficiency virus infection on macrophage function are unknown. To address this question we infected normal human macrophages in vitro with a monocytotropic strain of human immunodeficiency virus and performed assays to quantify their extra- and intracellular killing ability. Human immunodeficiency virus-infected macrophages were significantly less effective than control cells in mediating antibody-dependent cell-mediated cytotoxicity against leukemic cell targets and intracellular killing of Candida pseudotropicalis. The functional defects were profound, related temporarily to active virus production by the macrophages, and could not be overcome by granulocyte-macrophage colony-stimulating factor. Treatment of macrophages with 3'-azido-3'-deoxythymidine (AZT) 6 days after infection caused a marked decrease in virus production and prevented development of the intracellular killing functional defect. The results suggest that early antiviral therapy may be useful in preventing or mitigating some virus-induced mononuclear phagocyte dysfunction.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , HIV/immunology , Macrophages/immunology , Monocytes/immunology , Cells, Cultured , Colony-Stimulating Factors/pharmacology , Endotoxins/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor , Growth Substances/pharmacology , Humans , Kinetics , Macrophages/microbiology , Monocytes/drug effects , Monocytes/microbiology
17.
Blood ; 75(4): 851-7, 1990 Feb 15.
Article in English | MEDLINE | ID: mdl-1689190

ABSTRACT

Human granulocyte colony-stimulating factor (G-CSF) is a regulatory glycoprotein that stimulates the production of neutrophilic granulocytes from committed hematopoietic progenitor cells both in vitro and in vivo. In this report, we show that biosynthetic (recombinant) human G-CSF enhances colony formation by normal human bone marrow and the human myeloid leukemic cell lines, HL-60 and KG-1, as well as nonhematopoietic small cell lung cancer lines, H128 and H69. G-CSF also modulates multiple differentiated functions of human neutrophils, including enhanced oxidative metabolism in response to f-Met-Leu-Phe (f-MLP), increased antibody-dependent cell-mediated cytotoxicity (ADCC), and augmented arachidonic acid release in response to ionophore and chemotactic agents. These effects are all maximal at a concentration of 100 to 500 pmol/L. Using 125I-labeled recombinant human G-CSF, high affinity binding sites were identified on human neutrophils, the myeloid leukemia cell lines KG-1 and HL-60, and the small cell carcinoma cell lines, H128 and H69. G-CSF receptor numbers ranged between 138 and 285 sites per cell with a kd of 77 to 140 pmol/L, consistent with the concentrations of G-CSF that elicit biologic responses in vitro. Decreased specific binding of 125l-G-CSF by human neutrophils was consistently observed in the presence of excess unlabeled human granulocyte-macrophage colony-stimulating factor (GM-CSF), suggesting competition or down modulation by GM-CSF of the G-CSF receptor.


Subject(s)
Carcinoma, Small Cell/pathology , Colony-Stimulating Factors/metabolism , Hematopoietic System/cytology , Lung Neoplasms/pathology , Receptors, Cell Surface/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects , Arachidonic Acids/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/ultrastructure , Cell Division/drug effects , Cell Line , Colony-Stimulating Factors/pharmacology , Granulocyte Colony-Stimulating Factor , Granulocyte-Macrophage Colony-Stimulating Factor , Growth Substances/pharmacology , Hematopoietic System/metabolism , Hematopoietic System/ultrastructure , Humans , Iodine Radioisotopes/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/ultrastructure , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, Cell Surface/analysis , Receptors, Cell Surface/drug effects , Receptors, Granulocyte Colony-Stimulating Factor , Recombinant Proteins/metabolism , Superoxides/metabolism
18.
Ciba Found Symp ; 148: 62-71; discussion 71-5, 1990.
Article in English | MEDLINE | ID: mdl-2180650

ABSTRACT

Colony-stimulating factors (CSFs) have important effects on mature myeloid cells in addition to their regulatory role in haemopoiesis. Exposure of neutrophils to granulocyte macrophage-CSF (GM-CSF) increases chemotaxis, phagocytosis and cytotoxicity and primes the cells for enhanced oxidative metabolism in response to stimuli, such as formylated oligopeptides derived from bacteria (f-Met-Leu-Phe) and endogenous activated complement components (C5a). GM-CSF induces time-dependent changes in neutrophil f-Met-Leu-Phe receptor number and affinity that correspond to changes in functional activity. The neutrophil IgA Fc receptor is also modulated by GM-CSF such that it develops a high affinity state and transduces a phagocytic signal. The ability to regulate the number and activity of mature myeloid effector cells in vivo establishes unique therapeutic opportunities in the area of infectious disease, cancer treatment, bone marrow transplantation and augmentation of host defence in immunodeficient patients.


Subject(s)
Colony-Stimulating Factors/pharmacology , Growth Substances/pharmacology , Interleukin-3/pharmacology , Neutrophils/drug effects , Animals , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Macrophage Colony-Stimulating Factor , Neutrophils/physiology
20.
Blood ; 74(5): 1673-7, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2477084

ABSTRACT

Although the control of retroviral disease in animal systems often involves antibody-dependent cell-mediated cytotoxicity (ADCC), the role of cytotoxic function in human retroviral disorders is uncertain. The ability of the neutrophil to kill HIV-infected targets directed by antiviral antibody was examined. Neutrophils from patients with AIDS killed HIV-infected MOLT-3A cells in a manner equivalent to neutrophils obtained from normal volunteers. Both granulocyte- and granulocyte-macrophage colony-stimulating factors (G-CSF and GM-CSF) markedly augmented the cytotoxic function. Studies done with fractionated human antisera revealed that ADCC to HIV-infected cells was mediated only by antibody to the env glycoprotein. ADCC in this system was not dependent on oxidative metabolism because neutrophils from patients with chronic granulomatous disease (CGD) were capable of CSF-augmented cytotoxicity. Although ADCC can be mediated by various classes of lymphocytes and mononuclear phagocytes, such cells may be infected by HIV. Because the neutrophil apparently is not productively infected by the virus, it is an ideal cell to focus on with regard to cytotoxic function in AIDS patients. The findings regarding neutrophil ADCC in AIDS are clinically relevant because the availability of CSFs now permits therapeutic regulation of neutrophils in AIDS patients, and presumably natural antibody may be useful in targeting HIV-infected cells for neutrophil cytotoxicity in vivo.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Colony-Stimulating Factors/pharmacology , Granulomatous Disease, Chronic/blood , Growth Substances/pharmacology , HIV/growth & development , Neutrophils/immunology , Cell Line , Granulocyte Colony-Stimulating Factor , Granulocyte-Macrophage Colony-Stimulating Factor , Granulocytes/physiology , Granulomatous Disease, Chronic/immunology , HIV/drug effects , Humans , In Vitro Techniques , Neutrophils/drug effects , Recombinant Proteins/pharmacology , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...