Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 791
Filter
1.
Mol Biol Cell ; 11(9): 3169-76, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10982408

ABSTRACT

Cell fusion is a central phenomenon during the immune response that leads to formation of large elements called multinucleated giant cells (MGCs) of common occurrence at sites of granulomatous inflammation. We have previously reported on the involvement in this event of a novel receptor expressed to high level by mononuclear phagocytes, the purinergic P2X(7) receptor. Herein, we show that blockade of this receptor by a specific monoclonal antibody prevents fusion in vitro. In contrast, cell fusion is stimulated by addition of enzymes that destroy extracellular ATP (i.e., apyrase or hexokinase). Experiments performed with phagocytes selected for high (P2X(7) hyper) or low (P2X(7) hypo) P2X(7) expression show that fusion only occurs between P2X(7) hyper/P2X(7) hyper and not between P2X(7) hyper/P2X(7) hypo or P2X(7) hypo/P2X(7) hypo. During MGCs formation we detected activation of caspase 3, an enzyme that is powerfully stimulated by P2X(7). Finally, we observed that during MGCs formation, the P2X(7) receptor is preferentially localized at sites of cell-to-cell contact. These findings support the hypothesis originally put forward by our group that the P2X(7) receptor participates in multinucleated giant cell formation.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Fusion/physiology , Dendritic Cells/physiology , Giant Cells/cytology , Receptors, Purinergic P2/physiology , Animals , Antibodies, Monoclonal/pharmacology , Apyrase/metabolism , Caspase 3 , Caspases/metabolism , Cell Fusion/drug effects , Cell Line , Dendritic Cells/cytology , Giant Cells/physiology , Hexokinase/metabolism , Macrophages , Mice , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X7
2.
J Infect Dis ; 182(2): 607-10, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915097

ABSTRACT

FP-21399 is a bis(disulfonaphthalene) derivative that prevents human immunodeficiency virus (HIV) infection of uninfected cells by blocking entry of the virus. FP-21399 shows an affinity for lymph nodes. In this phase I study, FP-21399 was administered intravenously over 1 h as a single dose (0.9, 1.7, 2.8, and 4.2 mg/kg) or as a once-weekly infusion (1, 2, and 3 mg/kg) for 4 consecutive weeks to 34 HIV-1 infected patients with CD4(+) cell counts of 50-400 cells/microL. Concomitant antiretroviral therapy was permitted but not required. The most frequent adverse events involved the transient, dose-dependent appearance of drug- or metabolite-related color in the urine and skin. Plasma drug levels were linear with dose. The drug was cleared, with an elimination half-life of 4 h and a terminal half-life of 1.5-2 days; the terminal half-life represented redistribution and clearance from tissues. FP-21399 administered weekly for 4 weeks was well tolerated. Further studies are necessary to define the role of this fusion inhibitor in the treatment of HIV infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Cell Fusion/drug effects , Chlorobenzenes/therapeutic use , HIV Infections/drug therapy , Naphthalenes/therapeutic use , Adult , Anti-HIV Agents/pharmacokinetics , Chlorobenzenes/pharmacokinetics , Female , Humans , Male , Middle Aged , Naphthalenes/pharmacokinetics
3.
Biochem Biophys Res Commun ; 272(2): 351-6, 2000 Jun 07.
Article in English | MEDLINE | ID: mdl-10833417

ABSTRACT

To test the anti-human immunodeficiency virus type-1 (HIV-1) activity of 3,6,9,12-tetraazatetradecane-1,14-diylbis(zinc dithiocarbamate)-S,S'-dioxide (cyclic zinc-dithiocarbamate-S, S'-dioxide), MAGI and MAGIC-5 cells were used; the former express CXCR4 and the latter express both CXCR4 and CCR5, which are HIV-1 coreceptors. The compound markedly inhibited HIV-1 X4 (CXCR4-using) viral replication in both MAGI and MAGIC-5 cells. On the other hand, the replication of HIV-1 R5X4 (both CXCR4-and CCR5-using) in MAGI cells but not MAGIC-5 cells was inhibited by the compound. The compound was found to specifically inhibit HIV-1 (X4) envelope-mediated cell-to-cell fusion, binding of anti-CXCR4 monoclonal antibody (12G5) to CXCR4 expressed on the surface of cells, and calcium flux induced by stromal-derived factor-1alpha (SDF-1alpha) bound to CXCR4. The results suggest that the compound inhibited CXCR4-mediated HIV-1 infection by influencing to the HIV-1 coreceptor activity of CXCR4.


Subject(s)
Cyclic S-Oxides/pharmacology , HIV-1/drug effects , HIV-1/metabolism , Organometallic Compounds/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Calcium/metabolism , Cell Fusion/drug effects , Cell Line , Chemokine CCL5/pharmacology , Chemokine CXCL12 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/pharmacology , Cyclic S-Oxides/chemistry , Cytopathogenic Effect, Viral/drug effects , DNA, Viral/analysis , DNA, Viral/genetics , Flow Cytometry , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/pathology , Giant Cells/virology , HIV-1/genetics , HIV-1/physiology , Humans , Inhibitory Concentration 50 , Organometallic Compounds/chemistry , Proviruses/drug effects , Proviruses/genetics , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology
4.
FEBS Lett ; 474(2-3): 246-51, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10838094

ABSTRACT

We examined fusion mediated by the human immunodeficiency virus type 1 (HIV-1) and type 2 (HIV-2) envelope glycoproteins under various experimental conditions. Incubation of HeLa cells expressing HIV-2(ROD) and HIV-2(SBL/ISY) envelope glycoproteins with HeLa-CD4 target cells resulted in fusion at temperatures >/=25 degrees C whereas fusion with cells expressing HIV-1(Lai) occurred only at >/=31 degrees C. HIV-2 envelope glycoprotein-mediated fusion proceeded in the absence of Ca(2+) in the culture medium, whereas HIV-1 fusion required Ca(2+) ions for fusion. In contrast to HIV-2 envelope glycoprotein fusion, incubations in the presence of the 0.5 microM cytochalasin B completely inhibited HIV-1 envelope glycoprotein-mediated fusion. Our results suggest that in contrast to HIV-2, HIV-1 fusion is dependent on dynamic processes in the target membrane.


Subject(s)
Calcium/pharmacology , Cytochalasin B/pharmacology , Gene Products, env/metabolism , HIV-1/physiology , HIV-2/physiology , Actins/metabolism , Biopolymers/metabolism , CD4 Antigens/metabolism , Cell Fusion/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Giant Cells/cytology , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/virology , HIV-1/classification , HIV-1/drug effects , HIV-2/classification , HIV-2/drug effects , HeLa Cells , Humans , Temperature , Time Factors
5.
J Virol ; 74(12): 5629-38, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10823870

ABSTRACT

Using a recently developed model for in vitro generation of pp65-positive polymorphonuclear leukocytes (PMNLs), we demonstrated that PMNLs from immunocompetent subjects may harbor both infectious human cytomegalovirus (HCMV) and viral products (pp65, p72, DNA, and immediate-early [IE] and pp67 late mRNAs) as early as 60 min after coculture with human umbilical vein endothelial cells (HUVEC) or human embryonic lung fibroblasts (HELF) infected with a clinical HCMV isolate (VR6110) or other wild-type strains. The number of PMNLs positive for each viral parameter increased with coculture time. Using HELF infected with laboratory-adapted HCMV strains, only very small amounts of viral DNA and IE and late mRNAs were detected in PMNLs. A cellular mRNA, the vascular cell adhesion molecule-1 mRNA, which is abundantly present in both infected and uninfected HUVEC, was detected in much larger amounts in PMNLs cocultured with VR6110-infected cells than in controls. Coculture of PMNLs with VR6110-infected permissive cells in the presence or absence of RNA, protein, and viral DNA synthesis inhibitors showed that only IE genes were transcribed in PMNLs during coculture. Synthesis of IE transcripts in PMNLs was also supported by the finding that only the copy number of IE mRNA (and not the DNA or the pp67 mRNA) per infected PMNL increased markedly with time, and the pp67 to IE mRNA copy number ratio changed from greater than 10 in infected HUVEC to less than 1 in cocultured PMNLs. Fluorescent probe transfer experiments and electron microscopy studies indicated that transfer of infectious virus and viral products from infected cells to PMNLs is likely to be mediated by microfusion events induced by wild-type strains only. In addition, HCMV pp65 and p72 were both shown to localize in the nucleus of the same PMNLs by double immunostaining. Two different mechanisms may explain the virus presence in PMNLs: (i) one major mechanism consists of transitory microfusion events (induced by wild-type strains only) of HUVEC or HELF and PMNLs with transfer of viable virus and biologically active viral material to PMNLs; and (ii) one minor mechanism, i.e., endocytosis, occurs with both wild-type and laboratory strains and leads to the acquisition of very small amounts of viral nucleic acids. In conclusion, HCMV replicates abortively in PMNLs, and wild-type strains and their products (as well as cellular metabolites and fluorescent dyes) are transferred to PMNLs, thus providing evidence for a potential mechanism of HCMV dissemination in vivo.


Subject(s)
Cytomegalovirus/physiology , Endothelium, Vascular/virology , Neutrophils/virology , Virus Replication , Cell Fusion/drug effects , Cell Nucleus/metabolism , Cell Nucleus/virology , Cells, Cultured , Coculture Techniques , Cycloheximide/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus/ultrastructure , DNA, Viral/analysis , DNA, Viral/biosynthesis , DNA, Viral/genetics , Dactinomycin/pharmacology , Endocytosis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Foscarnet/pharmacology , Humans , Kinetics , Lung/cytology , Lung/embryology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/ultrastructure , Protein Biosynthesis/drug effects , RNA, Viral/analysis , RNA, Viral/biosynthesis , RNA, Viral/genetics , Transcription, Genetic/drug effects , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/genetics , Viral Load , Viral Proteins/analysis , Viral Proteins/biosynthesis , Virus Replication/drug effects
6.
Biochem Biophys Res Commun ; 271(3): 630-4, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10814513

ABSTRACT

SPARC (secreted protein acidic and rich in cysteine) is an extracellular Ca(2+)-binding glycoprotein associated with the morphogenesis and remodeling of various tissues. Here, involvement of SPARC in the myogenesis of skeletal myoblasts was investigated in vitro. First, the differential expression of SPARC mRNA during the myogenesis was initially identified by a differential display reverse transcription (DDRT)-PCR method. The expression of the SPARC gene was significantly up-regulated during the differentiation of C2C12 mouse myoblasts. Second, the treatment with anti-SPARC antibody almost completely prevented the differentiation of myoblasts. Third, the treatment with EGTA, a Ca(2+) chelator that is known to inhibit the fusion of C2C12 myoblasts, reversibly inhibited the up-regulation of SPARC gene expression. On the other hand, the treatment with A23187, a Ca(2+) ionophore, rapidly and dramatically increased the level of SPARC transcript. Taken together, these results suggest that SPARC may play a critical role(s) in the morphological change of myoblasts, and that the expression of SPARC gene may be controlled by Ca(2+)-dependent pathway in myogenesis.


Subject(s)
Muscle, Skeletal/embryology , Osteonectin/metabolism , Animals , Antibodies/pharmacology , Calcimycin/pharmacology , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Cell Fusion/drug effects , Cell Line , Chick Embryo , Egtazic Acid/pharmacology , Gene Expression Regulation, Developmental/drug effects , Mice , Morphogenesis , Osteonectin/genetics , RNA, Messenger/metabolism
7.
In Vitro Cell Dev Biol Anim ; 36(2): 133-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10718370

ABSTRACT

Ouabain, a specific inhibitor of the sodium- and potassium-activated adenosine triphosphatase, causes reversible inhibition of the fusion of myoblasts to form myotubes. We further examined this observation to investigate whether control of Na/K-ATPase activity may normally contribute to the regulation of myogenesis. In control cultures, fusion was preceded by a small decrease in intracellular sodium concentration, but intracellular sodium and potassium increased significantly during fusion. Levels of ouabain that produce prolonged inhibition of fusion (400 microM) virtually eliminated sodium and potassium gradients. However, lower ouabain levels (10-100 microM) also produced significant changes in intracellular potassium and/or sodium along with little apparent decrease in the eventual extent of fusion. The effect of ouabain on protein synthesis was also examined. Low levels of ouabain (<50 microM) that did not affect myogenesis also did not affect incorporation of radiolabeled amino acids, while higher concentrations produced a decline in protein synthesis that paralleled decreases in the rate of myoblast fusion. Levels of metabolic labeling were reduced 90% in cultures treated with 400 microM ouabain. Inhibition of protein synthesis would prevent membrane remodeling required for fusion and other events in myogenesis. Thus, our results do not support any specific role for the sodium- and potassium-activated adenosine triphosphatase in regulating myogenesis.


Subject(s)
Enzyme Inhibitors/pharmacology , Muscles/cytology , Ouabain/pharmacology , Protein Biosynthesis , Animals , Cell Differentiation/drug effects , Cell Fusion/drug effects , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Mice , Proteins/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism
8.
FEBS Lett ; 467(1): 123-7, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10664469

ABSTRACT

For reconstruction or repair of damaged tissues, an artificially regulated switch from proliferation to differentiation would be of great advantage. To achieve conditional myogenesis, we expressed MyoD in mouse C3H 10T1/2 fibroblastic cells, using a gene regulation system based on the synthetic steroid RU 486. By stable co-transfection of a plasmid construct with the RU 486 dependent activator and an integrating inducible MyoD construct, a cell clone, designated 10T-RM, was obtained in which MyoD expression was stringently controlled by RU 486. 12 h after addition of 10 nM RU 486 to 10T-RM cells, saturation levels of MyoD mRNA were observed and >/=2 days later, mRNA for embryonal myosin heavy chain (MyHC(emb)) was abundant and mononucleated cells fused into myotubes.


Subject(s)
Fibroblasts/cytology , Fibroblasts/drug effects , Mifepristone/pharmacology , Muscles/cytology , Muscles/drug effects , Animals , Cell Differentiation/drug effects , Cell Fusion/drug effects , Cell Line , Cell Size/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Genes, Reporter/genetics , Mice , Mice, Inbred C3H , Muscle Development , Muscles/embryology , MyoD Protein/genetics , Myosin Heavy Chains/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Transfection
9.
Biochim Biophys Acta ; 1500(2): 227-40, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10657592

ABSTRACT

It has been shown that deletion of the chemokine receptor, CXCR4, causes disordered angiogenesis in mouse models. In the present studies, we examined the distribution and trafficking of CXCR4 in human endothelial cells, tested their responses to the CXCR4 ligand, SDF-1, and asked whether endothelial cell CXCR4 can serve as a cell surface receptor for the binding of viruses. The results show that CXCR4 is present on endothelial cells from coronary arteries, iliac arteries and umbilical veins (HUVEC), but expression was heterogeneous, with some cells expressing CXCR4 on their surface, while others did not. Addition of SDF-1 caused a rapid decrease in CXCR4 surface expression. It also caused CXCR4-mediated activation of MAPK, release of PGI(2), endothelial migration, and the formation of capillary-like structures by endothelial cells in culture. Co-culture of HUVEC with lymphoid cells that were chronically infected with a CD4-independent/CXCR4-tropic variant of HIV-2 resulted in the formation of multinucleated syncytia. Formation of the syncytia was inhibited by each of several different CXCR4 antibodies. Thus, our findings indicate: (1) that CXCR4 is widely expressed on human endothelial cells; (2) the CXCR4 ligand, SDF-1, can evoke a wide variety of responses from human endothelial cells; and (3) CXCR4 on endothelial cells can serve as a receptor for isolates of HIV that can utilize chemokine receptors in the absence of CD4.


Subject(s)
Endothelium, Vascular/metabolism , HIV-2/physiology , Receptors, CXCR4/physiology , Anti-HIV Agents/pharmacology , Calcium Signaling/drug effects , Capillaries/cytology , Cell Fusion/drug effects , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Chemotaxis/drug effects , Collagen , Coronary Vessels/cytology , Cytopathogenic Effect, Viral/drug effects , Down-Regulation , Drug Combinations , Endothelium, Vascular/drug effects , Endothelium, Vascular/virology , Epoprostenol/metabolism , Flow Cytometry , Gene Expression , Humans , Iliac Artery/cytology , Immunoenzyme Techniques , Laminin , MAP Kinase Signaling System/drug effects , Microscopy, Fluorescence , Morphogenesis/drug effects , Proteoglycans , Receptor Cross-Talk , Receptor, PAR-1 , Receptors, CXCR4/genetics , Receptors, Thrombin/physiology , Tetradecanoylphorbol Acetate/pharmacology , Umbilical Veins/cytology
10.
Toxicon ; 38(1): 139-44, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10669019

ABSTRACT

One mAb reactive with mutalysin-II, a hemorrhagic metalloproteinase isolated from Lachesis muta muta venom, was produced in mice immunized with L. m. muta venom. Indirect ELISA was employed to compare the antigenic cross-reactivity among the venoms from Bothrops snakes. The mAb anti-mutalysin-II efficiently neutralized the hemorrhagic effect of both mutalysin-II and L. m. muta crude venom. Furthermore, the mAb were cross-reactive with B. alternatus, B. atrox, B. itapetiningae, B. jararaca and B. neuwiedii and showed variable potencies in neutralizing the hemorrhagic activity of several bothropic venoms.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bothrops , Crotalid Venoms/antagonists & inhibitors , Crotalid Venoms/toxicity , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Metalloendopeptidases/antagonists & inhibitors , Viper Venoms/antagonists & inhibitors , Viper Venoms/toxicity , Viperidae , Animals , Cell Fusion/drug effects , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Male , Metalloendopeptidases/toxicity , Mice , Mice, Inbred BALB C , Spleen/cytology
11.
Cell Motil Cytoskeleton ; 45(1): 27-41, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10618164

ABSTRACT

Previous studies have shown that a monoclonal antibody, H-145, inhibits myotube formation of quail myoblasts transformed with a temperature-sensitive mutant of Rous sarcoma virus (QM-RSV cells) [Hyodo and Kim, 1994: Exp. Cell Res. 212:120-131]. The antigen recognized by H-145 (H-145 antigen), which is a glycoprotein with a molecular mass of about 116 kDa, is related to a step immediately before myoblast fusion. To determine the functional significance of H-145 antigen, we examined its dynamic state during myogenic differentiation of QM-RSV cells. H-145 antigen showed a unique and discrete distribution. In immature myotubes immediately after myoblast fusion, many ring-like structures of H-145 antigen appeared on the ventral surface of the cells, encircling the actin dots detected simultaneously by immunofluorescence and interference reflection microscopy. The core of the ring-like structures was filled with the termini of actin bundles, mainly formed by alpha-actin. Other cytoskeletal-associated proteins, such as vinculin and alpha-actinin, were also associated with these structures. The ring-like structures of H-145 antigen were observed only during a restricted period when myoblasts fused actively, suggesting their relationships to myotube formation and an early stage of myofibril formation. With maturation of the myotubes, most of the H-145 antigen became redistributed in linear arrays on the apical cell surface and was probably associated with the termini of actin bundles to organize myofibrils, suggesting that the antigen was also related to maturation of myotubes. Experiments using monoclonal antibodies against chick beta1 integrin showed that H-145 antigen is beta1 integrin or a very closely related derivation. Thus H-145 antigen (beta1 integrin) is possibly involved in both myoblast fusion and the myofibril organization in myotubes.


Subject(s)
Antigens, Surface/analysis , Cell Differentiation , Muscle, Skeletal/chemistry , Actins/analysis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Surface/metabolism , Cell Fusion/drug effects , Cell Line, Transformed , Fluorescent Antibody Technique , Integrin beta1/immunology , Integrin beta1/metabolism , Microscopy, Confocal , Muscle Fibers, Skeletal/chemistry , Muscle, Skeletal/cytology , Myofibrils/metabolism , Quail
12.
J Gen Virol ; 81(Pt 2): 441-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644843

ABSTRACT

Proteolytic cleavage of the fusion protein (F) is an important control mechanism of the biological activity of paramyxoviruses. The sequence R-R-H-K-R(112) at the cleavage site of the F protein of measles virus (MV) was altered by site-directed mutagenesis to R-N-H-N-R(112), which is not recognized by the ubiquitous cellular protease furin. When transiently expressed in cell cultures standard F protein was cleaved, whereas the mutant remained in the uncleaved form. Syncytium formation by the mutant that was analysed after coexpression with haemagglutinin protein depended on the presence of trypsin. Recombinant MV containing the mutation required trypsin activation for fusion and infectivity in cell culture. Intranasal infection of transgenic mice susceptible to MV infection (Ifnar(tm)-CD46Ge) resulted in a moderately productive infection and inflammation of the lung. In contrast to parental virus, intracerebral inoculation did not induce neural disease. The possible effects of the change in cleavage activation on tissue tropism and pathogenicity are discussed.


Subject(s)
Measles virus/genetics , Measles virus/pathogenicity , Trypsin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Fusion/drug effects , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Humans , Measles/etiology , Measles/pathology , Measles/virology , Measles virus/drug effects , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Recombination, Genetic , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/physiology , Virulence/drug effects , Virulence/genetics , Virulence/physiology
14.
AIDS Res Hum Retroviruses ; 15(18): 1667-72, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10606089

ABSTRACT

The infection of human cells by HIV-1 virus can be mimicked by a fusion process between cells expressing the HIV envelope protein (Env) and cells expressing both human CD4 (huCD4) and appropriate human chemokine receptors. In this study, a macrophage-tropic (M-tropic) HIV cell-cell fusion assay was established that utilized huCD4, human CCR5 (huCCR5), and HIV ADAgpl60 as fusion components and a Gal4/VP16-activated luciferase as a reporter system. By combining CHO cells expressing huCD4 and huCCR5 with CHO cells expressing HIV ADAgpl60, a 300-fold increase in luciferase activity could be elicited relative to control. No luciferase activity was detected when HXB2gpl60 (T-tropic) was used instead of ADAgpl60 (M-tropic) as the fusion partner in the assay. Addition of anti-huCD4 (RPA-T4) or anti-huCCR5 (2D7) monoclonal antibodies in the assay significantly inhibited the fusion event; in contrast, an anti-CXCR4 (12G5) monoclonal antibody had little effect, indicating that the fusion assay was huCD4 and huCCR5 dependent. The cell-cell fusion occurred in a time-dependent manner; the maximum luciferase activity was detected about 8 hr after mixing the cells. The fusion events could also be monitored by another reporter system in which Gal4/VP16 activated green fluorescent protein (GFP) was used as the reporter instead of luciferase. In combination with fluorescence microscopy, the GFP reporter system allowed visualization of the fusion events in real time. Compared with previously described HIV fusion models, this system has several advantages, including simplicity, sensitivity, and the ability to allow continuous monitoring of the HIV cell-cell fusion event. Finally, this cell-cell fusion system is easily adapted to study other HIV fusion events.


Subject(s)
CD4 Antigens/physiology , Cell Fusion , Genes, Reporter , HIV Envelope Protein gp160/physiology , Luciferases/genetics , Luminescent Proteins/genetics , Macrophages/virology , Receptors, CCR5/physiology , Animals , Antibodies, Monoclonal/pharmacology , CHO Cells , Cell Fusion/drug effects , Cricetinae , Cricetulus , Green Fluorescent Proteins , Humans , Luciferases/biosynthesis , Luminescent Proteins/biosynthesis , Microscopy, Fluorescence , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sensitivity and Specificity , Transfection
15.
Biophys J ; 77(4): 2035-45, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10512823

ABSTRACT

Hemifusion, the linkage of contacting lipid monolayers of two membranes before the opening of a fusion pore, is hypothesized to proceed through the formation of a stalk intermediate, a local and strongly bent connection between membranes. When the monolayers' propensity to bend does not support the stalk (e.g., as it is when lysophosphatidylcholine is added), hemifusion is inhibited. In contrast, short-chain alcohols, reported to affect monolayer bending in a manner similar to that of lysophosphatidylcholine, were here found to promote hemifusion between fluorescently labeled liposomes and planar lipid bilayers. Single hemifusion events were detected by fluorescence microscopy. Methanol or ethanol (1.2-1.6 w/w %) added to the same compartment of the planar bilayer chamber as liposomes caused a 5-50 times increase in the number of hemifusion events. Alcohol-induced hemifusion was inhibited by lysophosphatidylcholine. Promotion of membrane hemifusion by short-chain alcohol was also observed for cell-cell fusion mediated by influenza virus hemagglutinin (HA). Alcohol promoted a fusion stage subsequent to the low pH-dependent activation of HA. We propose that binding of short-chain alcohol to the surface of membranes promotes hemifusion by facilitating the transient breakage of the continuity of each of the contacting monolayers, which is required for their subsequent merger in the stalk intermediate.


Subject(s)
Alcohols/chemistry , Alcohols/metabolism , Cell Membrane/metabolism , Lipid Bilayers/metabolism , Liposomes/metabolism , Membrane Fusion , 1-Butanol/metabolism , 1-Butanol/pharmacology , 3T3 Cells , Alkanes/metabolism , Alkanes/pharmacology , Animals , Cell Fusion/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Cholesterol/metabolism , Cholesterol/pharmacology , Electric Conductivity , Erythrocytes/drug effects , Erythrocytes/metabolism , Ethanol/metabolism , Ethanol/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Hydrogen-Ion Concentration , Lipid Bilayers/chemistry , Liposomes/chemistry , Liposomes/drug effects , Lysophosphatidylcholines/metabolism , Lysophosphatidylcholines/pharmacology , Membrane Fusion/drug effects , Methanol/metabolism , Methanol/pharmacology , Mice , Microscopy, Fluorescence , Molecular Weight , Propylene Glycols/chemistry , Propylene Glycols/metabolism , Propylene Glycols/pharmacology
16.
J Cell Biol ; 146(4): 893-904, 1999 Aug 23.
Article in English | MEDLINE | ID: mdl-10459022

ABSTRACT

The role of transmembrane 4 superfamily (TM4SF) proteins during muscle cell fusion has not been investigated previously. Here we show that the appearance of TM4SF protein, CD9, and the formation of CD9-beta1 integrin complexes were both regulated in coordination with murine C2C12 myoblast cell differentiation. Also, anti-CD9 and anti-CD81 monoclonal antibodies substantially inhibited and delayed conversion of C2C12 cells to elongated myotubes, without affecting muscle-specific protein expression. Studies of the human myoblast-derived RD sarcoma cell line further demonstrated that TM4SF proteins have a role during muscle cell fusion. Ectopic expression of CD9 caused a four- to eightfold increase in RD cell syncytia formation, whereas anti-CD9 and anti-CD81 antibodies markedly delayed RD syncytia formation. Finally, anti-CD9 and anti-CD81 monoclonal antibodies triggered apoptotic degeneration of C2C12 cell myotubes after they were formed. In summary, TM4SF proteins such as CD9 and CD81 appear to promote muscle cell fusion and support myotube maintenance.


Subject(s)
Antigens, CD/metabolism , Membrane Glycoproteins , Membrane Proteins/metabolism , Muscle, Skeletal/cytology , Animals , Antibodies, Monoclonal , Antigens, CD/genetics , Antigens, CD/immunology , Apoptosis/drug effects , Cell Differentiation , Cell Fusion/drug effects , Cell Line , DNA Fragmentation/drug effects , Desmin/metabolism , Gene Expression Regulation , Giant Cells/cytology , Giant Cells/drug effects , Giant Cells/metabolism , Histocompatibility Antigens/metabolism , Humans , Integrin beta1/immunology , Integrin beta1/metabolism , Membrane Proteins/immunology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Precipitin Tests , Tetraspanin 28 , Tetraspanin 29 , Time Factors , Tumor Cells, Cultured
17.
J Med Chem ; 42(17): 3203-9, 1999 Aug 26.
Article in English | MEDLINE | ID: mdl-10464007

ABSTRACT

Recent X-ray crystallographic determination of the HIV-1 envelope glycoprotein gp41 core structure opened up a new avenue to discover antiviral agents for chemotherapy of HIV-1 infection and AIDS. We have undertaken a systematic study to search for anti-HIV-1 lead compounds targeted to gp41. Using molecular docking techniques to screen a database of 20 000 organic molecules, we found 16 compounds with the best fit for docking into the hydrophobic cavity within the gp41 core and with maximum possible interactions with the target site. Further testing of these compounds by an enzyme-linked immunosorbent assay and virus inhibition assays discerned two compounds (ADS-J1 and ADS-J2) having inhibitory activity at micromolar concentrations on the formation of the gp41 core structure and on HIV-1 infection. These two compounds will be used as leads to design more effective HIV-1 inhibitors targeted to the HIV-1 gp41 core structure.


Subject(s)
Anti-HIV Agents/chemical synthesis , HIV Envelope Protein gp41/chemistry , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Cell Fusion/drug effects , Cell Line , Enzyme-Linked Immunosorbent Assay , HIV Envelope Protein gp41/metabolism , HIV-1/drug effects , Humans , Models, Molecular , Molecular Conformation , Naphthalenesulfonates/chemistry , Structure-Activity Relationship , Triazines/chemistry
18.
Am J Physiol ; 277(2): C320-9, 1999 08.
Article in English | MEDLINE | ID: mdl-10444409

ABSTRACT

Skeletal muscle is often the site of tissue injury due to trauma, disease, developmental defects or surgery. Yet, to date, no effective treatment is available to stimulate the repair of skeletal muscle. We show that the kinetics and extent of muscle regeneration in vivo after trauma are greatly enhanced following systemic administration of curcumin, a pharmacological inhibitor of the transcription factor NF-kappaB. Biochemical and histological analyses indicate an effect of curcumin after only 4 days of daily intraperitoneal injection compared with controls that require >2 wk to restore normal tissue architecture. Curcumin can act directly on cultured muscle precursor cells to stimulate both cell proliferation and differentiation under appropriate conditions. Other pharmacological and genetic inhibitors of NF-kappaB also stimulate muscle differentiation in vitro. Inhibition of NF-kappaB-mediated transcription was confirmed using reporter gene assays. We conclude that NF-kappaB exerts a role in regulating myogenesis and that modulation of NF-kappaB activity within muscle tissue is beneficial for muscle repair. The striking effects of curcumin on myogenesis suggest therapeutic applications for treating muscle injuries.


Subject(s)
Curcumin/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , NF-kappa B/antagonists & inhibitors , Regeneration/drug effects , Animals , Cell Count/drug effects , Cell Differentiation/drug effects , Cell Fusion/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , NF-kappa B/physiology , Stem Cells/pathology , Wounds and Injuries/pathology , Wounds and Injuries/physiopathology
19.
Berl Munch Tierarztl Wochenschr ; 112(5): 186-91, 1999 Jun.
Article in German | MEDLINE | ID: mdl-10399406

ABSTRACT

Signal transfer between neurons and between neurons and muscle cells is mediated by the secretion of neurotransmitters. The axon of the presynaptic cell contains synaptic vesicles, the storage organelles for neurotransmitters. Arrival of an action potential causes calcium-influx into the axon and leads to fusion of synaptic vesicles with the presynaptic plasma membrane. Recently, the events between calcium-influx and membrane fusion were elucidated on a molecular level. The family of SNARE-proteins was identified as the key players in neurosecretion. They are located on synaptic vesicles (VAMP) or on the presynaptic plasma membrane (syntaxin, SNAP-25). Intimate protein-protein interactions between the SNARE-proteins are responsible for the attachment and merger of vesicle and the plasma membrane. Fusion is triggered by calcium-binding to synaptotagmin, another protein recently identified on synaptic vesicles. The molecular mechanism of the action of clostridial neurotoxins was also elucidated. Botulinum-as well as Tetanus toxins are proteases which cleave neuronal SNARE-proteins. This explains the long known inhibition of neurosecretion caused by these toxins. The proteolytic action of Tetanus- and Botulinum toxin occurs in different types of neurons, resulting in a stimulatory or inhibitory effect on muscle cells. This selective degradation of SNAREs explains the opposing clinical signs of tetanus (cramps) and botulismus (paralysis).


Subject(s)
Botulinum Toxins/pharmacology , Neurons/physiology , Neurotransmitter Agents/metabolism , Synaptic Vesicles/physiology , Tetanus Toxin/pharmacology , Vesicular Transport Proteins , Animals , Cell Fusion/drug effects , Cell Fusion/physiology , Membrane Proteins/physiology , Molecular Biology , Muscles/drug effects , Muscles/physiology , Nerve Tissue Proteins/physiology , Neurons/drug effects , Neurotoxins/pharmacology , SNARE Proteins , Synaptic Vesicles/drug effects
20.
Development ; 126(15): 3381-90, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10393117

ABSTRACT

Cell-cell fusion is a component of many different developmental processes, but little is known about how cell-cell fusion is regulated. Here we investigate the regulation of a stereotyped cell-cell fusion event that occurs among the endodermal precursor cells of the glossiphoniid leech Helobdella robusta. We find that this fusion event is regulated inductively by a cell that does not itself fuse. We also show that biochemical arrest (by microinjection with ricin A chain or ribonuclease A) of the inducer or either of the fusion partners prevents fusion, but only if the arrest is initiated during a critical period long before the time at which fusion normally occurs. If the arrest occurs after this critical period, fusion occurs on schedule. These results suggest that both fusion partners play active roles in the process and that neither the induction nor the fusion itself requires concomitant protein synthesis.


Subject(s)
Cell Fusion/physiology , Leeches/cytology , Leeches/embryology , Animals , Cell Fusion/drug effects , Endoderm/cytology , Giant Cells/cytology , Leeches/drug effects , Ribonuclease, Pancreatic/pharmacology , Ricin/pharmacology , Signal Transduction , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...