Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Gene Ther ; 15(12): 787-94, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18654612

ABSTRACT

Previous studies have shown that intravenously applied bacteria can accumulate in tumors and lead to sporadic tumor regression. Recently, systemic administration of attenuated Salmonella typhimurium was demonstrated to generate no significant side effects in humans, but also no antitumor responses. We report the enhanced antitumor activity in preclinical mouse cancer models of nonvirulent S. typhimurium engineered to synthesize the cytokine Interleukin-18 (IL-18). IL-18-producing bacteria (but not control bacteria) inhibit the growth of primary subcutaneous tumors as well as pulmonary metastases in immunocompetent mice challenged with syngeneic multidrug-resistant clones of murine carcinoma cell lines, without overt toxicity to normal tissues. Antitumor activity was associated with increased accumulation of T-lymphocytes and NK cells in tumors, and massive infiltration of granulocytes, as well as increased intra-tumoral production of several cytokines. In summary, these findings provide evidence of promising preclinical antitumor activity of IL-18-expressing, attenuated S. typhimurium, suggesting a novel strategy for cancer immunotherapy.


Subject(s)
Interleukin-18/genetics , Neoplasms/therapy , Salmonella/genetics , Animals , Cytokines/metabolism , Genetic Therapy , Genetic Vectors , Interleukin-18/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Models, Animal , Neoplasms/metabolism , Protein Engineering , Salmonella/metabolism , Tumor Cells, Cultured
2.
Cell Death Differ ; 10(2): 153-62, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12700643

ABSTRACT

During inflammatory bowel diseases, commitment of extravased polymorphonuclear leucocytes (PMN) to apoptosis is required for the resolution of inflammation. To investigate the effect of transepithelial migration on PMN apoptotic rates, PMN transepithelial migration was reproduced in vitro using T84 intestinal monolayers. Transepithelial migration was found to delay neutrophil apoptosis, and this survival effect correlated with a downregulation of the surface expression of Fas ligand (FasL) and with a decrease in both procaspases-3, and -8 mRNA and procaspases-3, -6, -7 and -8 protein levels. Moreover, neutrophil survival and FasL shedding mediated by transepithelial migration were abrogated by a broad-spectrum metalloproteinase inhibitor, BB-94. Although Erk1/2 and p38 MAPK were activated in transmigrated PMN, inhibition of these MAP kinases did not impair transmigration-induced PMN survival. Taken together, our results show that trans-epithelial migration induces the downregulation of proapoptotic proteins expression in transmigrated PMN, which results in their increased lifespan.


Subject(s)
Caspases/metabolism , Intestinal Mucosa/cytology , Neutrophils/cytology , Phenylalanine/analogs & derivatives , fas Receptor/metabolism , Apoptosis/immunology , Cell Survival , Down-Regulation , Enzyme Activation , Epithelial Cells/cytology , Humans , Intestinal Mucosa/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
3.
Infect Immun ; 69(8): 5001-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11447179

ABSTRACT

Helicobacter pylori has been shown to induce chronic active gastritis and peptic ulcer and may contribute to the development of duodenal ulcer. Previous studies have shown that H. pylori mediates apoptosis of gastric epithelial cells via a Fas-dependent pathway. However, evidence for the induction of such a mechanism in intestinal epithelial cells (IEC) by H. pylori infection has not been demonstrated yet. This study was performed (i) to ascertain that H. pylori can induce IEC apoptosis; (ii) to delineate the role of the cag pathogenicity island (PAI), cagE, and vacA gene products in this process; and (iii) to verify whether the Fas-dependent pathway is involved in this phenomenon. When T84 cells were exposed to VacA(+)/cag PAI(+) H. pylori strains (CCUG 17874 and 60190), they exhibited apoptosis hallmarks as assessed by morphological studies, as well as annexin V and 3,3'-dihexyloxacarbocyanine iodide staining. In contrast, few or no apoptotic features could be detected after incubation with an isogenic mutant of strain 60190 in which the cagE gene was disrupted (60190:C(-) strain) or with a VacA(-)/cag PAI(-) H. pylori strain (G21). In addition, activation of caspase-3 during infection with VacA(+)/cag PAI(+) H. pylori strains was inhibited by pretreatment of IEC with an antagonistic anti-Fas antibody (ZB4). Taken together, these findings indicate that H. pylori triggers apoptosis in IEC via a Fas-dependent pathway following a process that depends on the expression of the cag PAI.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Intestinal Mucosa/microbiology , Caspase 3 , Caspases/metabolism , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/microbiology , Flow Cytometry/methods , Helicobacter pylori/physiology , Humans , Intestinal Mucosa/pathology , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Signal Transduction , Tumor Cells, Cultured , fas Receptor/metabolism
4.
J Leukoc Biol ; 68(4): 522-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11037974

ABSTRACT

Recruitment of polymorphonuclear leukocytes (PMNL) is a hallmark of both urinary and digestive infections caused by Escherichia coli. Cytotoxic necrotizing factor 1 (CNF-1) is a toxin produced by uropathogenic E. coli strains that mediates its effects via the activation of small GTP-binding proteins. However, the role and the consequences of CNF-1 on PMNL physiology remain largely unknown. In this study, we provide evidence that CNF-1 dramatically affects the PMNL cytoskeleton architecture by inducing an increased content of F-actin. Furthermore, we demonstrate that CNF-1 increases functional features of PMNL, such as superoxide generation and adherence on epithelial T84 monolayers, but significantly decreases their phagocytic function. Our results suggest that CNF-1 may behave as a virulence factor in urinary or digestive infection by stimulating PMNL cytotoxicity as a result of its enhancing effect on their adherence to epithelial cells as well as the production of radical oxygen products. Moreover, the decreased phagocytosis of PMNL induced by CNF-1 likely facilitates growth of bacteria. In these conditions, CNF-1 would intervene in the initiation and in the perpetuation of the inflammatory process.


Subject(s)
Bacterial Toxins/pharmacology , Cytotoxins/pharmacology , Escherichia coli Proteins , Escherichia coli/pathogenicity , Neutrophils/drug effects , Phagocytosis/drug effects , Respiratory Burst/drug effects , Actins/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Epithelial Cells/cytology , Humans , Inflammation/chemically induced , Intestinal Mucosa/cytology , Macrophage-1 Antigen/metabolism , Neutrophils/microbiology , Neutrophils/physiology , Neutrophils/ultrastructure , Opsonin Proteins/pharmacology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Virulence , Zymosan/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/physiology
5.
J Cell Biol ; 150(6): 1479-88, 2000 Sep 18.
Article in English | MEDLINE | ID: mdl-10995451

ABSTRACT

Acute colitis is characterized by a large number of polymorphonuclear leukocytes (PMNLs) migrating across the columnar epithelium in response to inflammatory stimuli. Several of these inflammatory factors have been characterized as proapoptotic inducers for intestinal epithelial cells. Our aim was to elucidate the role of PMNL transmigration in the onset of intestinal epithelial cell apoptosis. We found that PMNL migration, in response to N-formyl-methionyl-leucyl-phenylalanine across monolayers of intestinal epithelial cells (T84), was associated with activation of caspase-2, -3, and -9 and poly(ADP-ribose) polymerase cleavage within epithelial cells. Moreover, dihydrocytochalasin B treatment of T84 cells induced apoptosis with similar characteristics. Although Fas and Fas ligand were expressed on T84 cells and PMNLs, treatment of epithelial cells with an antagonistic anti-Fas antibody failed to prevent apoptosis induced by migrating PMNLs. Owing to the F-actin reorganization accompanying PMNL transmigration, these findings indicate a direct relationship between PMNL migration and induction of apoptosis in epithelial cells. This apoptotic process appears to involve remodeling of the actin cytoskeleton of enterocytes independent of the Fas/Fas ligand pathway.


Subject(s)
Apoptosis/immunology , Cell Movement/immunology , Epithelial Cells/cytology , Intestinal Mucosa/cytology , Neutrophils/cytology , Actins/metabolism , Caspase 2 , Caspase 3 , Caspase 9 , Caspases/metabolism , Colitis/immunology , Colitis/physiopathology , Colonic Neoplasms , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Enzyme Precursors/metabolism , Epithelial Cells/enzymology , Fas Ligand Protein , Flow Cytometry , Humans , Intestinal Mucosa/immunology , Membrane Glycoproteins/metabolism , Microscopy, Immunoelectron , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Polymers/metabolism , Proteins/metabolism , Tumor Cells, Cultured , fas Receptor/metabolism
6.
Infect Immun ; 68(2): 449-55, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10639403

ABSTRACT

The functionality of polymorphonuclear leukocytes (PMNs) once they migrate into the digestive lumen is still ill defined. More specifically, phagocytic function and bactericidal action of PMNs after transepithelial migration have not received much attention. The aim of the present study is to compare PMN behavior before and after transepithelial migration, in particular (i) phagocytosis and bactericidal activity; (ii) expression of surface molecules, particularly those involved in phagocytosis; and (iii) apoptosis. Cultured human intestinal epithelial T84 cell monolayers were used. The effect of transepithelial migration on phagocytosis was evaluated by immunofluorescence and electron microscopy and by flow cytometric assessment of the engulfment of a strain of Escherichia coli transfected with the green fluorescent protein. Superoxide production by PMNs was investigated by luminol-mediated chemiluminescence. Expression of various surface molecules on PMNs was evaluated by flow cytometry, while PMN apoptosis was assayed by morphologic changes and DNA fragmentation. E. coli phagocytosis by the PMNs was markedly increased after transepithelial migration without modification of superoxide production. CD11b/CD18 and CD47 expression was increased upon PMN transmigration, whereas CD16 expression was decreased and CD29, CD46, CD49e, CD49f, CD55, CD59, CD61, CD95 levels remained unchanged. Apoptosis in transmigrated PMNs was slightly advanced and was observed after 12 h compared to 16 h for nontransmigrated PMNs. In conclusion, the phagocytic capacity of the PMNs is augmented after transepithelial migration, with a dramatic increase in the level of CD11b/CD18 and preservation of the superoxide production. These results suggest a higher bactericidal activity of the PMNs once they have translocated into the digestive lumen.


Subject(s)
Escherichia coli/immunology , Intestinal Mucosa/microbiology , Neutrophils/immunology , Phagocytosis , Antigens, CD/analysis , Apoptosis , CD18 Antigens/analysis , CD47 Antigen , Carrier Proteins/analysis , Cell Movement , Colonic Neoplasms/microbiology , Humans , Macrophage-1 Antigen/analysis , Neutrophils/metabolism , Reactive Oxygen Species , Tumor Cells, Cultured
7.
Eur Cytokine Netw ; 10(2): 227-36, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10400829

ABSTRACT

The role of the polymorphonuclear leukocyte (PMN) cytoskeleton during the transmigration across colonic epithelial cells is not very well understood. In order to study the role of different components of the PMN cytoskeleton during transepithelial migration across a colonic epithelial cell monolayer (T84), PMN were preincubated with drugs affecting either the actin cytoskeleton (cytochalasin B, iota toxin of Clostridium perfringens, and phalloidin) or the microtubules (colchicine and taxol). The role of PMN myosin during transepithelial migration was investigated using the inhibitor 2,3-butanedione monoxime (BDM) and DC3B toxin. PMN intracellular Ca2+, during neutrophil adhesion and translocation across the epithelium, was assessed by the Ca2+ chelator 1, 2bis-(2-aminophenoxy)-ethane-N,N,N', N'-tetra-acetic acid tetrakis (acetoxymethyl) ester (BAPTA-AM). Transmigration of PMN was initiated by applying either interleukin-8 or formyl-met-leu-phe (fMLP). While colchicine and taxol preexposure did not influence PMN transepithelial migration, treatment with cytochalasin B, iota toxin, phalloidin, BDM, DC3B toxin and BAPTA-AM greatly diminished migration of PMN across T84 monolayers. Similarly, cell-cell contacts established between PMN and epithelial cells during the transmigration were diminished after treatment of PMN with iota toxin or cytochalasin B. These data show that the neutrophil actin cytokeleton and myosin, but not the microtubules, evoke a Ca2+ -dependent motility that facilitates migration across the colonic epithelial barrier.


Subject(s)
ADP Ribose Transferases , Actins/physiology , Chemotaxis, Leukocyte/drug effects , Interleukin-8/pharmacology , Intestinal Mucosa/cytology , Microtubules/physiology , Myosins/physiology , Neutrophils/physiology , Bacterial Toxins/pharmacology , Calcium/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Colchicine/pharmacology , Colonic Neoplasms/pathology , Cytochalasin B/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Diacetyl/analogs & derivatives , Diacetyl/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Epithelial Cells/cytology , Humans , Microtubules/drug effects , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/ultrastructure , Paclitaxel/pharmacology , Phalloidine/pharmacology , Tumor Cells, Cultured
8.
Infect Immun ; 66(6): 2494-500, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9596707

ABSTRACT

Cytotoxic necrotizing factor type 1 (CNF1), a 110-kDa toxin-like protein from pathogenic Escherichia coli strains, induces an actin cytoskeleton reorganization consisting of the formation of prominent stress fibers by permanent activation of the small GTP-binding protein Rho. Since p21Rho regulates tight-junction permeability and perijunctional actin reorganization in epithelial intestinal cells (A. Nusrat, M. Giry, J. R. Turner, S. P. Colgan, C. A. Parkos, E. Lemichez, P. Boquet, and J. L. Madara, Proc. Natl. Acad. Sci. USA 92:10629-10633, 1995), we used polarized T84 epithelial intestinal cell monolayers to examine whether CNF1 could affect microvillus structure, transepithelial resistance, and polymorphonuclear leukocyte (PMN) transmigration. Incubation of T84 cells with CNF1 did not influence transepithelial resistance, suggesting that barrier function and surface polarity were not affected by the toxin. However, CNF1 effaced intestinal cell microvilli and induced a strong decrease of PMN transepithelial migration in either the luminal-to-basolateral or the basolateral-to-luminal direction. CNF1 could thus be a virulence factor exhibiting a new type of combined activity consisting of effacing of microvilli and occlusion of the epithelial barrier to PMNs. Attenuated transepithelial migration of PMNs could result in the enhanced growth and protection of luminal bacteria.


Subject(s)
Bacterial Toxins/toxicity , Chemotaxis, Leukocyte/drug effects , Cytotoxins/toxicity , Escherichia coli Proteins , Intestinal Mucosa/drug effects , Microvilli/drug effects , Neutrophils/drug effects , Cell Polarity , Cytoskeleton/drug effects , Electric Impedance , Epithelial Cells/drug effects , Escherichia coli/pathogenicity , Intestinal Mucosa/cytology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/enzymology , Permeability/drug effects , Peroxidase/analysis
9.
J Leukoc Biol ; 63(3): 312-20, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9500518

ABSTRACT

A variety of bacterial enterocolitis in their active stages are characterized by the migration of polymorphonuclear leukocytes (PMNs) across epithelial surfaces. These mechanisms could explain some effects of enterotoxins observed in the intestinal mucosae. Here, using specific inhibitors, we investigated the potential role of CD10 (E.C. 3.4.24.11), present at the surface of human neutrophils, on formyl-Met-Leu-Phe (fMLP)-induced PMN migration across cultured monolayers of the human intestinal cell line T84. Transmigration of human neutrophils across T84 epithelial cells was observed for concentrations of fMLP as low as 10(-9) M, whereas maximal effect was achieved at 10(-7) M as determined by transepithelial resistances and PMN myeloperoxidase assays. RB25, a CD10 inhibitor, reduced by two orders of magnitude the concentration of fMLP required to obtain full neutrophil transmigration across T84 epithelial cell line. RB25 response was concentration dependent with half-maximal and maximal effect occurring at 10(-9) and 10(-7) M, respectively. These concentrations of RB25 corresponded exactly to the half-maximal and maximal inhibition of endopeptidase 24.11 at the neutrophil cell surface. However, the effect of CD10 inhibitors on PMN transmigration cannot be accounted for by a direct action on T84 epithelial cells, since these cells fail to express any detectable endopeptidase 24.11 activity. Moreover, blocking of CD10 enzymatic activity by various and selective inhibitors potentiated the effect of low concentrations of fMLP on PMN transmigration. Finally, RB25 failed to affect interleukin-8 (IL-8)-induced PMN transmigration across T84 epithelial cells, in agreement with the preference of CD10 for small peptidic substrates. Taken together, these results demonstrate that inhibition of CD10 significantly reduced the concentration of fMLP needed for eliciting transmigration of PMN across intestinal epithelia.


Subject(s)
Benzylidene Compounds/pharmacology , Chemotaxis, Leukocyte/physiology , Glycine/analogs & derivatives , Intestinal Mucosa/physiology , Neprilysin/antagonists & inhibitors , Neutrophils/physiology , Chemotaxis, Leukocyte/drug effects , Colonic Neoplasms , Drug Synergism , Enzyme Inhibitors/pharmacology , Glycine/pharmacology , Humans , In Vitro Techniques , Neprilysin/blood , Neutrophils/drug effects , Peroxidase/blood , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...