Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 6(6): e1000964, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585570

ABSTRACT

Entry of Staphylococcus aureus into the bloodstream can lead to metastatic abscess formation and infective endocarditis. Crucial to the development of both these conditions is the interaction of S. aureus with endothelial cells. In vivo and in vitro studies have shown that the staphylococcal invasin FnBPA triggers bacterial invasion of endothelial cells via a process that involves fibronectin (Fn) bridging to alpha(5)beta(1) integrins. The Fn-binding region of FnBPA usually contains 11 non-identical repeats (FnBRs) with differing affinities for Fn, which facilitate the binding of multiple Fn molecules and may promote integrin clustering. We thus hypothesized that multiple repeats are necessary to trigger the invasion of endothelial cells by S. aureus. To test this we constructed variants of fnbA containing various combinations of FnBRs. In vitro assays revealed that endothelial cell invasion can be facilitated by a single high-affinity, but not low-affinity FnBR. Studies using a nisin-inducible system that controlled surface expression of FnBPA revealed that variants encoding fewer FnBRs required higher levels of surface expression to mediate invasion. High expression levels of FnBPA bearing a single low affinity FnBR bound Fn but did not invade, suggesting that FnBPA affinity for Fn is crucial for triggering internalization. In addition, multiple FnBRs increased the speed of internalization, as did higher expression levels of FnBPA, without altering the uptake mechanism. The relevance of these findings to pathogenesis was demonstrated using a murine sepsis model, which showed that multiple FnBRs were required for virulence. In conclusion, multiple FnBRs within FnBPA facilitate efficient Fn adhesion, trigger rapid bacterial uptake and are required for pathogenesis.


Subject(s)
Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Sepsis/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/physiology , Virulence , Animals , Bacterial Adhesion , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/microbiology , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Sepsis/metabolism , Sepsis/pathology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Umbilical Veins/cytology , Umbilical Veins/microbiology
2.
Virulence ; 11(1): 1623-1639, 2020 12.
Article in English | MEDLINE | ID: mdl-33222653

ABSTRACT

Staphylococcus aureus is a leading cause of infections world-wide. Once this pathogen has reached the bloodstream, it can invade different parts of the human body by crossing the endothelial barrier. Infected endothelial cells may be lysed by bacterial products, but the bacteria may also persist intracellularly, where they are difficult to eradicate with antibiotics and cause relapses of infection. Our present study was aimed at investigating the fate of methicillin resistant S. aureus (MRSA) isolates of the USA300 lineage with different epidemiological origin inside endothelial cells. To this end, we established two in vitro infection models based on primary human umbilical vein endothelial cells (HUVEC), which mimic conditions of the endothelium when infection occurs. For comparison, the laboratory strain S. aureus HG001 was used. As shown by flow cytometry and fluorescence- or electron microscopy, differentiation of HUVEC into a cell barrier with cell-cell junctions sets limits to the rates of bacterial internalization, the numbers of internalized bacteria, the percentage of infected cells, and long-term intracellular bacterial survival. Clear strain-specific differences were observed with the HG001 strain infecting the highest numbers of HUVEC and displaying the longest intracellular persistence, whereas the MRSA strains reproduced faster intracellularly. Nonetheless, all internalized bacteria remained confined in membrane-enclosed LAMP-1-positive lysosomal or vacuolar compartments. Once internalized, the bacteria had a higher propensity to persist within the differentiated endothelial cell barrier, probably because internalization of lower numbers of bacteria was less toxic. Altogether, our findings imply that intact endothelial barriers are more likely to sustain persistent intracellular infection.


Subject(s)
Endothelial Cells/microbiology , Host-Pathogen Interactions , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcus aureus/pathogenicity , Cells, Cultured , Cytoplasm/microbiology , Flow Cytometry/methods , Humans , Microscopy, Electron/methods , Staphylococcus aureus/classification , Umbilical Veins/cytology , Umbilical Veins/microbiology
3.
J Clin Invest ; 91(4): 1301-9, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8473483

ABSTRACT

Marburg and Ebola virus, members of the family Filoviridae, cause a severe hemorrhagic disease in humans and primates. The disease is characterized as a pantropic virus infection often resulting in a fulminating shock associated with hemorrhage, and death. All known histological and pathophysiological parameters of the disease are not sufficient to explain the devastating symptoms. Previous studies suggested a nonspecific destruction of the endothelium as a possible mechanism. Concerning the important regulatory functions of the endothelium (blood pressure, anti-thrombogenicity, homeostasis), we examined Marburg virus replication in primary cultures of human endothelial cells and organ cultures of human umbilical cord veins. We show here that Marburg virus replicates in endothelial cells almost as well as in monkey kidney cells commonly used for virus propagation. Our data support the concept that the destruction of endothelial cells resulting from Marburg virus replication is a possible mechanism responsible for the hemorrhagic disease and the shock syndrome typical of this infection.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/microbiology , Hemorrhagic Fevers, Viral/etiology , Marburgvirus/physiology , Base Sequence , Cells, Cultured , Fluorescent Antibody Technique , Humans , Microscopy, Electron , Molecular Sequence Data , Umbilical Veins/cytology , Umbilical Veins/microbiology , Virus Replication
4.
Biochem Pharmacol ; 72(11): 1367-74, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16884694

ABSTRACT

Infection of endothelial cells by Listeria monocytogenes is an essential step in the pathogenesis of listeriosis. Small GTPases of the Rho family act as molecular switches in signal transduction. We tested the hypothesis that Rho GTPases contribute to the regulation of cytokine expression following L. monocytogenes infection. L. monocytogenes induced release of distinct CC and CXC, as well as Th1 and Th2 cytokines and growth factors by endothelial cells and activated RhoA and Rac1. Inhibition of Rac1 by inhibitor Nsc23766 reduced cytokine expression, and slightly yet significantly the uptake of bacteria. Blocking of Rho proteins by Clostridium difficile toxin B-10463 (TcdB) reduced Listeria-dependent cytokine expression, whereas activating Rho proteins by Escherichia coli CNF1 increased it. We analyzed regulation of IL-8 expression in more detail: Listeria-induced IL-8 release was reduced by inhibition of RhoA, Rac1 and Cdc42 (TcdB) or Rac1 while blocking of RhoA/B/C by Clostridium limosum C3 fusion toxin (C3FT) or Rho kinase by Y27632 reduced cytokine expression only slightly. Activation of RhoA, Rac1 and Cdc42 (CNF1), but not of RhoA alone (CNF(Y)), enhanced Listeria-dependent IL-8 release significantly. Furthermore, inhibition of RhoA, Rac1 and Cdc42 (TcdB) and Rac1 (Nsc23766), but not of RhoA (C3FT) reduced Listeria-related recruitment of NF-kappaB/p65 and RNA polymerase II to the il8 promoter, as well as acetylation of histone H4 and Ser10/Lys14-phosphorylation/acetylation of histone H3 at the il8 gene promoter in HUVEC. In conclusion, Rac1 contributed to L. monocytogenes-induced cytokine expression by human endothelial cells.


Subject(s)
Endothelium, Vascular/enzymology , Listeria monocytogenes/pathogenicity , Signal Transduction , Umbilical Veins/enzymology , rac1 GTP-Binding Protein/biosynthesis , Aminoquinolines/pharmacology , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Cells, Cultured , Cytokines/biosynthesis , Endothelium, Vascular/drug effects , Endothelium, Vascular/microbiology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/pharmacology , Humans , Pyrimidines/pharmacology , Umbilical Veins/drug effects , Umbilical Veins/microbiology
5.
Thromb Haemost ; 94(2): 295-303, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16113818

ABSTRACT

Streptococcus pneumoniae is the major pathogen of community-acquired pneumonia and a common cause of otitis, meningitis and sepsis. During pneumococci infection accompanied with bacterial invasion and hematogenous spreading, the endothelium is directly targeted by pneumococci and their virulence factors. Therefore, we tested the hypothesis that pneumococci induced endothelial apoptosis. Unencapsulated R6x pneumococci strongly induced apoptosis of human endothelial cells both from lung microvasculature and umbilical vein, whereas an encapsulated strain D39 mainly led to necrotic cell death. Deletion of the gene coding for pneumolysin reduced pneumococci-induced apoptosis in HUVEC. Furthermore, N-acetyl-L-cysteine, an antioxidant thiol, significantly reduced apoptosis caused by R6x, and LDH release induced by D39, pointing to a role for reactive oxygen species in the pathogenesis. Apoptotic cells showed increased cleavage and activity of caspases 6 and 9 but only late activation of caspase 3. Programmed cell death could be strongly reduced by pan-caspase inhibitor zVAD. Reduced levels of Bcl2 and cytosolic increase of apoptosis-inducing factor in pneumococci-infected cells implicated involvement of mitochondrial death pathways. Caspase activation and apoptosis were abolished by cAMP elevation. Moreover, p38 mitogen-activated protein kinase and c-Jun NH(2)-terminal kinase were activated in pneumococci-infected cells and inhibitors of both kinases strongly reduced pneumococci-induced caspase activation and apoptosis. Hence, kinase- and caspase-dependence of pneumococci-induced endothelial apoptosis may bear relevance to novel therapeutic approaches to pneumococci-related disease.


Subject(s)
Apoptosis , Caspases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/microbiology , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Annexin A5/pharmacology , Antioxidants/pharmacology , Bacterial Proteins/genetics , Blotting, Western , Caspase 6 , Caspase 9 , Cell Death , Cells, Cultured , Cyclic AMP/metabolism , Cytosol/metabolism , DNA Fragmentation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Humans , Hydrogen Peroxide/pharmacology , L-Lactate Dehydrogenase/metabolism , Lung/blood supply , Lung/microbiology , Microscopy, Fluorescence , Mitochondria/metabolism , Necrosis , Phosphorylation , Propidium/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species , Streptolysins/genetics , Sulfhydryl Compounds/chemistry , Time Factors , Umbilical Veins/cytology , Umbilical Veins/microbiology
6.
Pathog Glob Health ; 109(2): 84-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25736870

ABSTRACT

Neonatal liver abscess is an uncommon seen condition in neonatology and it holds a very high neonatal mortality because of difficulty in diagnosis and treatment. Till today, only few instances are reported that too are mainly in preterm. Its diagnosis requires a high index of suspicion. Fungal hepatic abscess is very rare and in medical literature very few case reports are there in the medical literature. Here, we report a case of Candida albicans liver abscess in a preterm neonate, secondary to malpositioned umbilical lines that presented with respiratory difficulty and other clinical features of sepsis that was managed medically and discharged successfully.


Subject(s)
Candida/isolation & purification , Candidiasis/microbiology , Catheterization, Central Venous/adverse effects , Infant, Premature , Liver Abscess/microbiology , Umbilical Veins/microbiology , Amphotericin B/administration & dosage , Antifungal Agents/administration & dosage , Candidiasis/complications , Candidiasis/drug therapy , Fluconazole/administration & dosage , Humans , Infant, Newborn , Liver Abscess/drug therapy , Male , Treatment Outcome
7.
FEBS Lett ; 532(3): 411-4, 2002 Dec 18.
Article in English | MEDLINE | ID: mdl-12482602

ABSTRACT

Helicobacter pylori upregulates endothelial adhesion molecules but the pattern is unclear. Human umbilical vein endothelial cells (HUVEC) were exposed to control medium or H. pylori 60190. Binding of monoclonal antibodies against P-selectin, E-selectin, vascular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) was determined using enzyme-linked immunosorbent assay. Binding of polymorphonuclear leukocytes to HUVEC was determined on cells exposed as above. After 6 h exposure to H. pylori, there were 30%, 124%, 167% and 100% increases in P-selectin, E-selectin, VCAM-1 and ICAM-1 levels and a 400% increase in polymorphonuclear leukocyte adhesion in HUVEC exposed to H. pylori. Effects of incubation for other intervals between 0 and 18 h are also described. H. pylori exerts some of its effects on gastric mucosa via gastric vasculature. This study gives insight into the pattern of H. pylori-associated endothelial adhesion molecule upregulation.


Subject(s)
Endothelium, Vascular/cytology , Helicobacter pylori/metabolism , Neutrophils/cytology , Neutrophils/microbiology , Umbilical Veins/cytology , Cell Adhesion , Cells, Cultured , E-Selectin/metabolism , Endothelium, Vascular/microbiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , P-Selectin/metabolism , Protein Binding , Time Factors , Umbilical Veins/microbiology , Vascular Cell Adhesion Molecule-1/metabolism
8.
Transplantation ; 52(5): 896-903, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1658993

ABSTRACT

Cardiac allograft vasculopathy (accelerated transplant atherosclerosis) is considered by most to involve a chronic allogeneic immune response to one or more constituents in the coronary vascular wall. Recent evidence suggests that there is an association between cytomegalovirus infection and the development of cardiac allograft vasculopathy (CAV). To determine whether CMV directly infects and/or potentially influences immunogenicity of vascular tissue, human umbilical vein (HU-VECs) or human aortic (HAECs) endothelial cells and human aortic smooth muscle cells (HASMCs) were isolated, cultured, and infected with CMV strain AD 169. Infection was detected using an immunoperoxidase-labeled monoclonal antibody to CMV immediate-early antigen (L-14). The presence and relative quantity of MHC class I and II antigens were determined flow cytometrically using monoclonal antibodies to monomorphic class I and class II HLA determinants. Gamma interferon was used as a positive control stimulant for the upregulation of MHC determinants. Both pooled HUVECs as well as 2 cell lines of HAECs served as targets for CMV infection though less than 10% of the cells were infected despite inocula of 10 pfu/cell. Infection of the pooled HUVECs resulted in no significant changes in the cell surface density of either MHC class I or II determinants. In contrast, HASMCs were excellent targets for CMV infection with virtually 100% of cells infected. CMV infection of 2 distinct HASMC cultures resulted in an increase of 254 +/- 158 relative fluorescence units (RFUs) in MHC class I antigen expression, as assessed by fluorescence intensity, in a variable portion of the HASMCs. A second population of cells exhibited a decrease of 73 +/- 16 RFUs in MHC class I antigen expression. No significant change in MHC class II antigen expression was noted. These results demonstrate that while HUVECs and HAECs are targets of CMV infection, human aortic smooth muscle cells can more readily be infected by CMV. Furthermore, CMV can regulate smooth muscle cell MHC class I expression, hence potentially altering immunogenicity. A pathophysiologic link between cardiac allograft vasculopathy and CMV disease can therefore be hypothesized.


Subject(s)
Cytomegalovirus/immunology , Gene Expression Regulation, Viral , Histocompatibility Antigens Class I/biosynthesis , Muscle, Smooth, Vascular/metabolism , Aorta/metabolism , Aorta/microbiology , Cells, Cultured , Endothelium/metabolism , Endothelium/microbiology , Histocompatibility Antigens Class II/biosynthesis , Humans , In Vitro Techniques , Interferon-gamma/pharmacology , Muscle, Smooth, Vascular/microbiology , Umbilical Veins/metabolism , Umbilical Veins/microbiology
9.
J Comp Pathol ; 104(4): 379-87, 1991 May.
Article in English | MEDLINE | ID: mdl-1651960

ABSTRACT

One of three mares in the last trimester of pregnancy became paraplegic 7 days after experimental infection with EHV-1 and was killed 10 days after infection (d.p.i.). The other two mares aborted foetuses at 12 and 14 d.p.i. In the first mare, virus was detected by immunofluorescence (IIF) and immunoperoxidase (IP) staining in endothelial cells of the endometrium, placenta and umbilical vein, but not in any other foetal tissues. In the experimentally aborted foetuses, and in two other independent field cases of abortions, endothelial cell infection was also detected in the foetuses, both in major blood vessels and in capillaries or sinusoidal cells associated with parenchymal lesions. In these four cases there were also positive endothelial placental lesions detected by IIF or IP, although it was not always possible to isolate virus from these tissues, as it was from the foetuses. The evidence suggests that infection of maternal endothelial cells has a major role in the pathogenesis of abortion, and that endothelial cells are also involved in dissemination of virus within the foetus.


Subject(s)
Abortion, Veterinary/microbiology , Endometrium/microbiology , Endothelium, Vascular/microbiology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/isolation & purification , Horse Diseases/microbiology , Placenta/microbiology , Abortion, Septic/veterinary , Animals , Antibodies, Viral/analysis , Female , Fetus/microbiology , Herpesvirus 1, Equid/immunology , Horse Diseases/transmission , Horses , Immunoenzyme Techniques , Pregnancy , Umbilical Arteries/microbiology , Umbilical Veins/microbiology
10.
J Am Vet Med Assoc ; 214(12): 1813-6, 1791-2, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10382024

ABSTRACT

A technique for laparoscopically assisted resection of umbilical structures in foals was developed. Eleven foals ranging from 8 to 42 days old underwent this procedure. Results of bacteriologic culture of umbilical structures were positive in 7 foals. Mean duration of anesthesia was 99 minutes, of which the initial 20 to 25 minutes were typically devoted to positioning and preparation of the foal for surgery. Major complications did not develop in any foal. Minor complications, such as slippage of the endoscopic ligating clip or laceration of the bladder during dissection, were dealt with successfully during the procedure. Potential benefits of use of laparoscopy (i.e., decreased postoperative morbidity, smaller incisions, and increased intraoperative access to structures) must be carefully weighed against the risks of increased duration of anesthesia. However, anesthetic-related complications were not observed in any foal.


Subject(s)
Animals, Newborn/surgery , Horses/surgery , Laparoscopy/veterinary , Umbilicus/surgery , Animals , Bacterial Infections/surgery , Bacterial Infections/veterinary , Female , Horse Diseases/surgery , Male , Umbilical Arteries/microbiology , Umbilical Arteries/surgery , Umbilical Veins/microbiology , Umbilical Veins/surgery , Umbilicus/microbiology , Urachus/microbiology , Urachus/surgery
14.
Microb Pathog ; 45(5-6): 315-22, 2008.
Article in English | MEDLINE | ID: mdl-18778767

ABSTRACT

Cell adhesion molecules (CAMs) are surface receptors present in eukaryotic cells that mediate cell-cell or cell-extracellular matrix interactions. Vascular endothelium stimulation in vitro that lead to the upregulation of CAMs was reported for the pathogenic spirochaetes, including rLIC10365 of Leptospira interrogans. In this study, we report the cloning of LIC10507, LIC10508, LIC10509 genes of L. interrogans using Escherichia coli as a host system. The rational for selecting these sequences is due to their location in L. interrogans serovar Copenhageni genome that has a potential involvement in pathogenesis. The genes encode for predicted lipoproteins with no assigned functions. The purified recombinant proteins were capable to promote the upregulation of intercellular adhesion molecule 1 (ICAM-1) and E-selectin on monolayers of human umbilical vein endothelial cells (HUVECS). In addition, the coding sequences are expressed in the renal tubules of animal during bacterial experimental infection. The proteins are probably located at the outer membrane of the bacteria since they are detected in detergent-phase of L. interrogans Triton X-114 extract. Altogether our data suggest a possible involvement of these proteins during bacterial infection and provide new insights into the role of this region in the pathogenesis of Leptospira.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Endothelial Cells/metabolism , Gene Expression , Leptospira interrogans/metabolism , Leptospirosis/microbiology , Umbilical Veins/metabolism , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/isolation & purification , Cells, Cultured , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/microbiology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Leptospira interrogans/chemistry , Leptospira interrogans/genetics , Leptospira interrogans/pathogenicity , Leptospirosis/genetics , Leptospirosis/metabolism , Mice , Molecular Sequence Data , Sequence Alignment , Umbilical Veins/cytology , Umbilical Veins/microbiology
15.
Curr Protoc Microbiol ; Chapter 9: Unit 9C.4, 2007 Feb.
Article in English | MEDLINE | ID: mdl-18770623

ABSTRACT

Many successful pathogens have developed the ability to adhere to and invade animal tissues. Recent experimental evidence suggests that S. aureus, generally perceived as an extracellular pathogen, can also invade and, in some cases, multiply within host cells. As a proxy to infections in animal hosts, the study of S. aureus interactions with tissue culture cells has become an important research tool in many aspects of bacterial pathogenesis. In this unit, we describe two cell culture models, including bovine mammary epithelial cells and human umbilical vein endothelial cells, that investigators have used to study the interactions of S. aureus with host cells.


Subject(s)
Endothelial Cells/microbiology , Epithelial Cells/microbiology , Mammary Glands, Animal/microbiology , Staphylococcus aureus/pathogenicity , Umbilical Veins/microbiology , Animals , Bacterial Adhesion , Cattle , Cells, Cultured , Culture Media , Female , Humans , Mammary Glands, Animal/cytology , Staphylococcus aureus/physiology , Umbilical Veins/cytology
16.
J Vasc Surg ; 3(1): 146-54, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3941479

ABSTRACT

This article describes the pathologic changes in 31 human umbilical vein grafts excised from 23 patients after implantations ranging from 24 hours to 5 years. Gross morphologic examination, light microscopy, and scanning electron microscopy demonstrated that the umbilical vein grafts appeared to be fragile and easily delaminated. Bacteremic colonization on the luminal surface was present in the grafts removed because of infections but in two instances had extended into the wall. The presence of lipid on the surface and in the subintimal layer was observed in five grafts, three of which had been implanted for less than 1 month. We concluded that human umbilical vein grafts pathologically exhibit fragility, biodegradation, lipid accumulation, and bacterial colonization in infected cases, and these characteristics may adversely affect the durability and long-term success of the prosthesis.


Subject(s)
Blood Vessel Prosthesis/adverse effects , Postoperative Complications/pathology , Umbilical Veins/pathology , Adult , Aged , Humans , Lipids/analysis , Microscopy, Electron, Scanning , Middle Aged , Muscle, Smooth, Vascular/pathology , Reoperation , Time Factors , Umbilical Veins/microbiology , Umbilical Veins/transplantation
17.
Proc Natl Acad Sci U S A ; 81(23): 7588-90, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6095308

ABSTRACT

We studied the effects of human T-lymphotropic virus type I (HTLV-I) on human endothelial cells in vitro. During cocultivation with an HTLV-I producer cell line (C91/PL), endothelial cells formed characteristic multinucleated syncytial giant cells. Inoculation with concentrated cell-free supernatant fluid from C91/PL cultures produced similar cytopathic effects, which were neutralized by pretreatment with HTLV-I specific human serum. HTLV-I antigens were detected in the cytoplasm of the multinucleated cells by indirect immunofluorescence. When endothelial cells showed maximal cytopathic changes, reverse transcriptase activity was demonstrated in the supernatant fluid and HTLV-I was isolated by cocultivation with peripheral blood mononuclear cells. This study demonstrates that HTLV-I tropism is not limited to lymphoid cells but extends to human endothelial cells as well.


Subject(s)
Deltaretrovirus/pathogenicity , Endothelium/microbiology , Umbilical Veins/microbiology , Antigens, Viral/isolation & purification , Cells, Cultured , Deltaretrovirus/isolation & purification , Female , Fluorescent Antibody Technique , Humans , Pregnancy
18.
Proc Natl Acad Sci U S A ; 81(23): 7591-5, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6095309

ABSTRACT

The effects of the human T-cell leukemia virus type I (HTLV-I) on cultured human endothelial cells were evaluated. Coculture of endothelial monolayers with either irradiated HTLV-producing lymphocytes or cell-free virus resulted in the production of multinucleated syncytia. The development of syncytia was inhibited by sera from patients with adult T-cell leukemia/lymphoma (ATLL). HTLV antigens were present on endothelial syncytia passaged in culture for greater than 3 months as detected by an anti-p19 monoclonal antibody, which detects a core protein of HTLV-I, and by ATLL sera. Moreover, these HTLV-infected endothelial cells were then able to infect and transform normal cord blood T lymphocytes with HTLV. These studies demonstrate that human endothelial cells are susceptible to productive HTLV-I infection in vitro and may have relevance for the spectrum of human disease associated with this family of retroviruses.


Subject(s)
Deltaretrovirus/pathogenicity , Endothelium/microbiology , Umbilical Veins/microbiology , Antibodies, Monoclonal , Antigens, Viral/analysis , Cell Transformation, Viral , Cells, Cultured , Endothelium/ultrastructure , Female , Humans , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/microbiology , Microscopy, Electron , Pregnancy
19.
Lab Invest ; 68(6): 645-51, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8390591

ABSTRACT

BACKGROUND: Herpes simplex virus (HSV)-infected endothelium is a model for vascular injury and possibly the development of atherosclerosis. In vitro infection of human umbilical vein endothelial cells (HUVEC) by HSV-1 results in a number of changes including the expression of a procoagulant activity (PCA) compatible with that due to tissue factor (TF) synthesis. In this study, we have further characterized this PCA using more stringent assays for TF, and examined whether virus rendered incapable of replication retains the ability to stimulate TF synthesis in HUVEC. EXPERIMENTAL DESIGN: Confluent monolayers of HUVEC were exposed to intact or ultraviolet/heat-inactivated HSV-1. At appropriate time intervals, TF PCA was assessed by clotting assays, and TF antigen by an enzyme-linked immunosorbent assay specific for TF. The appearance of mRNA specific for TF was performed by Northern blotting. RESULTS: TF activity was demonstrated by both 1-stage and 2-stage clotting assays; the dependence of the latter on factor VIIa, and the inhibition by specific blocking antibodies to human TF support the notion that the PCA is indeed due to TF. Furthermore, cellular TF antigen levels were found to parallel TF activity, and there was a transient de novo expression of TF mRNA. Tissue factor PCA in HSV-infected HUVEC remained "encrypted"; that is, full clotting activity was not expressed in the absence of cellular disruption in a situation analogous to that seen in all normal cells thus far examined that express TF PCA. However, this response did not appear to be dependent upon replicative infection of HSV-1 within the endothelial cell since a similar (although lesser) induction of TF PCA was present in cells that had been exposed to virus previously rendered incapable of replication. CONCLUSIONS: HSV-1 induces PCA in HUVEC which is clearly TF-dependent; this response does not require viral replication. These data indicate increased complexity in HSV interactions with vascular endothelium and imply induction of some procoagulant functions by nonproductive infection.


Subject(s)
Blood Coagulation Factors/metabolism , Endothelium, Vascular/metabolism , Herpes Simplex/metabolism , Simplexvirus/physiology , Thromboplastin/metabolism , Umbilical Veins/metabolism , Cells, Cultured , Endothelium, Vascular/microbiology , Humans , Umbilical Veins/microbiology , Virus Replication
20.
J Trauma ; 56(1): 105-10, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14749575

ABSTRACT

OBJECTIVE: We have previously documented that gut-derived lymph from rats subjected to trauma plus hemorrhagic shock (T/HS) is injurious to vascular endothelial cells and activates neutrophils (PMNs), two key events in postshock organ injury. Because T/HS leads to gut injury, intestinal bacterial overgrowth, and the loss of gut barrier function, the relative role of gut injury as opposed to intestinal bacterial overgrowth per se in the pathogenesis of biologically active intestinal lymph is unclear. We therefore studied whether mesenteric lymph can injure endothelial cells and/or active PMNs in an intestinal bacterial overgrowth model where there is no gut injury (monoassociation). METHODS: Bacterial overgrowth was established in male rats by treating the animals with 4 days of oral antibiotics followed by administration of a nonpathogenic, streptomycin-resistant strain of Escherichia coli C25. Mesenteric lymph was then collected from rats with normal flora and from E. coli C25 monoassociated rats. Its effects were tested on human umbilical vein endothelial cells (HUVECs) and human PMNs. As an additional control, lymph was collected from antibiotic-decontaminated rats that received antibiotics but were not colonized with E. coli C25. RESULTS: As compared with medium, normal flora intestinal lymph, antibiotic-decontaminated lymph, or portal plasma from the monoassociated rats, mesenteric lymph from the monoassociated rats killed HUVECs and increased the permeability of a HUVEC monolayer. In contrast to the effects on HUVECs, lymph from the monoassociated rats did not increase PMN CD11b expression or prime PMNs for an augmented respiratory burst, as compared with lymph from the rats with normal flora or from antibiotic-decontaminated rats. The effects of lymph from the monoassociated rats was not caused by bacteria, because these lymph samples were sterile. CONCLUSION: These results indicate that disruption of the normal intestinal microflora resulting in bacterial overgrowth with enteric bacilli may participate in the production of mesenteric lymph that is injurious to endothelial cells in shock, but this mechanism does not appear to be significantly involved in the activation of PMNs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/microbiology , Intestines/microbiology , Neutrophils/microbiology , Animals , Escherichia coli Infections/prevention & control , Humans , Intestines/drug effects , Lymph/microbiology , Male , Rats , Rats, Sprague-Dawley , Umbilical Veins/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL