Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 25(3): 753-60, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17313571

ABSTRACT

Recently, infections have been implicated in the pathogenesis of Alzheimer's disease. Apart from the direct effects of pathogens, it can be hypothesized that inflammatory mechanisms, such as the production of pro-inflammatory mediators by resident glia, may result in neurotoxicity. Here, we examined the inflammatory responses in murine microglial cell (MMC) and murine astrocyte cell (MAC) lines following infection with Chlamydia pneumoniae (Cpn), a pathogen that has recently been associated with Alzheimer's disease. Furthermore, we determined whether these inflammatory responses are sufficient to cause neuronal cell death in vitro. MMCs and MACs were infected with Cpn. Subsequently, various chemo- and cytokines were determined in the culture supernatant fluid of infected/control cells at different time points post-infection. Significantly higher levels of monocyte chemoattractant protein 1, interleukin (IL)-6, tumour necrosis factor (TNF)-alpha and IL-1beta were found in supernatant fluids of infected MMCs compared with controls. In contrast, in the supernatant fluid of infected MACs, only monocyte chemoattractant protein 1 and IL-6 displayed significantly higher levels compared with controls. Moreover, neurotoxicity was examined up to 72 h after transferring the conditioned supernatant fluid to a neuronal cell layer. No neuronal cell death was observed when supernatant fluids from infected/mock-treated MACs were transferred. However, when neurones were exposed to conditioned supernatant fluid from infected MMCs, a significant increase in cell death was observed compared with mock. Furthermore, adding neutralizing antibodies against IL-6 and TNF-alpha to that conditioned supernatant fluid prevented neuronal cell death by approximately 50%. In conclusion, these data suggest that Cpn infection results in a pro-inflammatory milieu, particularly by activating MMCs, that ultimately results in neurodegeneration with prominent roles for both IL-6 and TNF-alpha.


Subject(s)
Astrocytes/immunology , Chlamydophila Infections/immunology , Chlamydophila pneumoniae , Microglia/immunology , Animals , Antibodies/pharmacology , Apoptosis/immunology , Astrocytes/cytology , Astrocytes/microbiology , Carcinoma, Hepatocellular , Cell Line, Tumor , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Liver Neoplasms , Microglia/cytology , Microglia/microbiology , Muridae , Necrosis , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
2.
Transplant Proc ; 39(1): 261-7, 2007.
Article in English | MEDLINE | ID: mdl-17275518

ABSTRACT

Long-term survival of solid-organ transplants is limited as a result of chronic transplant dysfunction (CTD), which is characterized by occlusion of intragraft vascular tissue due to myointimal hyperplasia. Recent studies have shown a role for infections in vascular pathologies. For example, Chlamydia pneumoniae (Cpn) has been shown to aggravate atherosclerosis, and Cpn immunoglobulin (Ig)G titers correlate with severity of allograft atherosclerosis after cardiac transplantation. In this study, we evaluated the effect of Cpn infection on CTD using a rat aortic allograft model. Orthotopic abdominal aorta transplantations (Tx) were performed with Brown Norway rats as donors and Lewis rats as recipients. Rats were humanely killed at 1 or 8 weeks after surgery. The graft was processed for DNA isolation and histological examination. Influx of macrophages and T cells was assessed using immunohistochemistry. At 1 week after Tx, the perivascular influx of inflammatory cells in the graft was not affected by Cpn infection. Furthermore, only limited numbers of Cpn DNA copies were found in the graft at 1 week after Tx. In addition, Cpn did not alter the severity of myointimal hyperplasia in the rat aortic allograft model at 8 weeks after surgery. Our data suggested that, in the rat aortic allograft model, Cpn infection did not influence the influx of inflammatory cells or the severity of CTD.


Subject(s)
Aorta/transplantation , Chlamydia Infections/physiopathology , Chlamydophila pneumoniae , Transplantation, Homologous , Animals , Male , Models, Animal , Postoperative Complications/physiopathology , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Isogeneic
3.
Arch Virol ; 150(12): 2593-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16052287

ABSTRACT

We have identified and characterized two antisense transcripts from the rat cytomegalovirus (RCMV) major immediate early (MIE) region. These transcripts, designated IE-AS1 and IE-AS2, are complementary to part of the sense IE1 transcript. The IE-AS transcripts were first detected in peripheral blood leukocytes (PBL) of RCMV-infected rats at 7 days post-infection (pi) in the absence of IE1 transcription. Nevertheless, both the IE1 and IE-AS transcripts were found at the same time in the salivary glands of RCMV-infected rats at 7 and 120 days pi as well as in RCMV-infected rat embryo fibroblasts (REFs) at 48 h pi.


Subject(s)
Antigens, Viral/genetics , Immediate-Early Proteins/genetics , Muromegalovirus/genetics , RNA, Antisense/analysis , RNA, Messenger/analysis , RNA, Viral/analysis , Amino Acid Sequence , Animals , Base Sequence , Fibroblasts/virology , Leukocytes/virology , Molecular Sequence Data , RNA, Antisense/genetics , RNA, Complementary/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Rats , Salivary Glands/virology
4.
Cardiovasc Res ; 56(2): 269-76, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12393097

ABSTRACT

OBJECTIVE: Atherosclerosis is an inflammatory process and is characterised by the presence of T-lymphocytes in the lesions. To study the role of Chlamydophila pneumoniae (C. pneumoniae) in this process and the effect of infection on T-cell influx, we infected Apo E3-Leiden mice with C. pneumoniae and investigated the effect on lesion development and T-cell influx in atherosclerotic lesions at different time points post infection (pi). METHODS: Nine week old mice, fed an atherogenic diet, were either mock-infected or infected with C. pneumoniae and sacrificed at 1, 6 and 9 months pi. Longitudinal sections of the aortic arches of the mice were stained with hematoxylin-eosin for atherosclerotic lesion type and lesion area analysis, or with rabbit-anti-CD3(+) to detect the presence of T-cells in the atherosclerotic lesions. T-cell influx was expressed as number of T-lymphocytes/lesion area. RESULTS: At 1 month pi, type 1, 2 and 3 lesions were present. At other time points pi, more complex lesion types 4, 5a and 5b were also present. Although infection did not influence the total lesion number or area, we observed an effect of C. pneumoniae infection on lesion type. Infection resulted in a significant shift in lesion formation from type 3 to type 4 (P=0.022) at 6 months pi, and from type 4 to type 5a (P=0.002) at 9 months pi. T-cells were observed at every time point pi. At 1 month pi, a significant increase in T-cell influx in the C. pneumoniae-infected atherosclerotic lesions was observed (P=0.0005). CONCLUSION: This study shows that C. pneumoniae infection enhances the inflammatory process by increasing T-lymphocytes in the plaque and accelerates the formation of complex lesions.


Subject(s)
Arteriosclerosis/microbiology , Chlamydia Infections/complications , Chlamydophila pneumoniae , Animals , Aortitis/immunology , Aortitis/microbiology , Aortitis/pathology , Arteriosclerosis/immunology , Arteriosclerosis/pathology , Cholesterol/blood , Diet, Atherogenic , Disease Progression , Female , Hypercholesterolemia/complications , Male , Mice , Mice, Inbred C57BL , Risk Factors , T-Lymphocytes/pathology , Triglycerides/blood
5.
Arch Virol ; 147(4): 813-24, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12038690

ABSTRACT

We previously generated an RCMV strain in which the r144 gene, encoding a major histocompatibility complex class I homolog, had been deleted (RCMVdelta r144). To investigate the role of r144 during acute infection of neonatal rats, we infected three days-old neonatal rats with either RCMVdelta r144 or wild type (wt) RCMV and the presence of infectious virus as well as viral DNA in various organs was determined at either 3, 5 or 21 days p.i.. In addition, we assessed both type and number of inflammatory cells in these organs. Interestingly, a significantly lower concentration of infectious virus as well as viral DNA was found in spleens of RCMVdelta r144-infected rats than in those of wt RCMV-infected animals at 3 days p.i.. At the same time point, a significantly lower amount of infiltrating NK cells and monocytes/macrophages was seen in the spleens of RCMVdelta r144-infected rats than in spleens of rats infected with wt RCMV. At 21 days p.i., RCMVdelta r144-infected rats were found to have lower virus titers in the salivary glands than wt RCMV-infected animals. Significant differences between RCMVdelta r144- and wt RCMV-infected rats were detected neither at other time points nor at other sites. We conclude that after infection of neonatal rats, the replication of RCMVdelta r144 is severely restricted compared to wt RCMV.


Subject(s)
Genes, MHC Class I/genetics , Herpesviridae Infections/virology , Muromegalovirus/genetics , Viral Proteins/genetics , Acute Disease , Animals , Animals, Newborn , Cell Count , DNA, Viral/analysis , Disease Models, Animal , Female , Gene Deletion , Herpesviridae Infections/immunology , Leukocytes/virology , Macrophages/virology , Male , Muromegalovirus/chemistry , Rats , Salivary Glands/virology , Spleen/immunology , Spleen/virology , Time Factors
6.
Arch Virol ; 146(11): 2211-8, 2001.
Article in English | MEDLINE | ID: mdl-11765922

ABSTRACT

Rat cytomegalovirus (RCMV) open reading frame R44 is the homolog of human cytomegalovirus gene UL44, which encodes the DNA polymerase accessory protein. Here, we show that R44 is transcribed as a 3.6-kb mRNA within the early and late phases of infection in vitro. In order to find potential monoclonal antibodies (MoAbs) directed against the R44-encoded protein (pR44), a panel of anti-RCMV MoAbs was screened for binding to pR44 recombinant proteins. Thus, an anti-pR44 MoAb, termed RCMV8, was identified. By using this MoAb, pR44 could be detected as early as 8 h after RCMV infection in vitro. The pR44 protein was determined to have a molecular mass of approximately 55 kDa and was found to be localized to the nucleus of RCMV-infected cells.


Subject(s)
Cytomegalovirus/genetics , Genes, Viral , Nuclear Proteins/genetics , Open Reading Frames , Animals , Molecular Weight , Nuclear Proteins/analysis , Rats
7.
Virchows Arch ; 437(4): 413-21, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11097367

ABSTRACT

The salivary gland is the preferred organ for cytomegalovirus (CMV) replication and viral persistence. In order to identify the nature of infected cells and to study viral replication in more detail, several experiments were conducted. Using the rat CMV (RCMV) model, acutely infected young adult rats (6 weeks of age) and new-born rats (3 days of age) were infected, and submandibular, parotid and sublingual glands were harvested at different time points after infection. For identification of the nature of infected cells, immunohistochemistry, in situ hybridisation and electron microscopic techniques were used. In young adult animals, the submandibular gland was the preferred organ for RCMV replication. The parotid and sublingual glands contained fewer viruses than the submandibular gland. In contrast, in new-born rats, the main site of RCMV replication was the sublingual gland, while the submandibular and parotid glands contained low amounts of virus. No virus could be detected in the parotid glands. In all glands of RCMV-infected animals, the infection was exclusively confined to striated duct cells. Infection resulted in a cellular inflammatory response which was mostly located in the interlobular duct region, whereas only few inflammatory cells were found in the neighbourhood of infected striated duct cells. This phenomenon may contribute to the long persistence of the virus in this organ.


Subject(s)
Cytomegalovirus/isolation & purification , Salivary Glands/virology , Virus Replication , Animals , Cytomegalovirus/physiology , Immunohistochemistry , Male , Microscopy, Electron , Rats , Rats, Inbred Lew , Salivary Glands/ultrastructure
8.
Transpl Int ; 13(1): 54-63, 2000.
Article in English | MEDLINE | ID: mdl-10743691

ABSTRACT

Previous studies have demonstrated that both cytomegalovirus (CMV) infection and prolonged cold ischemia of the allograft (CI) are associated with chronic rejection of renal transplants. The purpose of this study is to investigate the effect of CMV infection, of CI and of the combination of both, on the progression of chronic rejection, and to obtain a more detailed insight in their effects on the expression of adhesion molecules. Therefore, a rat transplantation model was used. Lewis recipients of renal allografts (with and without CI) from MHC-incompatible Brown Norway rats were inoculated with rat CMV or left uninfected. CMV infection alone resulted in an increased influx of CD4+ cells and macrophages early after infection, and in an increase in glomerular sclerosis and intima proliferation. CI caused an increase in infiltrating NK cells and an effect on intimal proliferation, glomerular sclerosis, and tubular atrophy. When CMV infection and CI were combined, an additive effect could be measured. This was however not the case for the function of the kidney. The creatinin showed a synergistic effect of the two influencing factors. Due to the CMV infection, an increase in CD49d cells was detected. CI resulted in an increase in CD18 cells and an increase in the expression of CD62P on vessels, and CD54 and CD44 on tubules. When CMV infection and CI were combined, all the effects caused by CMV and CI alone were present in an additional way. The results of the present study suggest that special attention should be paid to the recipient of an ischemically injured graft when either the donor or the recipient is CMV-infected. The patterns seen in histology, the infiltration of leukocytes and the expression of adhesion molecules, suggest that CI and CMV infection both have an effect on rejection, but act by different mechanisms.


Subject(s)
Cytomegalovirus Infections/complications , Graft Rejection/physiopathology , Kidney Transplantation/physiology , Kidney , Animals , Antigens, Differentiation/analysis , Atrophy , CD4-Positive T-Lymphocytes/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Ischemia , Kidney Transplantation/immunology , Kidney Transplantation/pathology , Leukocytes/pathology , Lymphocytes/pathology , Male , Organ Preservation , Postoperative Complications , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Homologous
9.
J Virol ; 74(2): 1045-50, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623772

ABSTRACT

The rat cytomegalovirus (RCMV) r144 gene encodes a polypeptide homologous to major histocompatibility complex class I heavy chains. To study the role of r144 in virus replication, an RCMV r144 null mutant strain (RCMVDeltar144) was generated. This strain replicated with efficiency similar to that of wild-type (WT) RCMV in vitro. Additionally, WT RCMV and RCMVDeltar144 were found not to differ in their replication characteristics in vivo. First, the survival rate was similar among groups of immunosuppressed rats infected with either RCMVDeltar144 or WT RCMV. Second, the dissemination of virus did not differ in either RCMVDeltar144- or WT RCMV-infected, immunosuppressed rats, either in the acute phase of infection or approximately 1 year after infection. These data indicate that the RCMV r144 gene is essential neither for virus replication in the acute phase of infection nor for long-term infection in immunocompromised rats. Interestingly, in a local infection model in which footpads of immunosuppressed rats were inoculated with virus, a significantly higher number of infiltrating macrophage cells as well as of CD8(+) T cells was observed in WT RCMV-infected paws than in RCMVDeltar144-infected paws. This suggests that r144 might function in the interaction with these leukocytes in vivo.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Cytomegalovirus/physiology , Cytomegalovirus Infections/immunology , DNA, Viral , Genes, MHC Class I , Genes, Viral , Humans , Immunocompromised Host , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Molecular Sequence Data , Mutagenesis , Rats , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Time Factors , Viral Proteins/physiology , Virus Replication/physiology
10.
Antiviral Res ; 44(1): 55-65, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10588333

ABSTRACT

Cytomegalovirus (CMV) infection is a major problem in the immunosuppressed patient. It is thought that besides direct CMV induced cell lysis, immunological damage is part of CMV pathogenesis. New antiviral drugs, which combine immunomodulating and antiviral qualities, could be beneficial. Recently, it has been described that desferrioxamine (DFO) and calcium trinatrium diethylenetriaminepentaacetic acid (DTPA) exhibit both properties. In this report the antiviral effects of both compounds against rat CMV (RCMV) are described in vitro and in vivo using a generalised and local infection model. In vitro, both compounds exhibited a significant antiviral effect, DTPA being more potent than DFO. However, in the generalised infection model no effect was seen on mortality, morbidity or presence of virus in internal organs. In rats infected subcutaneously in the hind paw, no effect was seen locally on paw thickness, presence of viral antigens and inflammatory response. In addition, these rats suffered from a generalised infection of low magnitude at 15 days post infection, although both DFO and DTPA were able to lower the level of viral replication. In conclusion, our data indicate that despite in vitro activity, in vivo usage of DFO or DTPA for acute CMV infection is not warranted.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Deferoxamine/pharmacology , Pentetic Acid/pharmacology , Virus Replication/drug effects , Animals , Antigens, Viral/analysis , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Injections, Intraperitoneal , Injections, Subcutaneous , Liver/virology , Rats , Rats, Inbred Lew , Salivary Glands/virology , Spleen/virology , Viral Plaque Assay
11.
J Virol ; 73(9): 7218-30, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10438809

ABSTRACT

The rat cytomegalovirus (RCMV) R78 gene belongs to an uncharacterized class of viral G protein-coupled receptor (GCR) genes. The predicted amino acid sequence of the R78 open reading frame (ORF) shows 25 and 20% similarity with the gene products of murine cytomegalovirus M78 and human cytomegalovirus UL78, respectively. The R78 gene is transcribed throughout the early and late phases of infection in rat embryo fibroblasts (REF) in vitro. Transcription of R78 was found to result in three different mRNAs: (i) a 1.8-kb mRNA containing the R78 sequence, (ii) a 3.7-kb mRNA containing both R77 and R78 sequences, and (iii) a 5.7-kb mRNA containing at least ORF R77 and ORF R78 sequences. To investigate the function of the R78 gene, we generated two different recombinant virus strains: an RCMV R78 null mutant (RCMVDeltaR78a) and an RCMV mutant encoding a GCR from which the putative intracellular C terminus has been deleted (RCMVDeltaR78c). These recombinant viruses replicated with a 10- to 100-fold-lower efficiency than wild-type (wt) virus in vitro. Interestingly, unlike wt virus-infected REF, REF infected with the recombinants develop a syncytium-like appearance. A striking difference between wt and recombinant viruses was also seen in vivo: a considerably higher survival was seen among recombinant virus-infected rats than among RCMV-infected rats. We conclude that the RCMV R78 gene encodes a novel GCR-like polypeptide that plays an important role in both RCMV replication in vitro and the pathogenesis of viral infection in vivo.


Subject(s)
Cytomegalovirus/genetics , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Viral Proteins/genetics , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cytomegalovirus/physiology , DNA, Viral , Gene Deletion , Genes, Viral , Giant Cells/virology , Humans , Molecular Sequence Data , Mutagenesis , Rats , Receptors, Cell Surface/physiology , Sequence Analysis, DNA , Transcription, Genetic , Viral Proteins/physiology , Virus Replication
12.
Transplantation ; 65(10): 1298-304, 1998 May 27.
Article in English | MEDLINE | ID: mdl-9625009

ABSTRACT

BACKGROUND: The development of chronic rejection has emerged as a major cause of long-term graft failure. Previous studies have demonstrated that cytomegalovirus (CMV) infection is associated with an increased incidence of chronic allograft rejection in renal, cardiac, and aortic allografts. This study was designed to investigate the effects of the major histocompatibility complex (MHC) class I or class II mismatches on CMV-enhanced chronic rejection. METHODS: Aortic transplantation was performed between different inbred rat strain combinations; the Lewis to RP combination was class I-mismatched and Wag/Rij to RP class II-mismatched. At 7, 28, and 90 days after transplantation, the intensity of chronic rejection in mismatched grafts with or without CMV infection was evaluated using histological and immunohistological analysis. RESULTS: The results of this study demonstrated that CMV infection led to an increased influx of monocytes/ macrophages in class I-mismatched grafts at 1 week after transplantation and enhanced infiltration of T lymphocytes in class II-mismatched grafts at 4 weeks. Although more vascular lesions were observed in the class II-mismatched combinations, an intensified neointima formation by CMV infection was observed only in the MHC class I-mismatched allografts. CONCLUSIONS: CMV infection may increase neointima formation of allografts when an MHC class I disparity between donor and recipient is present. This may be associated with the increased perivascular influx of monocytes/macrophages observed in CMV-infected animals early after transplantation.


Subject(s)
Aorta/growth & development , Aorta/transplantation , Herpesviridae Infections/physiopathology , Muromegalovirus , Tunica Intima/growth & development , Animals , Aorta/immunology , Aortitis/pathology , Aortitis/virology , Graft Rejection/physiopathology , Histocompatibility , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Male , Rats , Rats, Inbred Strains , Transplantation, Homologous
13.
J Virol ; 72(3): 2352-63, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499096

ABSTRACT

We have identified a rat cytomegalovirus (RCMV) gene that encodes a G-protein-coupled receptor (GCR) homolog. This gene (R33) belongs to a family that includes the human cytomegalovirus UL33 gene. R33 was found to be transcribed during the late phase of RCMV infection in rat embryo fibroblasts. Unlike the mRNAs from all the other members of the UL33 family that have been studied to date, the R33 mRNA is not spliced. To study the function of the R33 gene, we constructed an RCMV strain in which the R33 open reading frame is disrupted. The mutant strain (RCMV deltaR33) did not show differences in replication from wild-type RCMV upon infection of several rat cell types in vitro. However, marked differences were seen between the mutant and wild-type strain in the pathogenesis of infection in immunocompromised rats. First, the mutant strain induced a significantly lower mortality than the wild-type virus did. Second, in contrast to wild-type RCMV, the mutant strain did not efficiently replicate in the salivary gland epithelial cells of immunocompromised rats. Although viral DNA was detected in salivary glands of RCMV deltaR33-infected rats up to 14 days postinfection, it could not be detected at later time points. This indicates that although the strain with R33 deleted is probably transported to the salivary glands in a similar fashion to that for wild-type virus, the mutant virus is not able to either enter or replicate in salivary gland epithelial cells. We conclude that the RCMV R33 gene plays a vital role in the pathogenesis of infection.


Subject(s)
Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cytomegalovirus/physiology , DNA, Viral , Gene Deletion , Genes, Viral , Humans , Molecular Sequence Data , Mutagenesis , RNA, Viral , Rats , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Transcription, Genetic , Viral Proteins/metabolism , Virus Replication
14.
Clin Exp Immunol ; 110(3): 349-57, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9409635

ABSTRACT

Infection with human cytomegalovirus (HCMV) mostly results in a chronic subclinical infection; the immune system is unable to eliminate the virus and is apparently in equilibrium with the persistent virus. In the immunosuppressed host this equilibrium is disturbed, resulting in clinical infection. Rat cytomegalovirus (RCMV) infection in its host can be used as a model for HCMV infection. Using flow cytometry we examined the effect of acute RCMV infection on the composition of leucocyte subsets in the peripheral blood of both immunocompetent and immunosuppressed (5 Gy total body irradiation) Lewis rats. Special attention was paid to the natural killer (NK) cells and the CD8+ T cells known to be involved in the control of viral infections. Furthermore, we determined the presence of leucocyte subsets in the internal organs by immunohistochemistry. In immunocompetent rats, infection caused a small increase in NK cells and a large increase in CD8+ T cells. In contrast, infection of immunosuppressed rats caused a marked increase in NK cells and a small increase in CD8+ T cells, consisting of T cells with reduced expression of both CD8 and TCR. This phenomenon is characteristic of anergic CD8+ T cells, possibly explaining the ability of the virus to escape elimination by the immune system. The increase of NK cells in the peripheral blood of immunosuppressed, RCMV-infected rats could also be detected in kidney, liver, lung and pancreas, but not in salivary gland. This could explain the long persistence of infectious virus in the salivary gland.


Subject(s)
CD8 Antigens/analysis , Cytomegalovirus Infections/immunology , Immune Tolerance , Receptors, Antigen, T-Cell/analysis , T-Lymphocytes/immunology , Animals , Immunocompromised Host , Killer Cells, Natural/immunology , Male , Rats , Rats, Inbred Lew
15.
Cardiovasc Res ; 36(2): 282-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9463640

ABSTRACT

OBJECTIVE: Recently, we have found that rat CMV (RCMV) infected smooth muscle cells (SMCs) in rat carotid arteries when administered 14 days after balloon injury. In the present study we investigated (1) the long term effects of CMV infection on neointimal cross-sectional area, and (2) whether the phenotype of the intimal SMCs influences their susceptibility to active CMV infection. METHODS: In the first part of the study, rats received RCMV intravenously, two weeks after balloon catheterisation of the left carotid artery and were sacrificed twenty weeks after catheterisation. Continuous BrdU infusion was performed by subcutaneously implanted osmotic pumps during the last two weeks of life. In the second part RCMV was administered eight weeks after catheterisation and rats were sacrificed two weeks later. Immunohistochemistry was used to detect viral antigens, to determine BrdU incorporation as well as the contents of alpha-actin, desmin and vimentin in the carotid arteries. Intima and media cross-sectional areas were determined using computerized morphometry. RESULTS AND CONCLUSIONS: RCMV infection did not induce any differences in intima or media cross-sectional areas of the injured carotid artery, nor in the extent of SMC proliferation as shown by BrdU incorporation, 20 weeks after balloon catheterisation. Eight weeks after balloon catheterisation, RCMV no longer infected neointimal SMCs. This non-responsiveness to RCMV was associated with "re-differentiation" of the eight weeks old neointima, compared with two weeks after catheterization, as shown by the contents of alpha-actin, desmin and vimentin. Our data suggest that intimal SMC phenotype determines its susceptibility to active RCMV infection in vivo. Since de-differentiation of neointimal SMCs is associated with enhanced proliferation of these cells it is stated that de-differentiation or proliferation is prerequisite for infection.


Subject(s)
Carotid Artery Injuries , Catheterization , Cytomegalovirus Infections/transmission , Muscle, Smooth, Vascular/virology , Tunica Intima/virology , Analysis of Variance , Animals , Antigens, Viral/analysis , Biomarkers/analysis , Carotid Artery, Common/pathology , Carotid Artery, Common/virology , Cell Differentiation , Cell Division , Cytomegalovirus Infections/pathology , Desmin/analysis , Disease Susceptibility , Muscle, Smooth, Vascular/pathology , Random Allocation , Rats , Rats, Inbred WKY , Time Factors , Tunica Intima/pathology
16.
Transpl Int ; 9 Suppl 1: S340-4, 1996.
Article in English | MEDLINE | ID: mdl-8959860

ABSTRACT

We previously demonstrated that cytomegalovirus (CMV) infection enhanced perivascular inflammation in rat aortic allografts. In this study, we investigated the relationship between the CMV infection load and the magnitude of perivasculitis (chronic rejection) in aortic transplants. Rats received orthotopic abdominal aortic grafts, different degrees of total body irradiation (TBI) for immunosuppression and CMV inoculation. The spleens of the rats receiving 5 Gy of TBI contained more infectious virus and viral antigens than those of rats receiving 3 Gy of TBI or no TBI. Although the number of inflammatory cells infiltrating the perivascular area was decreased after TBI, CMV infection resulted in increased perivasculitis in rats that received 5 Gy of TBI as compared to non-infected animals. This virus-induced effect was characterized predominantly by an increased T-cell infiltration, including CD4 and CD8 T-cells. It is concluded that an enhanced systemic CMV infection during severe immunosuppressive therapy can accelerate the development of chronic rejection, which seems to be mediated mainly by T-cells.


Subject(s)
Aorta/transplantation , Cytomegalovirus Infections/complications , Vasculitis/etiology , Animals , Graft Rejection/etiology , Male , Rats , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes/immunology , Transplantation, Homologous , Whole-Body Irradiation
18.
In Vivo ; 7(6A): 567-73, 1993.
Article in English | MEDLINE | ID: mdl-8193277

ABSTRACT

There is evidence in support of a viral etiology for atherosclerosis. In our study, we demonstrated that cytomegalovirus (CMV) infection in rats caused morphological alterations of the endothelium and subendothelial space of the large vessels and that these alterations are similar to those which are induced by hypercholesterolemia. These alterations consisted of swollen endothelial cells with a surface showing bleb and microvilli formation. In addition, adhesion of leukocytes to the endothelium and the presence of leukocytes (lymphocytes and macrophages) in the subendothelium were found. Lipid accumulation occurred in the endothelium and in the subendothelial space, especially in hypercholesterolemic animals. This lipid accumulation in the subendothelial space consisted of extracellular lipid deposition and of subendothelial located "foam cells". A characteristic phenomenon of the effect of CMV infection of rats was a loosening of the endothelial cells from the basement membrane. The space between the basement membrane and endothelium was expanded and was filed with an increased amount of reticular basal lamina-like material. These observations show that CMV infection is associated with a non-deniding aortic endothelial injury which is consistent with the early events in the process of atherosclerosis.


Subject(s)
Aorta/ultrastructure , Arteriosclerosis/pathology , Cytomegalovirus Infections/pathology , Endothelium, Vascular/ultrastructure , Muscle, Smooth, Vascular/ultrastructure , Animals , Aorta/pathology , Arteriosclerosis/microbiology , Cell Membrane/pathology , Cell Membrane/ultrastructure , Endothelium, Vascular/pathology , Male , Microscopy, Electron , Microscopy, Electron, Scanning , Muscle, Smooth, Vascular/pathology , Rats , Rats, Wistar , Reference Values
19.
Atherosclerosis ; 93(1-2): 41-52, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1317707

ABSTRACT

The role of cytomegalovirus (CMV) in the early development of atherosclerosis was studied in a rat model. Arterial samples derived from virus-infected normo- and hypercholesterolaemic animals were investigated by light microscopy at 1, 4, 8 and 16 weeks post infection. Early atherogenic lesions comparable to those seen in non-infected hypercholesterolaemic rats were found in CMV-infected normocholesterolaemic and hypercholesterolaemic animals, starting at 1 week post infection. The changes consisted of minimal endothelial cell damage, as shown by the en face technique, and a more than 10-fold increase in the number of leukocytes adhering to the aortic intima. The increased adhesion of leukocytes was observed in infected normocholesterolaemic rats but only in the non-infected rats which were hypercholesterolaemic. The infection of hypercholesterolaemic rats did not enhance this effect although it resulted in increased migration of the leukocytes into the subendothelial space. CMV infection of normocholesterolaemic rats induced lipid accumulation in the endothelium. In these animals approximately 1% of the endothelial cells contained lipid at 1 week post infection. In the non-infected hypercholesterol-fed animals 10% of the cells contained lipid. CMV infection in these rats induced an extra increase of the lipid-containing endothelial area. The changes in the CMV infected animals largely corresponded with the intimal injury observed in the hypercholesterolaemic rats. These results support the hypothesis that CMV may be one of the factors involved in atherogenesis.


Subject(s)
Cytomegalovirus Infections/pathology , Endothelium, Vascular/pathology , Animals , Aorta/pathology , Arteriosclerosis/etiology , Cell Adhesion , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/complications , Hypercholesterolemia/blood , Hypercholesterolemia/complications , Hypercholesterolemia/pathology , Leukocytes/pathology , Male , Rats , Rats, Inbred Strains , Triglycerides/blood
20.
J Leukoc Biol ; 45(5): 466-73, 1989 May.
Article in English | MEDLINE | ID: mdl-2540258

ABSTRACT

An intraperitoneal (rat) cytomegalovirus (RCMV) infection in the rat caused an influx of mononuclear cells, which have been altered in functions and arachidonic acid (AA) metabolism. Phagocytosis has been increased considerably 3 days postinfection (p.i.), whereas the release of prostacyclin, thromboxane A2, 12-hydroxyheptadecatrienoic acid (HHT), 5-hydroxyeicosatetraenoic acid (5-HETE), and leukotriene B4 (LTB4) was inhibited for more than 80%. The release of superoxide anions and the chemiluminescence response (CL) upon opsonized zymosan stimulation did not differ from those observed in resident peritoneal macrophages. Additionally, the levels of cyclic nucleotides (cAMP and cGMP) were low in both resident and influx macrophages (day 3 p.i.). In contrast, peritoneal macrophages harvested on day 10 p.i. still showed a high level of phagocytosis. However, the intracellular level of cyclic AMP had decreased fivefold, whereas CL response and superoxide anion release were inhibited significantly. Moreover, the production of prostacyclin, LTB4, and 5-HETE was still suppressed in contrast to thromboxane synthesis, which has selectively been restored in these macrophages. A direct regulatory role of AA metabolites in changes in macrophage functions that were due to a RCMV infection could not be demonstrated.


Subject(s)
Arachidonic Acids/metabolism , Cytomegalovirus Infections/immunology , Macrophages/metabolism , Animals , Arachidonic Acid , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cytomegalovirus Infections/metabolism , Epoprostenol/biosynthesis , Fatty Acids, Unsaturated/biosynthesis , In Vitro Techniques , Luminescent Measurements , Macrophages/physiology , Male , Rats , Rats, Inbred Strains , Superoxides/metabolism , Thromboxane A2/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...