Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
J Virol ; 86(24): 13745-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23055562

ABSTRACT

Atherosclerosis is a major pathogenic factor in cardiovascular diseases, which are the leading cause of mortality in developed countries. While risk factors for atherosclerosis tend to be systemic, the distribution of atherosclerotic plaques within the vasculature is preferentially located at branch points and curves where blood flow is disturbed and shear stress is low. It is now widely accepted that hemodynamic factors can modulate endothelial gene expression and function and influence the pathophysiological changes associated with atherosclerosis. Human cytomegalovirus (HCMV), a ubiquitous pathogen, has long been proposed as a risk factor for atherosclerosis. To date, the role of HCMV in atherogenesis has been explored only in static conditions, and it is not known how HCMV infection is influenced by the physiological context of flow. In this study, we utilized a parallel-plate flow system to simulate the effects of shear stresses in different regions of the vasculature in vitro. We found that endothelial cells cultured under low shear stress, which simulates the flow condition of atheroprone regions in vivo, are more permissive to HCMV infection than cells experiencing high shear stress or static conditions. Cells exposed to low shear stress show increased entry of HCMV compared to cells exposed to high shear stress or static conditions. Viral structural gene expression, viral titers, and viral spread are also enhanced in endothelial cells exposed to low shear stress. These results suggest that hemodynamic factors modulate HCMV infection of endothelial cells, thus providing new insights into the induction/acceleration of atherosclerosis by HCMV.


Subject(s)
Arteries/virology , Cytomegalovirus/physiology , Endothelium, Vascular/virology , Membrane Fusion , Base Sequence , Blotting, Western , Cells, Cultured , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , DNA Primers , DNA, Viral/biosynthesis , Fluorescent Antibody Technique , Gene Expression Regulation, Viral , Genes, Viral , Humans , Microscopy, Electron, Transmission , Polymerase Chain Reaction
3.
Thromb Haemost ; 106(5): 858-67, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22012133

ABSTRACT

It is currently unclear what causes the chronic inflammation within atherosclerotic plaques. One emerging paradigm suggests that infection with bacteria and/or viruses can contribute to the pathogenesis of atherosclerosis either via direct infection of vascular cells or via the indirect effects of cytokines or acute phase proteins induced by infection at non-vascular sites. This paradigm has been supported by multiple epidemiological studies that have established positive associations between the risk of cardiovascular disease morbidity and mortality and markers of infection. It has also been supported by experimental studies showing an acceleration of the development of atherosclerosis following infection of hyperlipidaemic animal models. There are now a large number of different infectious agents that have been linked with an increased risk of cardiovascular disease. These include: Chlamydia pneumoniae, Porphyromonas gingivalis, Helicobacter pylori , influenza A virus, hepatitis C virus, cytomegalovirus, and human immunodeficiency virus. However, there are significant differences in the strength of the data supporting their association with cardiovascular disease pathogenesis. In some cases, the infectious agents are found within the plaques and viable organisms can be isolated suggesting a direct effect. In other cases, the association is entirely based on biomarkers. In the following review, we evaluate the strength of the data for individual or groups of pathogens with regard to atherosclerosis pathogenesis and their potential contribution by direct or indirect mechanisms and discuss whether the established associations are supportive of the infectious disease paradigm. We also discuss the failure of antibiotic trials and the question of persistent infection.


Subject(s)
Arteries/microbiology , Arteries/virology , Atherosclerosis/microbiology , Atherosclerosis/virology , Bacterial Infections/microbiology , Inflammation/microbiology , Inflammation/virology , Virus Diseases/virology , Animals , Anti-Bacterial Agents/therapeutic use , Arteries/immunology , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Bacterial Infections/complications , Bacterial Infections/drug therapy , Bacterial Infections/immunology , Evidence-Based Medicine , Humans , Inflammation/drug therapy , Inflammation/immunology , Risk Assessment , Risk Factors , Treatment Failure , Virus Diseases/complications , Virus Diseases/drug therapy , Virus Diseases/immunology
4.
Atherosclerosis ; 208(1): 90-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19665123

ABSTRACT

OBJECTIVE: Influenza can trigger heart attacks, and vaccination against influenza reduces the risk of cardiovascular events. Currently, it is believed that influenza virus in general does not disseminate to extra-pulmonary tissues. We assessed the vascular effects of influenza infection and whether the virus can directly infect atherosclerotic arteries in mice. METHODS/RESULTS: We intranasally infected 4 different types of mice--atherosclerotic apo E-deficient (our primary model), LDL receptor knockout, C57BL/6, and outbred Swiss--with influenza A/HK (H3/N2) virus. On day 7 after infection, we cultured viable virus from lung, aorta, and heart tissue, but not from the blood of apo E-deficient mice. Immunofluorescence studies showed influenza A virus NP1 protein and real time polymerase chain reaction (PCR) assay showed RNA in the aorta of infected apo E-deficient mice. Infected mice had significantly higher blood levels of chemokines and cytokines than control mice. At the local level, gene expression for several chemokines and cytokines was increased and eNOS expression was decreased. Infected mice had a higher density of macrophages in plaque than did control mice. CONCLUSIONS: We have shown for the first time that influenza virus can directly infect and reside in atherosclerotic arteries and that infection was associated with systemic and arterial-level pro-inflammatory changes.


Subject(s)
Atherosclerosis/virology , Orthomyxoviridae/pathogenicity , Animals , Arteries/virology , Inflammation/virology , Mice
5.
Am J Physiol Heart Circ Physiol ; 296(4): H937-45, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19181963

ABSTRACT

Human cytomegalovirus (CMV) is implicated in vascular complications through endothelial dysfunction. However, the effect of in vivo infections on vascular function in isolated arteries has not been examined. In pregnancy, systemic and uterine vascular adaptations accommodate increased blood volume through several mechanisms, including decreased sensitivity to vasoconstrictors and increased production of endothelial-dependent vasodilators. We hypothesized that an active in vivo CMV infection would reduce vasodilation of isolated arteries to the endothelial-dependent vasodilator methacholine and increase vasoconstriction to the alpha(1)-adrenergic receptor agonist phenylephrine and that these CMV-induced changes would be accentuated in late pregnancy. A mouse CMV infection model was used to study vascular responses in isolated mesenteric and uterine arteries from nonpregnant and late pregnant mice. In the mouse, CMV is not transmitted to the fetus. Accordingly, there was no evidence of active infection in any fetus examined, even though an active infection was found in salivary glands, uterine and mesenteric arteries, and placentas. Contrary to our hypothesis, increased endothelial-dependent vasodilation was found in mesenteric arteries from infected compared with uninfected nonpregnant and pregnant mice These data implicate active CMV infections in hypotensive disorders. Similarly, increased vasodilation was found in uterine arteries from infected vs. uninfected nonpregnant mice. However, this was completely reversed in infected compared with uninfected late pregnant mice in which vasodilation in uterine arteries was significantly reduced. Uterine arteries from infected pregnant mice also showed increased vasoconstriction to phenylephrine. Maternal infection led to decreased placental weights but had no effect on fetal weights in late pregnancy. These novel data demonstrate abnormal systemic and uterine vascular responses during an active CMV infection in both nonpregnant and late pregnant mice. Importantly, despite reduced placental weights, fetal weights were maintained, suggesting effective intrauterine compensation in the mouse model.


Subject(s)
Arteries/physiology , Arteries/virology , Cytomegalovirus Infections/physiopathology , Mesentery/blood supply , Uterus/blood supply , Vasoconstriction/physiology , Vasodilation/physiology , Animals , Arteries/drug effects , Cytomegalovirus , Disease Models, Animal , Female , Fetal Weight/physiology , Male , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Phenylephrine/pharmacology , Placenta/physiology , Pregnancy , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , beta-Galactosidase/metabolism
6.
Arch Med Res ; 40(7): 612-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20082878

ABSTRACT

BACKGROUND AND AIMS: The identification of possible pathogens for an infectious etiology of atherosclerotic coronary artery disease (CAD) is an expanding field. The present study was undertaken to explore the role of parvovirus B19, a potent infectious agent. METHODS: A total of 565 patients were analyzed (90 patients with CAD, and 475 controls). Serologic analysis for human paravovirus B19 (B19) specific IgM and IgG was carried out in all patients. In addition, tissue specimens were obtained from five patients who received heart transplants. Direct in situ polymerase chain reaction (PCR) and immunocytochemistry were performed in the samples to localize B19 DNA. RESULTS: Enzyme immunoassay showed that the seropositive rate of anti-B19 IgG in patients with CAD was 1.5- to 2.7-fold more frequent than in healthy controls. Clinical characteristics did not affect the prevalence of seropositivity for B19. However, anti-B19 IgM and B19-specific DNA were not detected in healthy or individuals with CAD. Furthermore, nonradioactive in situ PCR found that the majority of B19-specific DNA was located in the endothelial cells of the thickened intima. CONCLUSIONS: Our results first demonstrate a seroepidemiological and histopathological association between chronic B19 infection and CAD, suggesting that B19 infection may have a potential role in the pathogenesis of coronary atherosclerosis.


Subject(s)
Coronary Artery Disease , Parvoviridae Infections/complications , Parvovirus B19, Human/genetics , Adult , Aged , Antibodies, Viral/metabolism , Arteries/virology , Coronary Artery Disease/etiology , Coronary Artery Disease/virology , DNA, Viral/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Middle Aged , Parvoviridae Infections/blood , Parvovirus B19, Human/immunology
7.
Am J Pathol ; 172(4): 1100-11, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18310503

ABSTRACT

Human immunodeficiency virus (HIV) infection is associated with accelerated atherosclerosis and vasculopathy, although the mechanisms underlying these findings have not been determined. Hypotheses for these observations include: 1) an increase in the prevalence of established cardiac risk factors observed in HIV-infected individuals who are currently experiencing longer life expectancies; 2) the dyslipidemia reported with certain HIV anti-retroviral therapies; and/or 3) the proinflammatory effects of infiltrating HIV-infected monocytes/macrophages. An unexplored possibility is whether HIV itself can infect vascular smooth muscle cells (SMCs) and, by doing so, whether SMCs can accelerate vascular disease. Our studies demonstrate that human SMCs can be infected with HIV both in vivo and in vitro. The HIV protein p24 was detected by fluorescence confocal microscopy in SMCs from tissue sections of human atherosclerotic plaques obtained from HIV-infected individuals. Human SMCs could also be infected in vitro with HIV by a mechanism dependent on CD4, the chemokine receptors CXCR4 or CCR5, and endocytosis, resulting in a marked increase in SMC secretion of the chemokine CCL2/MCP-1, which has been previously shown to be a critical mediator of atherosclerosis. In addition, SMC proliferation appeared concentric to the vessel lumen, and minimal inflammation was detected, unlike typical atherosclerosis. Our data suggest that direct infection of human arterial SMCs by HIV represents a potential mechanism in a multifactorial paradigm to explain the exacerbated atherosclerosis and vasculopathy reported in individuals infected with HIV.


Subject(s)
Arteries/virology , HIV Infections/virology , HIV/physiology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/virology , Vascular Diseases/pathology , Vascular Diseases/virology , Adult , Aged , Arteries/pathology , Atherosclerosis/pathology , Atherosclerosis/virology , CD4 Antigens/metabolism , Chemokine CCL2/metabolism , Endocytosis , Female , HIV-1/physiology , Humans , Macrophages/virology , Male , Middle Aged , Monocytes/virology , Receptors, Chemokine/metabolism
9.
Am J Pathol ; 164(2): 589-600, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742264

ABSTRACT

Human cytomegalovirus (CMV) is a possible co-factor in atherogenesis and vascular occlusion, but its ability to actively infect medium and large blood vessels is unclear. A vascular explant model was adapted to investigate CMV infection in human coronary artery, internal mammary artery (IMA), and saphenous vein (SV). Vascular explants were inoculated with CMV Towne or low-passage clinical isolate and examined in situ for CMV cytopathic effect and immediate-early and early antigens, as indicators of active infection. At 5 to 7 days after inoculation, we found that CMV Towne actively infected eight of eight different atherosclerotic blood vessel explants (coronary artery, n = 4; SV and IMA grafts, n = 4), whereas it only infected 2 of 14 nonatherosclerotic blood vessel explants (SV, n = 10; IMA, n = 4) (P = 0.001). The CMV clinical isolate actively infected none of six sets of nonatherosclerotic SV explants at 5 to 7 days after inoculation. The active CMV infections involved adventitial and, less frequently, intimal cells. A small subset of infected cells in atherosclerotic tissue expresses the endothelial cell marker CD31. Smooth muscle cells residing in both atherosclerotic and nonatherosclerotic blood vessels were free of active CMV infections even after all vascular tissue layers were exposed to the virus. In contrast, active CMV Towne infection was evident at 2 days after inoculation in smooth muscle cells and endothelial cells previously isolated from the SV tissues. We conclude that active CMV infection is enhanced in atherosclerotic blood vessels compared to atherosclerosis-free vascular equivalents, and this viral activity is restricted to subpopulations of intimal and adventitial cells.


Subject(s)
Arteries/virology , Arteriosclerosis/virology , Cytomegalovirus Infections , Cytomegalovirus/pathogenicity , Saphenous Vein/virology , Animals , Humans , Immunohistochemistry , Organ Culture Techniques , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tunica Intima/virology
10.
Chin Med J (Engl) ; 114(11): 1208-10, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11729522

ABSTRACT

OBJECTIVE: To investigate the association of cylomegalovirus with atherosclerosis. METHODS: The presence of cylomegalovirus (CMV) nucleic acids was demonstrated in carotid and coronary arteries of patients with and without atherosclerosis by polymerase chain reaction (PCR). CMV was detected by PCR in the blood of patients with and without atherosclerosis. RESULTS: 83.3%-86.7% of the samples obtained from atherosclerotic patient arterial walls were shown to contain viral nucleic acids (CMV), CMV could be found among 6.7% of patients' arterial wall without atherosclerosis, significant difference can be found between them (P < 0.01). In blood CMV could be found in 42.4% of patients with atherosclerosis, in the control group, only 3% of samples had CMV, P < 0.01. CONCLUSION: CMV plays an important role in the pathologic process of the atherosclerosis and the atherosclerotic cerebral infarction.


Subject(s)
Arteries/virology , Arteriosclerosis/virology , Cytomegalovirus/isolation & purification , Leukocytes/virology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prevalence
11.
Mol Med ; 7(3): 169-76, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11471553

ABSTRACT

BACKGROUND: The pathogenesis of HIV-1-related cardiomyopathy is poorly understood, but HIV-1 has been detected in cardiomyocytes. Whether HIV-1 penetrates into the myocardium by infection of coronary artery endothelial cells (CAEC) or using transcellular or paracellular routes across CAEC has not been resolved. MATERIALS AND METHODS: A model of the CAEC barrier was constructed with primary CAEC (derived from human coronary vessels). Polymerase chain reaction (PCR) assay, infectious assay, and immunofluorescence were employed to show abortive nature of HIV-1 infection of CAEC. Tight junction (TJ) and cell adhesion proteins were visualized by immunofluorescence. The time course of HIV-1 invasion was measured by HIV-1 RNA assay. Inulin permeability assay determined paracellular leakage. Transmission electron microscopy demonstrated virus-induced endothelial vacuolization. RESULTS: Despite a strong display on CAEC of CXCR4 and a lesser expression of CCR3 and CCR5, HIV-1 did not productively replicate in CAEC, as shown by infectious assay, immunofluorescence, and electron microscopy. HIV-1 infection of CAEC was abortive with minimal reverse transcription of strong stop DNA and pol but not full-length or two LTR DNA circles. Upon infection of the model with 1 million RNA copies of HIV-1JR-FL, virus penetration 2 hr postinfection (PI) was negligible but increased by 1,750% 24 hr PI. The paracellular permeability increased during this period by only 25%. Neither AOP-RANTES nor v-MIPII significantly reduced HIV-1JR-FL invasion. Virus infection did not alter the integral TJ protein occludin and the TJ-associated protein ZO-1. HIV-1 exposed CAEC and brain microvascular endothelial cells (BMVEC) developed extensive cytoplasmic vacuolization with retroviral-like particles in the vacuoles. CONCLUSIONS: The endothelium is not an impenetrable barrier to HIV-1. The virus opens a transcellular route across coronary and brain endothelia in cytoplasmic vacuoles.


Subject(s)
Arteries/virology , Coronary Vessels/virology , Endocytosis , HIV-1/physiology , Arteries/ultrastructure , Base Sequence , Cells, Cultured , Chemokines/physiology , Coronary Vessels/ultrastructure , DNA Primers , Gene Products, pol/genetics , HIV Long Terminal Repeat , Microscopy, Electron , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Virus Replication
12.
Neuropathology ; 20 Suppl: S65-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11037191

ABSTRACT

HTLV-I was first described as a pathogenic human retrovirus that causes adult T-cell leukemia (ATL). Soon after the discovery of HTLV-I, an association of this virus with a slowly progressive neurological disorder was found independently in Japan and Caribbean islands, and this new clinical entity (HTLV-I-associated myelopathy with tropical spastic paraparesis) was named HAM/TSP. Autopsy findings clarified the chronic inflammatory nature of the disease. Detailed neuropathological analysis demonstrated: (i) T-cell-dominant mononuclear cell infiltration; (ii) diffuse and symmetrical degeneration of the anterolateral and inner portion of the posterior columns involving both myelin and axons; (iii) the presence of cytotoxic T cells and apoptosis of helper/inducer T cells; (iv) in vivo localization of HTLV-I provirus in the perivascular infiltrated T cells; and (v) accentuation of inflammatory lesions at the site with slow blood flow. From these findings it is suggested that a T-cell-mediated chronic inflammatory process targeting the HTLV-I-infected T cells is the primary pathogenic mechanism of HAM/TSP.


Subject(s)
Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , Arteries/immunology , Arteries/pathology , Arteries/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Japan , Paraparesis, Tropical Spastic/immunology , Spinal Cord/blood supply , Spinal Cord/pathology , Spinal Cord/virology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Helper-Inducer/virology
13.
Am Heart J ; 138(5 Pt 2): S465-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10539849

ABSTRACT

BACKGROUND: This research was suggested after crystals that we observed in herpesvirus-infected cell cultures were identified as cholesterol. Other reports and the development of defined reagents led us to select the use of Marek's disease herpesvirus (MDV) infection of chickens to demonstrate a potential role of herpesviruses in the pathogenesis of atherosclerosis. Available for our use were a clone-purified strain of MDV of known virulence, genetically selected, specific pathogen-free chickens, and appropriate isolation facilities to design controlled experiments to fulfill Koch's postulates. METHODS AND RESULTS: Experiments were performed to test the roles of both MDV and dietary cholesterol in atherosclerosis. The birds were examined 7 months after MDV infection with and without cholesterol feeding for gross and microscopic arterial lesions. Atherosclerotic lesions were found only in infected normocholesterolemic or hypercholesterolemic birds. The character and distribution of these lesions closely resembled those found in the chronic human arterial disease. Atherosclerotic lesions were not found in uninfected birds even if the birds were hypercholesterolemic. CONCLUSIONS: Evidence was obtained from other experiments that after MDV infection, cholesterol and cholesteryl esters accumulated in cell cultures and in atherosclerotic lesions. These changes were associated with altered enzymatic activity of the cholesterol synthesis cycle. Immunization with turkey herpesvirus vaccine or SB-1 vaccine prevented atherosclerotic lesions.


Subject(s)
Arteriosclerosis/veterinary , Chickens/virology , Herpesvirus 2, Gallid/pathogenicity , Marek Disease/complications , Animals , Antigens, Viral/immunology , Arteries/pathology , Arteries/virology , Arteriosclerosis/pathology , Arteriosclerosis/prevention & control , Arteriosclerosis/virology , Cells, Cultured , Cytomegalovirus/pathogenicity , Disease Models, Animal , Lipid Metabolism , Marek Disease/immunology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/virology , Specific Pathogen-Free Organisms , Viral Vaccines/administration & dosage
14.
Circulation ; 99(13): 1656-9, 1999 Apr 06.
Article in English | MEDLINE | ID: mdl-10190872

ABSTRACT

BACKGROUND: Restenotic and atherosclerotic lesions often contain smooth muscle cells (SMCs), which display high rates of proliferation and apoptosis. Human cytomegalovirus (HCMV) may increase the incidence of restenosis and predispose to atherosclerosis. Although the mechanisms contributing to these processes are unclear, studies demonstrate that one of the immediate-early (IE) gene products of HCMV, IE2-84, binds to and inhibits p53 transcriptional activity. Given the role of p53 in mediating apoptosis, we studied the ability of IE2-84 to inhibit p53-dependent apoptosis in human coronary artery SMCs. METHODS AND RESULTS: Apoptosis of SMCs was induced either by use of an adenovirus vector encoding human wild-type p53 protein or by treatment with doxorubicin. HCMV IE1-72 and IE2-84, the major IE proteins of HCMV, were overexpressed separately with adenovirus vectors encoding each protein, and the effects on p53-induced apoptosis were examined by both nick end-labeling (TUNEL) assay and flow cytometry. Expression of IE2-84, but not IE1-72, protected SMCs from p53-mediated apoptosis. CONCLUSIONS: These data indicate that an HCMV IE protein antagonizes p53-mediated apoptosis, suggesting a pathway by which HCMV infection predisposes to SMC accumulation and thereby contributes to restenosis and atherosclerosis.


Subject(s)
Apoptosis , Coronary Vessels/virology , Cytomegalovirus/physiology , Immediate-Early Proteins/physiology , Membrane Glycoproteins , Muscle, Smooth, Vascular/virology , Trans-Activators , Tumor Suppressor Protein p53/physiology , Viral Envelope Proteins , Viral Proteins , Apoptosis/drug effects , Arteries/drug effects , Arteries/metabolism , Arteries/virology , Blotting, Western , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Doxorubicin/pharmacology , Gene Expression , Genes, p53/genetics , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Transcription, Genetic
15.
Hum Gene Ther ; 8(9): 1033-40, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9189761

ABSTRACT

We aimed to investigate whether infection of normal rabbit arteries with a recombinant adenovirus vector would result per se in alterations in contractile and endothelial functions. In one group of rabbits, right or left femoral and ear artery segments were injected in vivo with a replication-deficient adenoviral vector expressing a beta-galactosidase (beta-Gal) reporter gene (4 x 10(10) pfu/ml) to demonstrate efficient gene transfer. Contralateral arteries were injected with the same concentration of a recombinant adenoviral vector carrying no transgene (Ad.MLPnull). In another group of animals, Ad.MLPnull was injected into the lumen of femoral and ear artery segments. The contralateral arteries were used as controls with the injection of vehicle alone. Histochemical assessment of gene transfer using beta-Gal activity (group 1) or in vitro contractility and endothelial function (group 2) was performed 3 days after adenoviral infection. Gene transfer was efficient and reproducible in the endothelium and was associated with the presence of inflammatory cells in the media. In Ad.MLPnull-injected arteries, in vitro contractile response of femoral artery rings to either KCl 60 mM or phenylephrine (10 microM) was reduced to 10.5 +/- 2.3% (n = 14; p < 0.001) and 8.8 +/- 2.0% (n = 7; p < 0.001) of the control values, respectively. Furthermore, in arteries injected with Ad.MLPnull, the endothelium-dependent relaxation produced by acetylcholine (10 microM) was virtually abolished. Similarly, the relaxant effects of the alpha 2-adrenoreceptor agonist UK14304 (1 microM) or the Ca2+ ionophore A23187 (1 microM) were also abolished. By contrast, sodium nitroprusside (10 microM) was still able to relax adenovirus-infected arteries. We conclude that infection with a recombinant adenoviral vector can induce early severe vasomotor alterations in both contractile function and endothelium-mediated relaxation of normal rabbit arteries.


Subject(s)
Adenoviridae/genetics , Arteries/physiopathology , Arteries/virology , Genetic Vectors/adverse effects , Vasomotor System/virology , Acetylcholine/pharmacology , Animals , Arteries/drug effects , Endothelium, Vascular/physiopathology , Endothelium, Vascular/virology , Gene Transfer Techniques , Membrane Potentials , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Potassium/pharmacology , Rabbits , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Vasomotor System/physiopathology
16.
Transplantation ; 63(9): 1366-9, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9158037

ABSTRACT

Human cytomegalovirus (CMV) has been associated with allograft rejection and, in particular, with transplant-associated arteriosclerosis. However, the role CMV plays in the development of transplant-associated arteriosclerosis remains unclear. CMV can infect the endothelium, the interface between allograft tissue and the host immune cells, but the direct induction of endothelial human leukocyte antigen (HLA) class II by CMV remains controversial. Our previous studies with venous endothelial cells (EC) have shown that CMV does not directly induce this antigen on infected EC and, furthermore, renders these cells refractory to interferon (IFN)-gamma induction. However, questions have arisen regarding the relevance of these findings to arterial endothelia. Thus, we have extended these studies to determine whether similar interactions occur in arterial EC. EC derived from human coronary artery, aorta, and umbilical artery were assayed by immunofluorescence flow cytometry and dual immunohistochemical staining following IFN-gamma treatment and/or inoculation with CMV. Data generated by these experiments demonstrate that regardless of vascular origin: (1) CMV does not directly induce endothelial surface or cytoplasmic HLA class II, and (2) although uninfected arterial EC are HLA class II inducible by IFN-gamma, infected cells are completely refractory to this effect. These results suggest that CMV-mediated inhibition of HLA class II expression is a phenomenon shared by human arterial and venous endothelia of both fetal and adult origin.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , Histocompatibility Antigens Class II/biosynthesis , Adult , Arteries/cytology , Arteries/metabolism , Arteries/virology , Arteriosclerosis/etiology , Cells, Cultured , Cytomegalovirus Infections/immunology , Humans , Transplantation/adverse effects
17.
J Clin Invest ; 98(1): 225-35, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8690797

ABSTRACT

Injury to atherosclerotic arteries induces the expression of growth regulatory genes that stimulate cellular proliferation and intimal formation. Intimal expansion has been reduced in vivo in nonatherosclerotic balloon-injured arteries by transfer of genes that inhibit cell proliferation. It is not known, however, whether vascular cell proliferation can be inhibited after injury in more extensively diseased atherosclerotic arteries. Accordingly, the purpose of this study was to investigate whether expression of recombinant genes in atherosclerotic arteries after balloon injury could inhibit intimal cell proliferation. To test this hypothesis, we examined the response to balloon injury in atherosclerotic rabbit arteries after gene transfer of herpesvirus thymidine kinase gene (tk) and administration of ganciclovir. Smooth muscle cells from hyperlipidemic rabbit arteries infected with adenoviral vectors encoding tk were sensitive to ganciclovir, and bystander killing was observed in vitro. In atherosclerotic arteries, a human placental alkaline phosphatase reporter gene was expressed in intimal and medial smooth muscle cells and macrophages, identifying these cells as targets for gene transfer. Expression of tk in balloon-injured hyperlipidemic rabbit arteries followed by ganciclovir treatment resulted in a 64% reduction in intimal cell proliferation 7 d after gene transfer (P = 0.004), and a 35-49% reduction in internal area 21 d after gene transfer, compared with five different control groups (P < 0.05). Replication of smooth muscle cells and macrophages was inhibited by tk expression and ganciclovir treatment. These findings indicate that transfer of a gene that inhibits cellular proliferation limits the intimal area in balloon-injured atherosclerotic arteries. Molecular approaches to the inhibition of cell proliferation in atherosclerotic arteries constitute a possible treatment for vascular proliferative diseases.


Subject(s)
Arteries/pathology , Arteriosclerosis/therapy , Catheterization/adverse effects , Cell Cycle/genetics , Gene Transfer Techniques , Adenoviridae/genetics , Animals , Antiviral Agents/pharmacology , Arteries/growth & development , Arteries/virology , Ganciclovir/pharmacology , Gene Expression , Genes, Reporter , Herpesviridae/enzymology , Herpesviridae/genetics , Humans , Muscle Development , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/pathology , Rabbits , Thymidine Kinase/genetics , Tunica Intima/growth & development , Tunica Intima/pathology
18.
Semin Interv Cardiol ; 1(1): 77-83, 1996 Mar.
Article in English | MEDLINE | ID: mdl-9552497

ABSTRACT

Vascular gene transfer is the introduction of foreign DNA into host cells within the vessel wall. Expression of recombinant genes within vascular cells has been demonstrated in normal, injured and atherosclerotic arteries. Transfer of genes with biological activity has provided insights into the role of specific genes in normal and pathological states. The development of gene transfer as a form of vascular therapy will require safe and effective vectors and intravascular delivery systems. Viral and non-viral vectors and several delivery catheters are being evaluated to determine their clinical applicability. The current applications of vascular gene transfer to cardiovascular diseases are the prevention of restenosis following arterial injury and induction of angiogenesis in occlusive vascular disease.


Subject(s)
Arteriosclerosis/therapy , Genetic Therapy , Animals , Arteries/pathology , Arteries/virology , Arteriosclerosis/etiology , Arteriosclerosis/genetics , Catheterization, Peripheral , DNA, Viral/genetics , Drug Delivery Systems , Gene Transfer Techniques , Genetic Vectors , Humans , Plasmids/genetics , Secondary Prevention , Viruses/genetics
19.
J Biol Chem ; 270(33): 19630-7, 1995 Aug 18.
Article in English | MEDLINE | ID: mdl-7642651

ABSTRACT

Herpesvirus infection of arterial smooth muscle cells has been shown to cause cholesteryl ester (CE) accumulation. However, the effects of human herpes simplex virus type 1 (HSV-1) infection on cholesterol binding and internalization, intracellular metabolism, and efflux have not been evaluated. In addition, the effects of viral infection on signal transduction pathways that impact upon cholesterol metabolism have not been studied. We show in studies reported herein that HSV-1 infection of arterial smooth muscle cells enhances low density lipoprotein (LDL) binding and uptake which parallels an increase in LDL receptor steady state mRNA levels and transcription of the LDL receptor gene. HSV-2 also increases CE synthesis and 3-hydroxy- 3-methylglutaryl-CoA reductase activity but concomitantly reduces CE hydrolysis and cholesterol efflux. Interestingly, this viral infection was associated with a time-dependent decrease in protein kinase A activity and an increase in viral-induced protein kinase (VPK) activity commensurate with the accumulation of esterified cholesterol. The relationship between increased VPK activity and alterations in CE accumulation in virally infected cells was explored using an HSV-1 VPK- mutant in which the portion of the HSV-1 genome encoding VPK had been deleted. Cholesteryl ester accumulation was significantly increased (> 50-fold) in HSV-1-infected cells compared to uninfected cells. However, the HSV-1 VPK- mutant had no significant effect on CE accumulation. The relationship between VPK activity and these alterations in cholesterol metabolism was further supported by the observation that staurosporine and calphostin C (protein kinase inhibitors) reduced protein kinase activity in HSV-1-infected cells. These results suggest several potential mechanisms by which alterations in kinase activities in response to HSV-1 infection of vascular cells may alter cholesterol trafficking processes that eventually lead to CE accumulation.


Subject(s)
Arteries/metabolism , Cholesterol, LDL/metabolism , Herpesvirus 1, Human/physiology , Protein Kinases/metabolism , Arteries/cytology , Arteries/virology , Biological Transport , Cells, Cultured , Cholesterol Esters/metabolism , Herpesvirus 1, Human/enzymology , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...