Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Vascul Pharmacol ; 137: 106823, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33232769

RESUMEN

Currently, the world is suffering from the pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses angiotensin-converting enzyme 2 (ACE2) as a receptor to enter the host cells. So far, 60 million people have been infected with SARS-CoV-2, and 1.4 million people have died because of COVID-19 worldwide, causing serious health, economical, and sociological problems. However, the mechanism of the effect of SARS-CoV-2 on human host cells has not been defined. The present study reports that the SARS-CoV-2 spike protein alone without the rest of the viral components is sufficient to elicit cell signaling in lung vascular cells. The treatment of human pulmonary artery smooth muscle cells or human pulmonary artery endothelial cells with recombinant SARS-CoV-2 spike protein S1 subunit (Val16 - Gln690) at 10 ng/ml (0.13 nM) caused an activation of MEK phosphorylation. The activation kinetics was transient with a peak at 10 min. The recombinant protein that contains only the ACE2 receptor-binding domain of the SARS-CoV-2 spike protein S1 subunit (Arg319 - Phe541), on the other hand, did not cause this activation. Consistent with the activation of cell growth signaling in lung vascular cells by the SARS-CoV-2 spike protein, pulmonary vascular walls were found to be thickened in COVID-19 patients. Thus, SARS-CoV-2 spike protein-mediated cell growth signaling may participate in adverse cardiovascular/pulmonary outcomes, and this mechanism may provide new therapeutic targets to combat COVID-19.


Asunto(s)
COVID-19/metabolismo , Células Endoteliales/metabolismo , Pulmón/irrigación sanguínea , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/patología , COVID-19/virología , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/virología , Interacciones Huésped-Patógeno , Humanos , Cinética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/virología , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/virología , Receptores Virales/metabolismo , SARS-CoV-2/patogenicidad , Transducción de Señal
2.
J Dent Res ; 97(11): 1214-1221, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29775416

RESUMEN

The nitric oxide (NO)-sensitive soluble guanylyl cyclase (sGC) is a heterodimeric enzyme with an α and ß subunit. NO binds to heme of the ß1-subunit of sGC, activates the enzyme in the reduced heme iron state in vascular smooth muscle cells (VSMCs), and generates cGMP-inducing vasodilatation and suppression of VSMC proliferation. In the complex tumor milieu with higher levels of reactive oxygen species (ROS), sGC heme iron may become oxidized and insensitive to NO. To change sGC from an NO-insensitive to NO-sensitive state or NO-independent manner, protein expression of sGC in VSMC is required. Whether sGCα1ß1 exists at the protein level in arterial VSMCs of oropharyngeal squamous cell carcinoma (OPSCC) is unknown. In addition, whether differences in the genetic profile between human papillomavirus (HPV)-positive and HPV-negative OPSCC contributes to the regulation of sGCα1ß1 is unclear. Therefore, we compared the effects of HPV-positive and HPV-negative OPSCC on the expression of sGCα1ß1 in arterial VSMCs from tumor-free and tumor-containing regions of human tissue sections using quantitative immunohistochemistry. In comparison to the tumor-free region, we found a decrease in expression of both α1- and ß1-subunits in the arterial VSMC layer of the tumor-containing areas. The OPSCC-induced significant downregulation of the α1- and ß1-subunits of sGC in arterial VSMC was HPV-independent. We conclude that the response of sGC to NO in tumor arterial VSMCs may be impaired by oxidation of the heme of the ß1-subunit, and thus, α1- and ß1-subunits of sGC could be targeted to degradation under oxidative stress in OPSCC in an HPV-independent manner. The degradation of sGCα1ß1 in VSMCs may result in increased proliferation of VSMCs, promoting tumor arteriogenesis in OPSCC. This can be interrupted by preserving the active heterodimer sGCα1ß1 in arterial VSMCs.


Asunto(s)
Carcinoma de Células Escamosas/irrigación sanguínea , Músculo Liso Vascular/virología , Neoplasias Orofaríngeas/irrigación sanguínea , Infecciones por Papillomavirus/metabolismo , Guanilil Ciclasa Soluble/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virología , Estudios de Casos y Controles , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Neovascularización Patológica/metabolismo , Óxido Nítrico/metabolismo , Neoplasias Orofaríngeas/enzimología , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/enzimología , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo
3.
Atherosclerosis ; 271: 111-119, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29486395

RESUMEN

BACKGROUND AND AIMS: Despite the potential life-threatening consequences of thoracic aortic aneurysms (TAAs), the pathogenesis of these diseases is still poorly understood. While some aspects of TAA formation have been elucidated, the role of vascular smooth muscle cells (SMCs) in both bicuspid aortic valve (BAV)-associated and degenerative tricuspid aortic valve (TAV)-associated TAAs has not yet been fully unravelled. Thus, this work was aimed at uncovering processes in SMC biology that may contribute to TAA formation. METHODS: Using isolated SMCs and tissue samples from TAAs linked to BAV syndrome, TAV-associated degenerative TAAs and control aortas, we performed targeted mRNA expression profile analyses and conducted immunohistological analyses on aortic wall tissue sections. RESULTS: While SMC expression profiles and tissue analyses in TAV-TAAs clearly point toward a pro-proliferative state of the aortic media SMCs, BAV-TAA SMCs and tissue provide evidence for DNA damage, DNA damage response signalling as well as profound TLR-3 signalling. CONCLUSIONS: The data presented in this study emphasizes the importance of SMCs in TAA development. Furthermore, our results provide evidence that the state of SMCs in the BAV-TAA (senescent) and TAV-TAA (pro-proliferative) differs significantly. For the first time, we also present findings that may argue for the occurrence of a viral infection in BAV-TAA SMCs.


Asunto(s)
Aorta Torácica/patología , Aneurisma de la Aorta Torácica/genética , Válvula Aórtica/anomalías , Proliferación Celular , Senescencia Celular , Enfermedades de las Válvulas Cardíacas/genética , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Transcriptoma , Virosis/virología , Adulto , Anciano , Aorta Torácica/metabolismo , Aorta Torácica/virología , Aneurisma de la Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/patología , Aneurisma de la Aorta Torácica/virología , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Válvula Aórtica/virología , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Casos y Controles , Proliferación Celular/genética , Células Cultivadas , Senescencia Celular/genética , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Enfermedades de las Válvulas Cardíacas/virología , Interacciones Huésped-Patógeno , Humanos , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Riesgo , Virosis/metabolismo , Virosis/patología
4.
Transplantation ; 101(3): 531-540, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27362315

RESUMEN

BACKGROUND: Renal transplantation is the preferred treatment for patients with end-stage renal disease. Human cytomegalovirus (HCMV) activation is associated with decreased renal graft function and survival. Human cytomegalovirus encodes several immune modulatory proteins, including the G protein-coupled receptor US28, which scavenges human chemokines and modulates intracellular signaling. METHODS: Our aim was to identify the expression and localization of US28 in renal allograft biopsies by immunohistochemistry and determine its role in viral spreading in vitro. RESULTS: Immunohistochemistry revealed US28 in 31 of 34 renal transplant biopsies from HCMV-seropositive donors. Expression was independent of HCMV viremia or IgG serostatus. US28 was predominantly expressed in the cytoplasm of vascular smooth muscle cells (VSMCs) and tubular epithelial cells, with a median positivity of 20% and 40%, respectively. Also, US28-positive cells were present within arterial neointima. In contrast to US28, HCMV-encoded immediate early antigen was detected in less than 5% of VSMCs, tubular epithelial cells, interstitial endothelium, interstitial inflammatory infiltrates, and glomerular cells.Primary VSMCs were infected with green fluorescent protein-tagged wild type or US28-deficient HCMV. The viral spreading of US28-deficient HCMV, via culture medium or cell-to-cell transmission, was significantly impeded as shown by green fluorescent protein (ie, infected) cell quantification and quantitative real-time polymerase chain reaction. Additionally, the number and size of foci was smaller. CONCLUSIONS: In summary, HCMV-encoded US28 was detected in renal allografts from HCMV-positive donors independent of viremia and serostatus. Also, US28 facilitates HCMV spreading in VSMCs in vitro. Because the vasculature is affected in chronic renal transplant dysfunction, US28 may provide a potential target for therapeutic intervention.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Trasplante de Riñón/efectos adversos , Riñón/metabolismo , Receptores de Quimiocina/metabolismo , Donantes de Tejidos , Proteínas Virales/metabolismo , Adulto , Anciano , Aloinjertos , Anticuerpos Antivirales/sangre , Biomarcadores/sangre , Biopsia , Células Cultivadas , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunohistoquímica , Riñón/inmunología , Riñón/cirugía , Riñón/virología , Trasplante de Riñón/métodos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/virología , Receptores de Quimiocina/inmunología , Estudios Retrospectivos , Factores de Tiempo , Proteínas Virales/inmunología , Virulencia , Adulto Joven
5.
Cell Physiol Biochem ; 39(5): 1804-1812, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27744449

RESUMEN

BACKGROUND: The present study was designed to observe the infection of human cytomegalovirus (HCMV) to human vascular smooth muscle cells (VSMCs), and the effect of viral infection on lipid metabolism in VSMCs. METHODS: The cytopathic effects were observed by inverted microscopy and viral infection were examined by electron microscopy and RT-PCR. The lipid metabolism related gene profiling of VSMCs after HCMV infection was assayed by cDNA assay and the abnormal expression of genes were validated by quantitative RT-PCR. The content of cholesterol in VSMCs after HCMV infection was assayed by cholesterol detection kit. RESULTS: VSMCs showed obvious cytopathic effects after HCMV infection. Intact viral particles could be detected in VSMCs using electron microscope. By use of RT-PCR technology, IE gene of HCMV could be amplified from VSMCs. The expression of cell lipid metabolism related gene profiling showed obvious disorders. The expression levels of HMG-CoA synthase and HMG-CoA reductase after infection increased significantly. The cellular cholesterol content (µmol/106 cells) was significantly higher than that of mock infected group at 72h post infection. CONCLUSION: HCMV can infect VSMCs and the infection can affect cellular lipid metabolism related gene expression, which get involved in the occurrence and development of atherosclerosis (AS).


Asunto(s)
Colesterol/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Metabolismo de los Lípidos/genética , Miocitos del Músculo Liso/metabolismo , Oxidorreductasas/genética , Células Cultivadas , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Hidroximetilglutaril-CoA Sintasa/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología , Oxidorreductasas/metabolismo , Transducción de Señal
6.
PLoS One ; 10(7): e0130264, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26132411

RESUMEN

Intimal hyperplasia (IH) is the primary cause of vein bypass graft failure. The smooth muscle cell (SMC) is a key element of IH as it phenotypically switches from a contractile to a synthetic state which can become pathological. R7020, which is an engineered strain of Herpes Simplex Virus-1, inhibits IH in animal models. Although it has many characteristics which make it a strong candidate for use as a prophylactic agent how it inhibits IH is not well understood. The objective of this study was to identify modes of action used by R7020 to function in blood vessels that may also contribute to its inhibition of IH. The cytopathic effect of R7020 on SMCs was determined in vitro and in a rabbit IH model. In vitro assays with R7020 infected SMCs were used to quantify the effect of dose on the release kinetics of the virus as well as the effects of R7020 on cell viability and the adhesion of peripheral blood mononuclear cells (PBMCs) to SMCs in the absence and presence of tumor necrosis factor alpha (TNF-α). The observed cytopathic effect, which included R7020 positive filopodia that extend from cell to cell and the formation of syncytia, suggests that R7020 remains cell associated after egress and spreads cell to cell instead of by diffusion through the extracellular fluid. This would allow the virus to rapidly infect vascular cells while evading the immune system. The directionality of the filopodia in vivo suggests that the virus preferentially travels from the media towards the intima targeting SMCs that would lead to IH. The formation of syncytia would inhibit SMC proliferation as incorporated cells are not able to multiply. It was also observed that R7020 induced the fusion of PBMCs with syncytia suggesting the virus may limit the effect of macrophages on IH. Furthermore, R7020 inhibited the proliferative effect of TNF-α, an inflammatory cytokine associated with increased IH. Thus, the results of this study suggest that R7020 inhibits IH through multiple mechanisms.


Asunto(s)
Proliferación Celular , Herpesvirus Humano 1/fisiología , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología , Túnica Íntima/virología , Animales , Movimiento Celular , Células Cultivadas , Herpesvirus Humano 1/patogenicidad , Humanos , Hiperplasia/virología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Conejos , Túnica Íntima/patología
7.
Basic Res Cardiol ; 109(2): 401, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24442486

RESUMEN

Both human cytomegalovirus (HCMV) and arginase II (ARG II) have been implicated in the pathogenesis of cardiovascular diseases. The effects of HCMV on ARG II are unknown. The aim of this study was to investigate the effects of HCMV on ARG II expression in endothelial and vascular smooth muscle cells (SMC) both in vitro and ex vivo. Endothelial and SMC were infected with either HCMV or UV-irradiated HCMV. Expression of ARG II, endothelial or inducible nitric oxide synthase (eNOS and iNOS, respectively) and viral immediate early (IE) was quantified using quantitative PCR. Ganciclovir and short interfering RNA were used to determine the viral gene mediating the effects on ARG II. Detection of viral antigens and ARG II expression was performed by immunofluorescence or immunohistochemistry. HCMV infection increased both ARG II mRNA and protein levels in the examined cells; this effect was mediated by the HCMV IE2-p86 protein. The upregulation of ARG II was accompanied by a downregulation of eNOS but an induction of iNOS in HCMV-infected endothelial cells. Both eNOS and iNOS expressions were induced in HCMV-infected SMC. ARG II was abundantly expressed in endothelial cells, foam cells and SMC and was importantly significantly upregulated in HCMV-immunoreactive human carotid atherosclerotic plaques. HCMV IE2-p86 mediates ARG II upregulation in vitro and ARG II is co-expressed with HCMV antigens in human carotid atherosclerotic plaques. We speculate that HCMV may contribute to endothelial dysfunction via ARG II induction and reduced eNOS production.


Asunto(s)
Arginasa/genética , Enfermedades de las Arterias Carótidas/virología , Infecciones por Citomegalovirus/complicaciones , Citomegalovirus/genética , Vasculitis/enzimología , Vasculitis/virología , Antivirales/farmacología , Aorta/citología , Aorta/virología , Arginasa/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/patología , Células Endoteliales/citología , Células Endoteliales/virología , Ganciclovir/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Inmediatas-Precoces/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/virología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/virología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Transactivadores/genética , Regulación hacia Arriba/genética , Vasculitis/patología
8.
Gene Ther ; 20(12): 1158-64, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24005577

RESUMEN

Ex vivo gene therapy during coronary artery bypass grafting (CABG) holds great potential to prevent excessive smooth muscle cell (SMC) proliferation, neointima formation and graft failure. The most successful preclinical strategies to date have utilised vectors based on the species C adenovirus, Ad5, which engages the Coxsackie and Adenovirus receptor (CAR) as its primary attachment receptor. Profiling receptors on human SMCs demonstrated the absence of CAR but substantial expression of the species B receptor CD46. We performed transduction experiments using Ad5 and the CD46-utilising adenovirus Ad35, and found Ad35 significantly more efficient at transducing SMCs. To evaluate whether transduction could be further augmented, we evaluated chimeric CD46-utilising Ad5/Ad35 vectors comprising the Ad5 capsid pseudotyped with the Ad35 fibre alone (Ad5/F35) or in combination with the Ad35 penton (Ad5/F35/P35). In human smooth muscle cells (hSMCs), Ad5/F35/P35 mediated significantly higher levels of transduction than either parental vector or Ad5/F35. Ex vivo transduction experiments using mouse aortas from CD46 transgenics demonstrated that Ad5/F35/P35 was significantly more efficient at transducing SMCs than the other vectors tested. Finally, ex vivo transduction and immunofluorescent colocalisation experiments using human tissue from CABG procedures confirmed the preclinical potential of Ad5/F35/P35 as an efficient vector for vascular transduction during CABG.


Asunto(s)
Adenovirus Humanos/genética , Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Transducción Genética , Adenovirus Humanos/clasificación , Animales , Aorta , Células Cultivadas , Puente de Arteria Coronaria , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/cirugía , Enfermedad de la Arteria Coronaria/terapia , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Terapia Genética , Vectores Genéticos , Humanos , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/virología , Neointima/patología , Neointima/terapia
9.
PLoS Pathog ; 9(7): e1003470, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874198

RESUMEN

Hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) are diseases caused by hantavirus infections and are characterized by vascular leakage due to alterations of the endothelial barrier. Hantavirus-infected endothelial cells (EC) display no overt cytopathology; consequently, pathogenesis models have focused either on the influx of immune cells and release of cytokines or on increased degradation of the adherens junction protein, vascular endothelial (VE)-cadherin, due to hantavirus-mediated hypersensitization of EC to vascular endothelial growth factor (VEGF). To examine endothelial leakage in a relevant in vitro system, we co-cultured endothelial and vascular smooth muscle cells (vSMC) to generate capillary blood vessel-like structures. In contrast to results obtained in monolayers of cultured EC, we found that despite viral replication in both cell types as well as the presence of VEGF, infected in vitro vessels neither lost integrity nor displayed evidence of VE-cadherin degradation. Here, we present evidence for a novel mechanism of hantavirus-induced vascular leakage involving activation of the plasma kallikrein-kinin system (KKS). We show that incubation of factor XII (FXII), prekallikrein (PK), and high molecular weight kininogen (HK) plasma proteins with hantavirus-infected EC results in increased cleavage of HK, higher enzymatic activities of FXIIa/kallikrein (KAL) and increased liberation of bradykinin (BK). Measuring cell permeability in real-time using electric cell-substrate impedance sensing (ECIS), we identified dramatic increases in endothelial cell permeability after KKS activation and liberation of BK. Furthermore, the alterations in permeability could be prevented using inhibitors that directly block BK binding, the activity of FXIIa, or the activity of KAL. Lastly, FXII binding and autoactivation is increased on the surface of hantavirus-infected EC. These data are the first to demonstrate KKS activation during hantavirus infection and could have profound implications for treatment of hantavirus infections.


Asunto(s)
Capilares/virología , Permeabilidad Capilar , Endotelio Vascular/virología , Activación Enzimática , Factor XII/metabolismo , Infecciones por Hantavirus/virología , Sistema Calicreína-Quinina , Bradiquinina/antagonistas & inhibidores , Bradiquinina/metabolismo , Capilares/efectos de los fármacos , Capilares/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Orthohantavirus/fisiología , Infecciones por Hantavirus/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógeno de Alto Peso Molecular/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/virología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Precalicreína/antagonistas & inhibidores , Precalicreína/metabolismo , Inhibidores de Proteasas/farmacología , Proteolisis/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/virología , Propiedades de Superficie , Replicación Viral
10.
Biochem Biophys Res Commun ; 426(4): 486-91, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22960172

RESUMEN

Pulmonary arterial hypertension (PAH) is a rare but fatal condition in which raised pulmonary vascular resistance leads to right heart failure and death. Endothelin-1 is a potent endogenous vasoconstrictor, which is considered to be central to many of the events that lead to PAH, and is an important therapeutic target in the treatment of the condition. In many cases of PAH, the aetiology is unknown but inflammation is increasingly thought to play an important role and viruses have been implicated in the development of disease. The Toll Like Receptors (TLRs) play a key role in innate immune responses by initiating specific anti-bacterial and anti-viral defences in recognition of signature molecular motifs on the surface of invading pathogens. In this study, we set out to examine the expression of bacterial and viral TLRs in human pulmonary artery smooth muscle cells and to establish whether their activation could be relevant to PAH. We found that the viral TLR3 and bacterial TLRs 4 and 6 were most abundantly expressed in human pulmonary artery smooth muscle cells. Using specific TLR ligands, we found that activation of TLRs 3 and 4 resulted in IL-8 release by human pulmonary artery smooth muscle cells but that only TLR3 stimulation resulted in IP10 and endothelin-1 release. These data suggest that human pulmonary artery smooth muscle cells express significant levels of viral TLR3 and respond to its activation by releasing endothelin-1. This may have importance in understanding the association between viruses and the development of PAH.


Asunto(s)
Endotelina-1/biosíntesis , Hipertensión Pulmonar/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor Toll-Like 3/metabolismo , Células Cultivadas , Quimiocinas/genética , Citocinas/genética , Hipertensión Pulmonar Primaria Familiar , Expresión Génica , Humanos , Hipertensión Pulmonar/virología , Interleucina-8/genética , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología , Poli I-C/farmacología , Receptores de Citocinas/genética , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/genética , Factor de Necrosis Tumoral alfa/farmacología
11.
Cell Signal ; 24(10): 1909-17, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22709828

RESUMEN

OBJECTIVE: In our previous study, we have demonstrated that tissue factor pathway inhibitor (TFPI) gene could induce vascular smooth muscle cell (VSMC) apoptosis. This study was conducted to investigate whether the overexpression of the TFPI gene can induce VSMC apoptosis by inhibiting JAK-2/STAT-3 pathway phosphorylation and thereby inhibiting the expression of such downstream targets as the apoptotic protein Bcl-2 and cell cycle protein cyclin D1. The effect of TFPI on the expression of survivin, a central molecule in cell survival, was also investigated. METHODS: Rat VSMCs were infected with recombinant adenovirus containing either the TFPI (Ad-TFPI) or LacZ (Ad-LacZ) gene or DMEM in vitro. TFPI expression was detected by ELISA. TUNEL staining and electron microscope were carried out to determine the apoptosis of VSMCs. The expression levels of JAK-2, p-JAK-2, STAT-3, p-STAT-3, cyclin D1, Bcl-2 and survivin were examined by western blot analysis. RESULTS: TFPI protein was detected in the TFPI group after gene transfer and the peak expression was at the 3rd day. At the 3rd, 5th and 7th days after gene transfer, the apoptotic rates by TUNEL assay in the TFPI group were 10.91 ± 1.66%, 13.46 ± 1.28% and 17.04 ± 1.95%, respectively, whereas those in the LacZ group were 3.28 ± 0.89%, 4.01 ± 0.72% and 4.89 ± 1.17%, respectively. We observed cell contraction, slight mitochondrial swelling, nuclear pyknosis and apoptotic body formation in TFPI-treated VSMCs using electron microscopy. JAK-2, p-JAK-2, STAT-3, p-STAT-3, cyclin D1 and Bcl-2, which are all involved in the JAK-2/STAT-3 pathway, were detected in the VSMCs on the 3rd, 5th and 7th days after gene transfer, which is consistent with previously demonstrated time points when VSMCs apoptosis occurred. The expression levels of p-JAK-2, p-STAT-3, cyclin D1 and Bcl-2 were significantly decreased over time in the TFPI group (each P<0.05) but not in the Ad-LacZ and DMEM groups. However, this attenuation of expression was not observed for JAK-2 and STAT-3 in any of the groups at any time points after gene transfer (each P>0.05). The expression level of survivin in the TFPI group also weakened significantly over time compared with the levels in the Ad-LacZ and DMEM groups (each P<0.05) at the 3rd, 5th and 7th days after gene transfer. CONCLUSION: The results demonstrated that TFPI played an apoptosis-inducing role in VSMCs in a manner that involves both the suppression of JAK-2/STAT-3 pathway phosphorylation and the down-regulation of survivin. Our data show for the first time that targeting the JAK-2/STAT-3 pathway and survivin by overexpressing TFPI may be a new avenue for the treatment of restenosis.


Asunto(s)
Adenoviridae/fisiología , Apoptosis , Interacciones Huésped-Patógeno , Lipoproteínas/genética , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/virología , Adenoviridae/genética , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/patología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Lipoproteínas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Ratas , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Survivin , Regulación hacia Arriba
12.
Hypertension ; 56(2): 247-52, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20530298

RESUMEN

Calcium is an essential signaling molecule that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and differentiation. Here, we show that the calcium antagonist nifedipine inhibits VSMC dedifferentiation in vitro and in vivo. Differentiated VSMCs cultured on laminin-coated dishes were transferred to laminin-free dishes to induce dedifferentiation. Induction of dedifferentiation resulted in the upregulation of nonmuscle myosin heavy chain expression, a marker of dedifferentiation, and the downregulation of smooth muscle myosin heavy chain expression, a marker of differentiation. Nifedipine significantly inhibited both the induction of these phenotypic changes and upregulation of Akt signaling in these cells. Administration of nifedipine at a low concentration that did not affect blood pressure could inhibit the increase in nonmuscle myosin heavy chain expression and decrease in smooth muscle myosin heavy chain expression in a rat balloon-injury model. Furthermore, nifedipine suppressed neointimal hyperplasia and upregulation of Akt signaling. However, phospho-Akt expression was not suppressed in the regenerating arterial endothelium of the nifedipine-treated rats. The inhibitory effect of the downregulation of Akt signaling by dominant-negative Akt on the induction of VSMC dedifferentiation in the intima was identical to that of nifedipine. In contrast, upregulation of Akt signaling by transfection of the cells with a constitutively active Akt reversed the nifedipine-induced inhibition of VSMC dedifferentiation. In conclusion, nifedipine inhibits VSMC dedifferentiation by suppressing Akt signaling, thereby preventing neointimal thickening.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Nifedipino/farmacología , Proteínas Proto-Oncogénicas c-akt/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/fisiología , Aorta/virología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/virología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Túnica Íntima/efectos de los fármacos , Túnica Íntima/fisiología , Regulación hacia Arriba/efectos de los fármacos , Vasodilatadores/farmacología
13.
Neurol Res ; 30(9): 903-9, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18534055

RESUMEN

OBJECTIVES: To explore the role of angiotensin receptors (AT1 and AT2) and human cytomegalovirus (HCMV) infection in atherosclerosis, and to observe effect of HCMV UL83 gene on angiotensin receptor AT receptors. METHODS: Cell model of RNA interference (RNAi) HCMV UL83 gene was set up by transfection RNAi into HCMV infected human cerebral artery vascular smooth muscle cells cultured in vitro by application of transfection technique and adoption of RT-PCR, etc. Effect of HCMV on gene expression of AT1 and AT2 receptors in human cerebral artery vascular smooth muscle cells cultured in vitro under the action of Ganciclovir (GCV), AT1 and AT2 receptor antagonists by immunofluorescence technique, RT-PCR and protein electrophoresis. RESULTS: The duplication and protein expression of HCMV UL83 gene mRNA was successfully interfered using RNAi technique. After silencing HCMV UL83 gene, gene expression of AT1 receptor was down-regulated while that of AT2 was up-regulated. AT1 and AT2 receptor antagonists and GCV, under certain condition, could inhibit the activation on AT1 and AT2 receptor in human cerebral artery vascular smooth muscle cells induced by HCMV infection. CONCLUSION: Cell model of HCMV infected UL83 silence gene was successfully set up in human cerebral artery vascular smooth muscle cells cultured in vitro. HCMV UL83 might play a role by activating AT1 receptor and inhibiting expression of AT2 receptor.


Asunto(s)
Citomegalovirus/genética , Músculo Liso Vascular/metabolismo , Fosfoproteínas/genética , Interferencia de ARN , Receptores de Angiotensina/genética , Proteínas de la Matriz Viral/genética , Aterosclerosis/metabolismo , Aterosclerosis/virología , Western Blotting , Células Cultivadas , Arterias Cerebrales/citología , Citomegalovirus/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/virología , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/metabolismo , Receptores de Angiotensina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección/métodos
14.
J Exp Med ; 205(1): 19-24, 2008 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-18180307

RESUMEN

Leukotrienes (LTs) are powerful proinflammatory lipid mediators that may play a central role in cardiovascular diseases, including arteriosclerosis, myocardial infarction, and stroke. Owing to restricted expression of 5-lipoxygenase (5-LO), the enzyme required for their synthesis, LTs are almost exclusively produced by myeloid cells. Here, we report that human cytomegalovirus (HCMV) infection of human vascular smooth muscle cells (SMCs) increases 5-LO mRNA levels by up to 170-fold in a dose- and time-dependent manner. Infected cells expressed 5-LO protein, as shown by immunohistochemistry, enabling them to synthesize bioactive LTB(4). HCMV-infected vascular SMCs expressing 5-LO protein were readily detected in tissue samples from CMV-infected patients with inflammatory bowel disease or AIDS. Thus, pathogen-induced LT production in HCMV-infected tissues may contribute to local inflammation, consistent with the ability of HCMV to control cellular and immunological functions. HCMV-induced LT biosynthesis in SMCs offers a molecular mechanism to explain HCMV-induced pathogenesis in inflammatory diseases.


Asunto(s)
Araquidonato 5-Lipooxigenasa/biosíntesis , Infecciones por Citomegalovirus/metabolismo , Regulación de la Expresión Génica , Leucotrieno B4/biosíntesis , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Células Cultivadas , Fibroblastos/virología , Humanos , Técnicas para Inmunoenzimas , Inmunohistoquímica , Inflamación , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología
15.
Cell Signal ; 19(10): 2127-37, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17669626

RESUMEN

Enterovirus 71 (EV71) is a widespread virus that causes severe and fatal diseases in patients, including circulation failure. The mechanisms underlying EV71-initiated intracellular signaling pathways to influence host cell functions remain unknown. In this study, we identified a requirement for PDGFR, PI3-K/Akt, p38 MAPK, JNK, and NF-kappaB in the regulation of VCAM-1 expression by rat vascular smooth muscle cells (VSMCs) in response to viral infection. EV71 induced VCAM-1 expression in a time- and viral concentration-dependent manner. Infection of VSMCs with EV71 stimulated VCAM-1 expression and phosphorylation of PDGFR, Akt, and p38 MAPK which were attenuated by AG1296, wortmannin, and SB202190, respectively. The phosphorylation of JNK stimulated by EV71 was not detected under present conditions. In contrast, JNK inhibitor SP600125 inhibited EV71-induced VCAM-1 expression. Furthermore, VCAM-1 expression induced by EV71 was significantly attenuated by a selective NF-kappaB inhibitor (helenalin). Consistently, EV71-stimulated translocation of NF-kappaB into the nucleus and degradation of IkappaB-alpha as well as VCAM-1 mRNA expression was blocked by helenalin, AG1296, SB202190, SP600125, wortmannin, and LY294002. Moreover, the involvement of p38 MAPK, PI3-K/Akt, and NF-kappaB in EV71-induced VCAM-1 expression was reveled by that transfection with dominant negative plasmids of p38 MAPK, p85, Akt, NIK, IKK-alpha, and IKK-beta attenuated these responses. These findings suggest that in VSMCs, EV71-induced VCAM-1 expression was mediated through activation of PDGFR, PI3-K/Akt, p38 MAPK, JNK, and NF-kappaB pathways.


Asunto(s)
Enterovirus/fisiología , Músculo Liso Vascular/virología , Transducción de Señal , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Animales , Adhesión Celular , Células Cultivadas , Expresión Génica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Neutrófilos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Virol ; 81(8): 3816-26, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17251289

RESUMEN

Rat cytomegalovirus (RCMV) is a beta-herpesvirus with a 230-kbp genome containing over 167 open reading frames (ORFs). RCMV gene expression is tightly regulated in cultured cells, occurring in three distinct kinetic classes (immediate early, early, and late). However, the extent of viral-gene expression in vivo and its relationship to the in vitro expression are unknown. In this study, we used RCMV-specific DNA microarrays to investigate the viral transcriptional profiles in cultured, RCMV-infected endothelial cells, fibroblasts, and aortic smooth muscle cells and to compare these profiles to those found in tissues from RCMV-infected rat heart transplant recipients. In cultured cells, RCMV expresses approximately 95% of the known viral ORFs with few differences between cell types. By contrast, in vivo viral-gene expression in tissues from rat heart allograft recipients is highly restricted. In the tissues studied, a total of 80 viral genes expressing levels twice above background (5,000 to 10,000 copies per mug total RNA) were detected. In each tissue type, there were a number of genes expressed exclusively in that tissue. Although viral mRNA and genomic DNA levels were lower in the spleen than in submandibular glands, the number of individual viral genes expressed was higher in the spleen (60 versus 41). This finding suggests that the number of viral genes expressed is specific to a given tissue and is not dependent upon the viral load or viral mRNA levels. Our results demonstrate that the profiles, as well as the amplitude, of viral-gene expression are tissue specific and are dramatically different from those in infected cultured cells, indicating that RCMV gene expression in vitro does not reflect viral-gene expression in vivo.


Asunto(s)
Expresión Génica , Trasplante de Corazón , Infecciones por Herpesviridae/virología , Muromegalovirus/genética , ARN Viral/biosíntesis , Animales , Aorta/virología , Células Cultivadas , ADN Viral/análisis , Células Endoteliales/virología , Fibroblastos/virología , Perfilación de la Expresión Génica , Muromegalovirus/crecimiento & desarrollo , Músculo Liso Vascular/citología , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Bazo/virología , Glándula Submandibular/virología , Trasplante Homólogo
17.
Thorac Cardiovasc Surg ; 54(4): 219-26, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16755441

RESUMEN

OBJECTIVE: To further elucidate the mechanism by which cytomegalovirus (CMV) may promote atherosclerosis, we studied the expression pattern of cellular inflammatory and proliferative signals in the aortic wall of CMV(+) and CMV(-) patients undergoing coronary artery bypass grafting (CABG). METHODS: Aortic biopsies and blood samples of 68 CABG patients were investigated for CMV-DNA by PCR and IN SITU hybridisation. Expression of pp65 antigen, adhesion molecules (ICAM-1, VCAM-1, E-selectin), growth factors (PDGF-AA, TGF-beta), and the cellular proliferation factor Ki-67 was studied by immunohistochemistry. Logistic regression was used to test the correlation between the presence of CMV, vascular inflammation, and traditional noninflammatory risk factors for atherosclerosis. RESULTS: CMV-DNA was detected in the aortic tissue of 52 (76%) patients, and was localised predominantly in vascular smooth muscle cells. In CMV(+) patients, the expression of adhesion molecules and growth factors in the aortic endothelium was increased compared with CMV(-) patients. A positive correlation of elevated CRP, the induction of adhesion molecules and growth factors and CMV(+) was found. Female gender, smoking, and hyperlipidaemia were identified as risk factors for CMV(+). CONCLUSIONS: CMV-DNA in smooth muscle cells induces local growth factor expression as well as endothelial activation, both of which can promote the progression of atherosclerosis. Since traditional atherogenic risk factors increase the likelihood of aortic CMV manifestation, we suggest that CMV plays a crucial role in mediating the progression of atherosclerosis.


Asunto(s)
Aterosclerosis/virología , Proliferación Celular , Puente de Arteria Coronaria , Citomegalovirus/fisiología , Músculo Liso Vascular/virología , Anciano , Aorta/metabolismo , Aorta/patología , Aorta/virología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Citomegalovirus/genética , ADN Viral/sangre , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/virología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factores de Riesgo , Factores Sexuales , Fumar/efectos adversos , Factor de Crecimiento Transformador beta/biosíntesis , Molécula 1 de Adhesión Celular Vascular/biosíntesis
18.
Am J Pathol ; 168(2): 370-85, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16436653

RESUMEN

Hepatitis C virus (HCV) infection is frequently complicated by glomerulonephritis with immune complexes containing viral RNA. We examined the potential influence of Toll-like receptors (TLRs), specifically TLR3 recognition of viral dsRNA exemplified by polyriboinosinic:polyribocytidylic acid [poly(I:C) RNA]. Normal human kidney stained positive for TLR3 on mesangial cells (MCs), vascular smooth muscle cells, and collecting duct epithelium. Cultured MCs have low TLR3 mRNA levels with predominant intracellular protein localization, which was increased by tumor necrosis factor-alpha, interleukin (IL)-1beta, interferon (IFN)-gamma, and the TLR3 ligand poly(I:C) RNA. Poly(I:C) RNA stimulation of MCs increased mRNA and protein synthesis of IL-6, IL-1beta, M-CSF, IL-8/CXCL8, RANTES/CCL5, MCP-1/CCL2, and ICAM-I; it also increased anti-proliferative and proapoptotic effects, the latter of which was decreased by inhibiting caspase-8. In microdissected glomeruli of normal and non-HCV membranoproliferative glomerulonephritis biopsies, TLR3 mRNA expression was low. In contrast TLR3 mRNA expression was significantly increased in hepatitis C-positive glomerulonephritis and was associated with enhanced mRNA for RANTES/CCL5 and MCP-1/CCL2. We hypothesize that immune complexes containing viral RNA activate mesangial TLR3 during HCV infection, thereby contributing to chemokine/cytokine release and effecting proliferation and apoptosis. Thus, TLR3 expression on renal cells, and especially MCs, may establish a link between viral infections and glomerular diseases.


Asunto(s)
Glomerulonefritis/metabolismo , Glomerulonefritis/virología , Hepacivirus/genética , Hepatitis C/complicaciones , Receptor Toll-Like 3/metabolismo , Apoptosis , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Epitelio/metabolismo , Epitelio/virología , Glomerulonefritis/inmunología , Hepacivirus/patogenicidad , Humanos , Interferón gamma/farmacología , Interleucina-1/farmacología , Riñón/citología , Riñón/metabolismo , Riñón/virología , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/virología , Ligandos , Células Mesangiales/metabolismo , Células Mesangiales/virología , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Poli I-C/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptor Toll-Like 3/genética , Factor de Necrosis Tumoral alfa/farmacología
19.
Mol Ther ; 13(4): 694-704, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16431160

RESUMEN

Restenosis is a pathological condition involving intimal hyperplasia and negative arterial remodeling. Gene therapy vectors have shown modest therapeutic effects, but the level of infectivity has been relatively poor. In the present study we have designed a modified lentiviral vector (LV) pseudotyped with a strain of Hantavirus (HTNV) to improve the transduction efficiency into vascular smooth muscle and endothelial cells in vitro and in vivo. In vivo studies using adult New Zealand White rabbits demonstrated that local delivery of HTNV-pseudotyped LV (2 x 10(7) TU) into balloon-injured carotid arteries led to highly efficient transduction into endothelial and smooth muscle cells more effectively than VSV-G-pseudotyped LV (2 x 10(7) TU) or replication-defective adenoviral vectors (1-1.5 x 10(9) pfu) as determined by beta-gal immunohistochemistry. Overexpression of extracellular superoxide dismutase in balloon-injured carotid arteries 6 weeks after LV administration resulted in a significant reduction (P = 0.0024) of the intima/media ratio (0.18 +/- 0.09; n = 4) compared to vehicle-infused carotid arteries (0.69 +/- 0.08; n = 7). No beta-gal immunostaining was detected in other systemic organs, including the spleen, liver, heart, lung, kidneys, and brain. Moreover, no changes in plasma alanine aminotransferase or aspartate aminotransferase were detected following LV administration. In all, these data show that LV pseudotyped with Hantaviral glycoproteins can be a useful vector for targeting therapeutic genes to the vasculature in vivo.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Lentivirus/genética , Orthohantavirus/genética , Angioplastia de Balón , Animales , Arterias Carótidas/patología , Arterias Carótidas/virología , Bovinos , Línea Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Células Endoteliales/virología , Humanos , Inmunohistoquímica , Operón Lac , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/virología , Conejos , Superóxido Dismutasa/análisis , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
20.
Cardiovasc Res ; 67(1): 151-60, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15949479

RESUMEN

BACKGROUND: Cytomegaloviruses have been shown to promote atherogenesis in animal models. In humans, several epidemiological and clinical studies suggest involvement of the human cytomegalovirus (HCMV) in the development of atherosclerosis. HCMV is suspected to be associated with an enhanced restenosis rate and the occurrence of vasculopathies after solid organ transplantation. However, knowledge about the cellular and molecular bases of these findings is very limited. METHODS AND RESULTS: Human coronary artery smooth muscle cells (HCASMC) were successfully infected with HCMV in vitro. Infection of HCASMC with all HCMV strains analyzed resulted in a substantial upregulation of the beta-receptor of platelet-derived growth factor (PDGFR-beta) expression as demonstrated by immunohistochemistry, immunofluorescence, FACS, and Western blot analysis. The amount of PDGFR-beta protein present in HCASMC rapidly increased after 12 h of infection and this difference persisted for 72 h post-infection. We showed by quantitative FACS analysis that the extent of PDGFR-beta upregulation differed significantly between the HCMV strains TB40E, Toledo, and AD169. The expression of insulin-like growth factor receptors as well as hepatocyte growth factor receptors, however, was down-modulated in HCMV-infected HCASMC. Most importantly, the HCMV-associated upregulation of PDGFR-beta protein resulted in functionally intact receptors. A significantly higher increase of proliferative activity following stimulation with PDGF-BB was observed in HCMV-infected HCASMC compared to the uninfected control. CONCLUSIONS: Our data suggest that HCMV directly activates the PDGF system, which could promote atherogenesis and restenosis by activation of smooth muscle cell proliferation and neointima formation.


Asunto(s)
Aterosclerosis/virología , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus , Regulación Viral de la Expresión Génica , Músculo Liso Vascular/virología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Aterosclerosis/metabolismo , Western Blotting/métodos , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica/métodos , Especificidad de la Especie , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...