Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Signal ; 76: 109783, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32956789

RESUMO

Pathological neointimal growth can develop in patients as a result of vascular injury following percutaneous coronary intervention and coronary artery bypass grafting using autologous saphenous vein, leading to arterial or vein graft occlusion. Neointima formation driven by intimal hyperplasia occurs as a result of a complex interplay between molecular and cellular processes involving different cell types including endothelial cells, vascular smooth muscle cells and various inflammatory cells. Therefore, understanding the intercellular communication mechanisms underlying this process remains of fundamental importance in order to develop therapeutic strategies to preserve endothelial integrity and vascular health post coronary interventions. Extracellular vesicles (EVs), including microvesicles and exosomes, are membrane-bound particles secreted by cells which mediate intercellular signalling in physiological and pathophysiological states, however their role in neointima formation is not fully understood. The purification and characterization techniques currently used in the field are associated with many limitations which significantly hinder the ability to comprehensively study the role of specific EV types and make direct functional comparisons between EV subpopulations. In this review, the current knowledge focusing on EV signalling in neointima formation post vascular injury is discussed.


Assuntos
Células Endoteliais , Vesículas Extracelulares/metabolismo , Músculo Liso Vascular , Miócitos de Músculo Liso , Neointima/metabolismo , Lesões do Sistema Vascular , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia
2.
Cell Signal ; 75: 109751, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32860954

RESUMO

Atherosclerosis is a major cardiovascular disease and in 2016, the World Health Organisation (WHO) estimated 17.5 million global deaths, corresponding to 31% of all global deaths, were driven by inflammation and deposition of lipids into the arterial wall. This leads to the development of plaques which narrow the vessel lumen, particularly in the coronary and carotid arteries. Atherosclerotic plaques can become unstable and rupture, leading to myocardial infarction or stroke. Extracellular vesicles (EVs) are a heterogeneous population of vesicles secreted from cells with a wide range of biological functions. EVs participate in cell-cell communication and signalling via transport of cargo including enzymes, DNA, RNA and microRNA in both physiological and patholophysiological settings. EVs are present in atherosclerotic plaques and have been implicated in cellular signalling processes in atherosclerosis development, including immune responses, inflammation, cell proliferation and migration, cell death and vascular remodeling during progression of the disease. In this review, we summarise the current knowledge regarding EV signalling in atherosclerosis progression and the potential of utilising EV signatures as biomarkers of disease.


Assuntos
Aterosclerose/metabolismo , Comunicação Celular , Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Humanos , Transdução de Sinais , Remodelação Vascular
3.
Sci Rep ; 9(1): 3154, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30816157

RESUMO

The counter-regulatory axis, Angiotensin Converting Enzyme 2, Angiotensin-(1-7), Mas receptor (ACE2/Ang-1-7/MasR), of the renin angiotensin system (RAS) is a potential therapeutic target in stroke, with Ang-(1-7) reported to have neuroprotective effects in pre-clinical stroke models. Here, an extensive investigation of the functional and mechanistic effects of Ang-(1-7) was performed in a rodent model of stroke. Using longitudinal magnetic resonance imaging (MRI) it was observed that central administration of Ang-(1-7) following transient middle cerebral artery occlusion (MCAO) increased the amount of tissue salvage compared to reperfusion alone. This protective effect was not due to early changes in blood brain barrier (BBB) permeability, microglia activation or inflammatory gene expression. However, increases in NADPH oxidase 1 (Nox1) mRNA expression were observed in the treatment group compared to control. In order to determine whether Ang-(1-7) has direct cerebrovascular effects, laser speckle contrast imaging (LSCI) was performed to measure dynamic changes in cortical perfusion following reperfusion. Delivery of Ang-(1-7) did not have any effect on cortical perfusion following reperfusion however; it showed an indication to prevent the 'steal phenomenon' within the contralateral hemisphere. The comprehensive series of studies have demonstrated a moderate protective effect of Ang-(1-7) when given alongside reperfusion to increase tissue salvage.


Assuntos
Angiotensina I/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , NADPH Oxidase 1/genética , Fragmentos de Peptídeos/farmacologia , Peptidil Dipeptidase A/genética , Acidente Vascular Cerebral/tratamento farmacológico , Enzima de Conversão de Angiotensina 2 , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Meios de Contraste/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Infarto da Artéria Cerebral Média/diagnóstico por imagem , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/diagnóstico por imagem , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/patologia , Imageamento por Ressonância Magnética , Microglia/efeitos dos fármacos , Microglia/patologia , Artéria Cerebral Média/diagnóstico por imagem , Artéria Cerebral Média/efeitos dos fármacos , Fármacos Neuroprotetores , Proto-Oncogene Mas , RNA Mensageiro/genética , Ratos , Sistema Renina-Angiotensina/genética , Reperfusão/métodos , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
4.
Gene Ther ; 23(7): 592-6, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27014840

RESUMO

Intravenous delivery of adenoviruses is the optimal route for many gene therapy applications. Once in the blood, coagulation factor X (FX) binds to the adenovirus capsid and protects the virion from natural antibody and classical complement-mediated neutralisation in mice. However, to date, no studies have examined the relevance of this FX/viral immune protective mechanism in human samples. In this study, we assessed the effects of blocking FX on adenovirus type 5 (Ad5) activity in the presence of human serum. FX prevented human IgM binding directly to the virus. In individual human sera samples (n=25), approximately half of those screened inhibited adenovirus transduction only when the Ad5-FX interaction was blocked, demonstrating that FX protected the virus from neutralising components in a large proportion of human sera. In contrast, the remainder of sera tested had no inhibitory effects on Ad5 transduction and FX armament was not required for effective gene transfer. In human sera in which FX had a protective role, Ad5 induced lower levels of complement activation in the presence of FX. We therefore demonstrate for the first time the importance of Ad-FX protection in human samples and highlight subject variability and species-specific differences as key considerations for adenoviral gene therapy.


Assuntos
Adenoviridae/imunologia , Fator X/imunologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Adenoviridae/genética , Linhagem Celular Tumoral , Vetores Genéticos/sangue , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Células HEK293 , Humanos , Imunoglobulina M/imunologia , Injeções Intravenosas/métodos
5.
J Physiol ; 589(Pt 4): 939-51, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21173078

RESUMO

The renin­angiotensin system (RAS) regulates blood pressure mainly via the actions of angiotensin (Ang)II, generated via angiotensin converting enzyme (ACE). The ACE homologue ACE2 metabolises AngII to Ang1-7, decreasing AngII and increasing Ang1-7, which counteracts AngII activity via the Mas receptor. However, ACE2 also converts AngI to Ang1-9, a poorly characterised peptide which can be further converted to Ang1-7 via ACE. Ang1-9 stimulates bradykinin release in endothelium and has antihypertrophic actions in the heart, attributed to its being a competitive inhibitor of ACE, leading to decreased AngII, rather than increased Ang1-7. To date no direct receptor-mediated effects of Ang1-9 have been described. To further understand the role of Ang1-9 in RAS function we assessed its action in cardiomyocyte hypertrophy in rat neonatal H9c2 and primary adult rabbit left ventricular cardiomyocytes, compared to Ang1-7. Cardiomyocyte hypertrophy was stimulated with AngII or vasopressin, significantly increasing cell size by approximately 1.2-fold (P < 0.05) as well as stimulating expression of the hypertrophy gene markers atrial natriuretic peptide, brain natriuretic peptide, ß-myosin heavy chain and myosin light chain (2- to 5-fold, P < 0.05). Both Ang1-9 and Ang1-7 were able to block hypertrophy induced by either agonist (control, 186.4 µm; AngII, 232.8 µm; AngII+Ang1-7, 198.3 µm; AngII+Ang1-9, 195.9 µm; P < 0.05). The effects of Ang1-9 were not inhibited by captopril, supporting previous evidence that Ang1-9 acts independently of Ang1-7. Next, we investigated receptor signalling via angiotensin type 1 and type 2 receptors (AT1R, AT2R) and Mas. The AT1R antagonist losartan blocked AngII-induced, but not vasopressin-induced, hypertrophy. Losartan did not block the antihypertrophic effects of Ang1-9, or Ang1-7 on vasopressin-stimulated cardiomyocytes. The Mas antagonist A779 efficiently blocked the antihypertrophic effects of Ang1-7, without affecting Ang1-9. Furthermore, Ang1-7 activity was also inhibited in the presence of the bradykinin type 2 receptor antagonist HOE140, without affecting Ang1-9. Moreover, we observed that the AT2R antagonist PD123,319 abolished the antihypertrophic effects of Ang1-9, without affecting Ang1-7, suggesting Ang1-9 signals via the AT2R. Radioligand binding assays demonstrated that Ang1-9 was able to bind the AT2R (pKi = 6.28 ± 0.1). In summary, we ascribe a direct biological role for Ang1-9 acting via the AT2R. This has implications for RAS function and identifying new therapeutic targets in cardiovascular disease.


Assuntos
Angiotensina II/fisiologia , Angiotensina I/fisiologia , Miócitos Cardíacos/patologia , Fragmentos de Peptídeos/fisiologia , Receptor Tipo 2 de Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Linhagem Celular Transformada , Sobrevivência Celular/fisiologia , Células Cultivadas , Células HeLa , Humanos , Hipertrofia/patologia , Miócitos Cardíacos/fisiologia , Fragmentos de Peptídeos/farmacologia , Coelhos , Ratos
6.
Artigo em Inglês | MEDLINE | ID: mdl-22654822

RESUMO

G protein-coupled receptors (GPCRs) remain the best studied class of cell surface receptors and the most tractable family of proteins for novel small molecule drug discovery. Despite this, a considerable number of GPCRs remain poorly characterized and in a significant number of cases, endogenous ligand(s) that activate them remain undefined or are of questionable physiological relevance. GPR35 was initially discovered over a decade ago but has remained an "orphan" receptor. Recent publications have highlighted novel ligands, both endogenously produced and synthetic, which demonstrate significant potency at this receptor. Furthermore, evidence is accumulating which highlights potential roles for GPR35 in disease and therefore, efforts to characterize GPR35 more fully and develop it as a novel therapeutic target in conditions that range from diabetes and hypertension to asthma are increasing. Recently identified ligands have shown marked species selective properties, indicating major challenges for future drug development. As we begin to understand these issues, the continuing efforts to identify novel agonist and antagonist ligands for GPR35 will help to decipher its true physiological relevance; translating multiple assay systems in vitro, to animal disease systems in vivo and finally to man.

7.
J Mol Cell Cardiol ; 46(6): 891-901, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19249308

RESUMO

In this study a Ca(2+) sensitive protein was targeted to the mitochondria of adult rabbit ventricular cardiomyocytes using an adenovirus transfection technique. The probe (Mitycam) was a Ca(2+)-sensitive inverse pericam fused to subunit VIII of human cytochrome c oxidase. Mitycam expression pattern and Ca(2+) sensitivity was characterized in HeLa cells and isolated adult rabbit cardiomyocytes. Cardiomyocytes expressing Mitycam were voltage-clamped and depolarized at regular intervals to elicit a Ca(2+) transient. Cytoplasmic (Fura-2) and mitochondrial Ca(2+) (Mitycam) fluorescence were measured simultaneously under a range of cellular Ca(2+) loads. After 48 h post-adenoviral transfection, Mitycam expression showed a characteristic localization pattern in HeLa cells and cardiomyocytes. The Ca(2+) sensitive component of Mitycam fluorescence was 12% of total fluorescence in HeLa cells with a K(d) of approximately 220 nM. In cardiomyocytes, basal and beat-to-beat changes in Mitycam fluorescence were detected on initiation of a train of depolarizations. Time to peak of the mitochondrial Ca(2+) transient was slower, but the rate of decay was faster than the cytoplasmic signal. During spontaneous Ca(2+) release the relative amplitude and the time course of the mitochondrial and cytoplasmic signals were comparable. Inhibition of mitochondrial respiration decreased the mitochondrial transient amplitude by approximately 65% and increased the time to 50% decay, whilst cytosolic Ca(2+) transients were unchanged. The mitochondrial Ca(2+) uniporter (mCU) inhibitor Ru360 prevented both the basal and transient components of the rise in mitochondrial Ca(2+). The mitochondrial-targeted Ca(2+) probe indicates sustained and transient phases of mitochondrial Ca(2+) signal, which are dependent on cytoplasmic Ca(2+) levels and require a functional mCU.


Assuntos
Cálcio/metabolismo , Ventrículos do Coração/citologia , Mitocôndrias Cardíacas/metabolismo , Sondas Moleculares/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Western Blotting , Eletrofisiologia , Células HeLa , Humanos , Sondas Moleculares/genética , Técnicas de Patch-Clamp , Coelhos , Proteínas Recombinantes de Fusão/genética , Transfecção
8.
Gene Ther ; 15(6): 443-51, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18004401

RESUMO

Targeted delivery of biological agents to atherosclerotic plaques may provide a novel treatment and/or useful tool for imaging of atherosclerosis in vivo. However, there are no known viral vectors that possess the desired tropism. Two plaque-targeting peptides, CAPGPSKSC (CAP) and CNHRYMQMC (CNH) were inserted into the capsid of adeno-associated virus 2 (AAV2) to assess vector retargeting. AAV2-CNH produced significantly higher levels of transduction than unmodified AAV2 in human, murine and rat endothelial cells, whereas transduction of nontarget HeLa cells was unaltered. Transduction studies and surface plasmon resonance suggest that AAV2-CNH uses membrane type 1 matrix metalloproteinase as a surface receptor. AAV2-CAP only produced higher levels of transduction in rat endothelial cells, possibly because the virus was found to be affected by proteasomal degradation. In vivo substantially higher levels of both peptide-modified AAV2 vectors was detected in the brachiocephalic artery (site of advanced atherosclerotic plaques) and aorta, whereas reduced levels were detected in all other organs examined. These results suggest that in the AAV2 platform the peptides are exposed on the capsid surface in a way that enables efficient receptor binding and so creates effective atherosclerotic plaque targeted vectors.


Assuntos
Aterosclerose/terapia , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Animais , Apolipoproteínas E/genética , Linhagem Celular , Dependovirus/metabolismo , Marcação de Genes , Engenharia Genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Heparina/metabolismo , Humanos , Metaloproteinase 1 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/genética , Ligação Proteica , Ratos , Ressonância de Plasmônio de Superfície , Transdução Genética/métodos , Transgenes
9.
Gene Ther ; 12(20): 1534-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15944729

RESUMO

Transduction of the vascular endothelium by adeno-associated virus (AAV) vectors would have broad appeal for gene therapy. However, levels of transduction by AAV serotype-2 are low, an observation linked to deficiencies in endothelial cell binding, sequestration of virions in the extracellular matrix and/or virion degradation by the proteasome. Strategies to improve transduction of endothelial cells include AAV-2 capsid targeting using small peptides isolated by phage display or the use of alternate serotypes. Previously, we have shown that AAV serotypes-3 through -6 transduce endothelial cells with poor efficiency. Recently, AAV serotypes-7 and -8 have been shown to mediate efficient transduction of the skeletal muscle and liver, respectively, although their infectivity profile for vascular cells has not been addressed. Here, we show that AAV-7 and -8 also transduce endothelial cells with poor efficiency and the levels of transgene expression are markedly enhanced by inhibition of the proteasome. In both cases proteasome blockade enhances the nuclear translocation of virions. We further show that this is vascular cell-type selective since transduction of smooth muscle cells is not sensitive to proteasome inhibition. Analysis in intact blood vessels corroborated these findings and suggests that proteasome degradation is a common limiting factor for endothelial cell transduction by AAV vectors.


Assuntos
Dependovirus/genética , Células Endoteliais/virologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Músculo Liso Vascular/virologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Aorta Torácica/virologia , Linhagem Celular , Células Cultivadas , DNA Viral/análise , Células Endoteliais/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Leupeptinas/uso terapêutico , Camundongos , Músculo Liso Vascular/metabolismo , Inibidores de Proteassoma , Ratos , Ratos Endogâmicos WKY , Veia Safena , Sorotipagem , Transdução Genética/métodos
10.
Gene Ther ; 11(16): 1296-300, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15292916

RESUMO

Adenovirus (Ad)-mediated gene delivery is a promising approach for genetic manipulation of the vasculature and is being used in both preclinical models and clinical trials. However, safety concerns relating to infection of nontarget tissue and the poor infectivity of vascular cells compared to other cell types necessitates Ad vector refinement. Here, we combine a transductional targeting approach to improve vascular cell infectivity through RGD peptide insertion into adenovirus fibers, combined with transcriptional targeting to endothelial cells using a approximately 1 kb fragment of the fms-like tyrosine kinase receptor-1 (FLT-1) promoter. Single- and double-modified vectors were characterized in human cell lines that either support or have silenced FLT-1 expression. In rat hepatocytes and endothelial cells, the double modification substantially shifted transduction profiles toward vascular endothelial cells. Furthermore, in intact aortae derived from spontaneously hypertensive rats that display enhanced alphav integrin expression on dysfunctional endothelium, enhanced levels of transduction were observed using the double-modified vector but not in aortae derived from normotensive control rats. Our data indicate that Ad-mediated transduction can be beneficially modified in vitro and in vivo by combining fiber modification and a cell-selective promoter within a single-component vector system.


Assuntos
Infecções por Adenoviridae/genética , Adenoviridae/genética , Marcação de Genes/métodos , Animais , Aorta/fisiopatologia , Linhagem Celular , Células Cultivadas , Células Endoteliais/fisiologia , Terapia Genética/métodos , Vetores Genéticos/genética , Hepatócitos/fisiologia , Humanos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Proteína Tirosina Quinases , Receptores Virais/genética , Transcrição Gênica/genética , Transdução Genética/métodos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
11.
Curr Gene Ther ; 4(1): 89-113, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15032617

RESUMO

It has become apparent that the clinical success anticipated in the field of gene therapy has been limited by progress in several of the fundamental areas of genetics, molecular and cellular biology relevant to its application. Whilst a great deal of effort has been made in the evaluation of transgenes, it is only more recently with the advance of vector systems that attention has begun to be focused upon the means and control of transgene expression. Until recently, the majority of constructs have employed ubiquitous viral promoters to drive expression from simple gene expression cassettes using viral promoters and lacking introns, 3' untranslated regions (UTRs), locus control regions (LCR's), matrix attachment regions (MAR's) and other such genetic components. It has consequently emerged that these elements may have a key role in determining the levels and longevity of gene expression attainable in vivo, irrespective of the vector system utilised. The majority of gene therapy applications would also benefit from the specific optimisation of 'tailor-made' expression cassettes to optimise their therapeutic efficacy. In conjunction with modification of vector tropism and strategies to limit their immunogenicity, this should create vectors suitable for the clinical application of gene therapy. This review aims to highlight some of the principle considerations of gene expression in vivo, and the means by which it may most effectively be achieved, whether this is via the minimal modification of an existing eukaryotic promoter or by the more extensive design of a novel promoter and associated elements.


Assuntos
Regulação da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/química , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Animais , Células Eucarióticas/metabolismo , Vetores Genéticos/genética , Humanos , Neoplasias/terapia , Fatores de Transcrição/metabolismo , Vírus/genética
12.
Gene Ther ; 9(2): 110-7, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11857069

RESUMO

Gene transfer may be appropriate for therapeutic protocols targeted at the vascular endothelium. Endothelial dysfunction is the principal phenotype associated with atherosclerosis and hypertension. Oxidative stress has been implicated in the development of endothelial dysfunction. We have explored the ability of overexpressing anti-oxidant genes (superoxide dismutases; SODs) in vitro and in vivo to assess their potential for reversing endothelial dysfunction in a rat model, the stroke-prone spontaneously hypertensive rat (SHRSP). Western blotting and immunofluorescence assays in vitro showed efficient overexpression of MnSOD and ECSOD with respect to localisation to the mitochondria and extracellular surface, respectively. Transgene functional activity was quantified with SOD activity assays. MnSOD and ECSOD overexpression in intact SHRSP vessels in vivo led to endothelial and adventitial overexpression. Pharmacological assessment of transduced vessels following in vivo delivery by basal NO availability quantification demonstrated that the "null" adenovirus and MnSOD adenovirus did not significantly increase NO availability. However, AdECSOD-treated carotid arteries showed a significant increase in NO availability (1.91 +/- 0.04 versus 0.75 +/- 0.08 g/g, n = 6, P = 0.029). In summary, efficient overexpression of ECSOD, but not MnSOD in vivo, results in improved endothelial function in a rat model of hypertension and has important implications for the development of endothelial-based vascular gene therapy.


Assuntos
Endotélio Vascular/fisiopatologia , Sequestradores de Radicais Livres/metabolismo , Terapia Genética/métodos , Hipertensão/terapia , Superóxido Dismutase/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Células Cultivadas , Técnicas de Transferência de Genes , Vetores Genéticos/uso terapêutico , Hipertensão/enzimologia , Hipertensão/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos SHR , Superóxido Dismutase/genética
13.
Mol Ther ; 4(6): 534-42, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11735337

RESUMO

Adenovirus type 5 (Ad) based vectors transduce vascular endothelial cells (EC) and have been widely used for vascular gene transfer. However, many cell types express the Ad receptor (cox-sackievirus adenovirus receptor; CAR), preventing selective EC infection and precluding clinical use. We previously isolated the human EC-binding peptides SIGYPLP and LSNFHSS by phage display and demonstrated by means of a bispecific antibody that SIGYPLP directs efficient, high-level, EC-selective Ad-mediated gene transfer. We now generate genetically modified Ad fiber proteins with selective EC tropism by engineering these peptides into the HI loop of the Ad fiber. SIGYPLP, but not LSNFHSS, enhanced EC selectivity, demonstrating maintenance of peptide-cell binding fidelity upon incorporation into virions. Combining fiber mutations that block CAR binding (detargeting) with SIGYPLP insertion (retargeting) generated a novel Ad vector, AdKO1SIG, in a single component system. AdKO1SIG demonstrated efficient and selective tropism for EC compared with control Ad vectors. This is the first demonstration of genetic incorporation of a novel, mammalian, cell-selective ligand that retains its targeting fidelity in the Ad fiber HI loop, in combination with point mutations that abolish fiber-CAR interaction. This study demonstrates the potential for improving the cell-selectivity and safety of adenoviral vectors.


Assuntos
Adenoviridae/fisiologia , Endotélio Vascular/virologia , Terapia Genética/métodos , Fragmentos de Peptídeos/genética , Receptores Virais/metabolismo , Sítios de Ligação , Ligação Competitiva , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Primers do DNA/química , Citometria de Fluxo , Marcação de Genes/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Immunoblotting , Proteínas Luminescentes/metabolismo , Plasmídeos/genética , Receptores Virais/genética , Linfócitos T/metabolismo , Transdução Genética , Transfecção
14.
Mol Ther ; 4(3): 174-81, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11545607

RESUMO

Gene therapy vectors based on adeno-associated virus-2 (AAV2) offer considerable promise for human gene therapy. Applications for AAV vectors are limited to tissues efficiently transduced by the vector due to its natural tropism, which is predominantly skeletal muscle, neurons, and hepatocytes. Tropism modification to elevate efficiency and/or selectivity to individual cell types would enhance the scope of AAV for disease therapies. The vascular endothelium is implicitly important in cardiovascular diseases and cancer, but is relatively poorly transduced by AAV vectors. We therefore genetically incorporated the peptide SIGYPLP, which targets endothelial cells (EC), into position I-587 of AAV capsids. SIGYPLP-modified AAV (AAVsig) showed enhanced transduction of human EC compared with AAV with a wild-type capsid (AAVwt), a phenotype independent of heparan sulphate proteoglycan (HSPG) binding. In contrast, AAVsig did not enhance transduction of primary human vascular smooth muscle cells or human hepatocytes, principal targets for AAV vectors in local or systemic gene delivery applications, respectively. Furthermore, infection of EC in the presence of bafilomycin A(2) indicated that intracellular trafficking of AAV particles was altered by targeting AAV by means of SIGYPLP. AAV vectors with enhanced tropism for EC will be useful for diverse gene therapeutics targeted at the vasculature.


Assuntos
Dependovirus/genética , Endotélio Vascular/metabolismo , Macrolídeos , Transdução Genética/métodos , Sequência de Aminoácidos , Antibacterianos/farmacologia , Transporte Biológico/efeitos dos fármacos , Capsídeo/química , Capsídeo/genética , Capsídeo/metabolismo , Células Cultivadas , Dependovirus/efeitos dos fármacos , Dependovirus/metabolismo , Dependovirus/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/virologia , Expressão Gênica/efeitos dos fármacos , Engenharia Genética , Terapia Genética/métodos , Células HeLa , Proteoglicanas de Heparan Sulfato/antagonistas & inibidores , Proteoglicanas de Heparan Sulfato/metabolismo , Heparina/farmacologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virologia , Mutação , Especificidade de Órgãos , Receptores Virais/antagonistas & inibidores , Receptores Virais/metabolismo
15.
Nat Biotechnol ; 19(9): 838-42, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11533642

RESUMO

The promise of gene therapy for health care will not be realized until gene delivery systems are capable of achieving efficient, cell-specific gene delivery in vivo. Here we describe an adenoviral system for achieving cell-specific transgene expression in pulmonary endothelium. The combination of transductional targeting to a pulmonary endothelial marker (angiotensin-converting enzyme, ACE) and an endothelial-specific promoter (for vascular endothelial growth factor receptor type 1, flt-1) resulted in a synergistic, 300,000-fold improvement in the selectivity of transgene expression for lung versus the usual site of vector sequestration, the liver. This combined approach should be useful for the design of other gene delivery systems.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Transcrição Gênica , Transdução Genética , Transgenes , Adenoviridae/genética , Animais , Linhagem Celular , Fatores de Crescimento Endotelial/genética , Endotélio/metabolismo , Endotélio Vascular/metabolismo , Vetores Genéticos , Imuno-Histoquímica , Fígado/metabolismo , Luciferases/metabolismo , Pulmão/metabolismo , Linfocinas/genética , Masculino , Camundongos , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Hypertension ; 38(1): 65-70, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11463761

RESUMO

The use of viral vectors for vascular gene therapy targeted at the endothelium is limited by the promiscuous tropism of vectors and nonspecificity of viral promoters, resulting in high-level transgene expression in multiple tissues. To evaluate suitable endothelial cell (EC)-specific promoters for vascular gene therapy, we directly compared the ability of the fms-like tyrosine kinase-1 (FLT-1), intercellular adhesion molecule-2 (ICAM-2), and von Willebrand factor (vWF) promoters to drive EC-restricted transcription after cloning into adenoviral vectors upstream of lacZ. Vastly different expression profiles were observed. Whereas both FLT-1 and ICAM-2 promoters generated transgene expression levels similar to cytomegalovirus in ECs in vitro, vWF expression levels were extremely low. Analysis of non-EC types revealed that ICAM-2 but not FLT-1 evoked leaky transgene expression, thus identifying FLT-1 as the most selective promoter. With an ex vivo human gene therapy model, the FLT-1 promoter demonstrated EC-specific transgene expression in intact human vein but no detectable expression from infected exposed smooth muscle cells in EC-denuded vein. Furthermore, when adenoviruses were systemically administered to mice, the FLT-1 promoter demonstrated extremely low-level gene expression in the liver, the major target organ for adenoviral transduction in vivo. This study highlights the potential of using the FLT-1 promoter for local and systemic human gene therapy in hypertension and its complications.


Assuntos
Endotélio Vascular/fisiologia , Terapia Genética , Hipertensão/terapia , Regiões Promotoras Genéticas/fisiologia , Adenoviridae/genética , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Perfilação da Expressão Gênica , Marcação de Genes , Vetores Genéticos , Humanos , Hipertensão/patologia , Veia Safena/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , beta-Galactosidase/metabolismo
17.
Hypertension ; 37(2 Pt 2): 449-55, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11230317

RESUMO

Current gene delivery vectors demonstrate inefficient and nonselective gene transfer to vascular endothelial cells, limiting their use in cardiovascular gene transfer and therapy. The lectinlike oxidized LDL receptor (LOX-1) is expressed selectively at low levels on endothelial cells but is strongly upregulated in dysfunctional endothelial cells associated with hypertension and atherogenesis. Using LOX-1 as a target receptor, we have sought to isolate peptide ligands that mediate binding to the extracellular domain of LOX-1 as a definitive step in the development of targeted gene transfer aimed at dysfunctional endothelium. To achieve this, we ectopically overexpressed LOX-1 in cells lacking endogenous LOX-1 by using an episomally maintained expression system and designed a novel subtractive phage display strategy to identify peptides selective for LOX-1. After extensive biopanning, we sequenced individual phage and identified 60 novel peptides. This population of peptides contained a number of potential consensus motifs. To define the selectivity of individual peptides for LOX-1 with the use of an independent gene transfer system, we developed a novel adenoviral vector to overexpress LOX-1 transiently in primary cells and cell lines. We then quantified recovery of each peptide from LOX-1-positive and LOX-1-negative cells after adenovirus-mediated gene transfer. This strategy confirmed selectivity to LOX-1 for many peptides and highlighted the peptides LSIPPKA, FQTPPQL, and LTPATAI as principal candidates. These peptides will be useful for the selective targeting of viral and nonviral gene transfer vectors to endothelial cells expressing the LOX-1 receptor in vitro and in vivo and in particular dysfunctional endothelial cells associated with hypertension and atherosclerosis.


Assuntos
Endotélio Vascular/metabolismo , Peptídeos/genética , Receptores de LDL/genética , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Arteriosclerose/fisiopatologia , Western Blotting , Linhagem Celular , Células Cultivadas , Endotélio Vascular/fisiopatologia , Imunofluorescência , Técnicas de Transferência de Genes , Vetores Genéticos , Hipertensão/fisiopatologia , Ligantes , Peptídeos/química , Receptores de LDL/biossíntese , Receptores de LDL/metabolismo , Receptores de LDL Oxidado , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Circulation ; 102(2): 231-7, 2000 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-10889136

RESUMO

BACKGROUND: Gene transfer to vascular cells is a highly inefficient and nonselective process, defined by the lack of specific cell-surface receptors for both nonviral and viral gene delivery vectors. METHODS AND RESULTS: We used filamentous phage display to isolate a panel of peptides that have the ability to bind selectively and efficiently to quiescent human umbilical vein endothelial cells (HUVECs) with reduced or negligible binding to nonendothelial cells, including vascular smooth muscle cells and hepatocytes. By direct biopanning on HUVECs and a second approach involving preclearing steps before panning on HUVECs, we isolated and sequenced 140 individual phages and identified 59 peptides. We selected 7 candidates for further investigation by secondary screening of homogeneous phages on a panel of cell types. Using adenovirus-mediated gene transfer as a model gene delivery system, we cloned the peptide SIGYPLP and the positive control peptide KKKKKKK upstream of the S11e single-chain Fv ("adenobody") directed against the knob domain of the adenovirus to create fusion proteins. Adenovirus-mediated gene transfer via fiber-dependent infection was blocked with S11e, whereas inclusion of the KKKKKKK peptide retargeted gene transfer. The peptide SIGYPLP, however, retargeted gene delivery specifically to endothelial cells with a significantly enhanced efficiency over nontargeted adenovirus and without transduction of nontarget cells. CONCLUSIONS: Our study demonstrates the feasibility of using small, novel peptides isolated via phage display to target gene delivery specifically and efficiently to HUVECs and highlights their use for retargeting both viral and nonviral gene transfer to vascular endothelial cells for future clinical applications.


Assuntos
Adenoviridae/genética , Endotélio Vascular/fisiologia , Técnicas de Transferência de Genes , Biblioteca de Peptídeos , Infecções por Adenoviridae/genética , Células Cultivadas , Clonagem Molecular , Endotélio Vascular/citologia , Células HeLa , Humanos , Fígado/citologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fragmentos de Peptídeos/genética , Polilisina/genética , Veias Umbilicais/citologia
19.
Methods Mol Med ; 30: 271-83, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-21341032

RESUMO

Adenoviruses are icosahedral viruses 70-90 nm in diameter with a double-stranded, linear DNA genome of approx 36 kb. They are widely utilized in gene transfer protocols owing to their relative ease of genetic manipulation, ability to grow to high titres (10(9)-10(12) plaque forming units [pfu]/mL), and their ability to infect both dividing and nondividing cells efficiently. The latter reason makes them extremely suitable for investigations in vascular systems where the low proliferative indices of cells is a limiting factor for retroviral gene transfer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...