Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 118(1): 5, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36700983

RESUMEN

Long non-coding RNAs (lncRNAs) can act as regulatory RNAs which, by altering the expression of target genes, impact on the cellular phenotype and cardiovascular disease development. Endothelial lncRNAs and their vascular functions are largely undefined. Deep RNA-Seq and FANTOM5 CAGE analysis revealed the lncRNA LINC00607 to be highly enriched in human endothelial cells. LINC00607 was induced in response to hypoxia, arteriosclerosis regression in non-human primates, post-atherosclerotic cultured endothelial cells from patients and also in response to propranolol used to induce regression of human arteriovenous malformations. siRNA knockdown or CRISPR/Cas9 knockout of LINC00607 attenuated VEGF-A-induced angiogenic sprouting. LINC00607 knockout in endothelial cells also integrated less into newly formed vascular networks in an in vivo assay in SCID mice. Overexpression of LINC00607 in CRISPR knockout cells restored normal endothelial function. RNA- and ATAC-Seq after LINC00607 knockout revealed changes in the transcription of endothelial gene sets linked to the endothelial phenotype and in chromatin accessibility around ERG-binding sites. Mechanistically, LINC00607 interacted with the SWI/SNF chromatin remodeling protein BRG1. CRISPR/Cas9-mediated knockout of BRG1 in HUVEC followed by CUT&RUN revealed that BRG1 is required to secure a stable chromatin state, mainly on ERG-binding sites. In conclusion, LINC00607 is an endothelial-enriched lncRNA that maintains ERG target gene transcription by interacting with the chromatin remodeler BRG1 to ultimately mediate angiogenesis.


Asunto(s)
ARN Largo no Codificante , Animales , Humanos , Ratones , Cromatina , ADN Helicasas/genética , ADN Helicasas/metabolismo , Células Endoteliales/metabolismo , Ratones SCID , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/genética , Neovascularización Fisiológica
2.
Arterioscler Thromb Vasc Biol ; 39(2): 224-236, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30580571

RESUMEN

Objective- PDI (protein disulfide isomerase A1) was reported to support Nox1 (NADPH oxidase) activation mediated by growth factors in vascular smooth muscle cells. Our aim was to investigate the molecular mechanism by which PDI activates Nox1 and the functional implications of PDI in Nox1 activation in vascular disease. Approach and Results- Using recombinant proteins, we identified a redox interaction between PDI and the cytosolic subunit p47phox in vitro. Mass spectrometry of crosslinked peptides confirmed redox-dependent disulfide bonds between cysteines of p47phox and PDI and an intramolecular bond between Cys 196 and 378 in p47phox. PDI catalytic Cys 400 and p47phox Cys 196 were essential for the activation of Nox1 by PDI in vascular smooth muscle cells. Transfection of PDI resulted in the rapid oxidation of a redox-sensitive protein linked to p47phox, whereas PDI mutant did not promote this effect. Mutation of p47phox Cys 196, or the redox active cysteines of PDI, prevented Nox1 complex assembly and vascular smooth muscle cell migration. Proximity ligation assay confirmed the interaction of PDI and p47phox in murine carotid arteries after wire injury. Moreover, in human atheroma plaques, a positive correlation between the expression of PDI and p47phox occurred only in PDI family members with the a' redox active site. Conclusions- PDI redox cysteines facilitate Nox1 complex assembly, thus identifying a new mechanism through which PDI regulates Nox activity in vascular disease.


Asunto(s)
Disulfuros/química , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasa 1/metabolismo , NADPH Oxidasas/química , Proteína Disulfuro Isomerasas/química , Animales , Movimiento Celular , Células Cultivadas , Activación Enzimática , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Oxidación-Reducción , Superóxidos/metabolismo
3.
Circulation ; 136(1): 65-79, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28351900

RESUMEN

BACKGROUND: The angiogenic function of endothelial cells is regulated by numerous mechanisms, but the impact of long noncoding RNAs (lncRNAs) has hardly been studied. We set out to identify novel and functionally important endothelial lncRNAs. METHODS: Epigenetically controlled lncRNAs in human umbilical vein endothelial cells were searched by exon-array analysis after knockdown of the histone demethylase JARID1B. Molecular mechanisms were investigated by RNA pulldown and immunoprecipitation, mass spectrometry, microarray, several knockdown approaches, CRISPR-Cas9, assay for transposase-accessible chromatin sequencing, and chromatin immunoprecipitation in human umbilical vein endothelial cells. Patient samples from lung and tumors were studied for MANTIS expression. RESULTS: A search for epigenetically controlled endothelial lncRNAs yielded lncRNA n342419, here termed MANTIS, as the most strongly regulated lncRNA. Controlled by the histone demethylase JARID1B, MANTIS was downregulated in patients with idiopathic pulmonary arterial hypertension and in rats treated with monocrotaline, whereas it was upregulated in carotid arteries of Macaca fascicularis subjected to atherosclerosis regression diet, and in endothelial cells isolated from human glioblastoma patients. CRISPR/Cas9-mediated deletion or silencing of MANTIS with small interfering RNAs or GapmeRs inhibited angiogenic sprouting and alignment of endothelial cells in response to shear stress. Mechanistically, the nuclear-localized MANTIS lncRNA interacted with BRG1, the catalytic subunit of the switch/sucrose nonfermentable chromatin-remodeling complex. This interaction was required for nucleosome remodeling by keeping the ATPase function of BRG1 active. Thereby, the transcription of key endothelial genes such as SOX18, SMAD6, and COUP-TFII was regulated by ensuring efficient RNA polymerase II machinery binding. CONCLUSION: MANTIS is a differentially regulated novel lncRNA facilitating endothelial angiogenic function.


Asunto(s)
Sistemas CRISPR-Cas/fisiología , Epigénesis Genética/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Microvasos/fisiología , Neovascularización Fisiológica/fisiología , ARN Largo no Codificante/biosíntesis , Animales , Línea Celular , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Histona Demetilasas con Dominio de Jumonji/genética , Macaca fascicularis , Masculino , Ratones , Ratones SCID , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética
6.
Mol Ther ; 24(4): 779-87, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26732878

RESUMEN

Inhibition of vascular smooth muscle cell (VSMC) proliferation by drug eluting stents has markedly reduced intimal hyperplasia and subsequent in-stent restenosis. However, the effects of antiproliferative drugs on endothelial cells (EC) contribute to delayed re-endothelialization and late stent thrombosis. Cell-targeted therapies to inhibit VSMC remodeling while maintaining EC health are necessary to allow vascular healing while preventing restenosis. We describe an RNA aptamer (Apt 14) that functions as a smart drug by preferentially targeting VSMCs as compared to ECs and other myocytes. Furthermore, Apt 14 inhibits phosphatidylinositol 3-kinase/protein kinase-B (PI3K/Akt) and VSMC migration in response to multiple agonists by a mechanism that involves inhibition of platelet-derived growth factor receptor (PDGFR)-ß phosphorylation. In a murine model of carotid injury, treatment of vessels with Apt 14 reduces neointimal formation to levels similar to those observed with paclitaxel. Importantly, we confirm that Apt 14 cross-reacts with rodent and human VSMCs, exhibits a half-life of ~300 hours in human serum, and does not elicit immune activation of human peripheral blood mononuclear cells. We describe a VSMC-targeted RNA aptamer that blocks cell migration and inhibits intimal formation. These findings provide the foundation for the translation of cell-targeted RNA therapeutics to vascular disease.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/terapia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Semivida , Humanos , Ratones , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Neointima/metabolismo , Fosforilación , Ratas
7.
Clin Sci (Lond) ; 130(3): 151-65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26678171

RESUMEN

Since its discovery in 1999, a number of studies have evaluated the role of Nox1 NADPH oxidase in the cardiovascular system. Nox1 is activated in vascular cells in response to several different agonists, with its activity regulated at the transcriptional level as well as by NADPH oxidase complex formation, protein stabilization and post-translational modification. Nox1 has been shown to decrease the bioavailability of nitric oxide, transactivate the epidermal growth factor receptor, induce pro-inflammatory signalling, and promote cell migration and proliferation. Enhanced expression and activity of Nox1 under pathologic conditions results in excessive production of reactive oxygen species and dysregulated cellular function. Indeed, studies using genetic models of Nox1 deficiency or overexpression have revealed roles for Nox1 in the pathogenesis of cardiovascular diseases ranging from atherosclerosis to hypertension, restenosis and ischaemia/reperfusion injury. These data suggest that Nox1 is a potential therapeutic target for vascular disease, and drug development efforts are ongoing to identify a specific bioavailable inhibitor of Nox1.


Asunto(s)
Enfermedades Cardiovasculares/etiología , NADPH Oxidasas/metabolismo , Animales , Enfermedades Cardiovasculares/enzimología , Humanos , Isoenzimas/metabolismo , Estructura Molecular , Terapia Molecular Dirigida , NADPH Oxidasa 1 , NADPH Oxidasas/química
8.
Circ Res ; 115(11): 911-8, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25228390

RESUMEN

RATIONALE: Activation of Nox1 initiates redox-dependent signaling events crucial in the pathogenesis of vascular disease. Selective targeting of Nox1 is an attractive potential therapy, but requires a better understanding of the molecular modifications controlling its activation. OBJECTIVE: To determine whether posttranslational modifications of Nox1 regulate its activity in vascular cells. METHODS AND RESULTS: We first found evidence that Nox1 is phosphorylated in multiple models of vascular disease. Next, studies using mass spectroscopy and a pharmacological inhibitor demonstrated that protein kinase C-beta1 mediates phosphorylation of Nox1 in response to tumor necrosis factor-α. siRNA-mediated silencing of protein kinase C-beta1 abolished tumor necrosis factor-α-mediated reactive oxygen species production and vascular smooth muscle cell migration. Site-directed mutagenesis and isothermal titration calorimetry indicated that protein kinase C-beta1 phosphorylates Nox1 at threonine 429. Moreover, Nox1 threonine 429 phosphorylation facilitated the association of Nox1 with the NoxA1 activation domain and was necessary for NADPH oxidase complex assembly, reactive oxygen species production, and vascular smooth muscle cell migration. CONCLUSIONS: We conclude that protein kinase C-beta1 phosphorylation of threonine 429 regulates activation of Nox1 NADPH oxidase.


Asunto(s)
NADH NADPH Oxidorreductasas/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Aorta/citología , Sitios de Unión , Movimiento Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Mutación , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Fosforilación , Unión Proteica , Proteína Quinasa C beta/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/química , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 34(10): 2301-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25147340

RESUMEN

OBJECTIVE: Reactive oxygen species regulate canonical Wnt signaling. However, the role of the redox regulatory protein p66(Shc) in the canonical Wnt pathway is not known. We investigated whether p66(Shc) is essential for canonical Wnt signaling in the endothelium and determined whether the canonical Wnt pathway induces vascular endothelial dysfunction via p66(Shc)-mediated oxidative stress. APPROACH AND RESULTS: The canonical Wnt ligand Wnt3a induced phosphorylation (activation) of p66(Shc) in endothelial cells. Wnt3a-stimulated dephosphorylation of ß-catenin, and ß-catenin-dependent transcription, was inhibited by knockdown of p66(Shc). Exogenous H2O2-induced ß-catenin dephosphorylation was also mediated by p66(Shc). Moreover, p66(Shc) overexpression dephosphorylated ß-catenin and increased ß-catenin-dependent transcription, independent of Wnt3a ligand. P66(Shc)-induced ß-catenin dephosphorylation was inhibited by antioxidants N-acetyl cysteine and catalase. Wnt3a upregulated endothelial NADPH oxidase-4, and ß-catenin dephosphorylation was suppressed by knocking down NADPH oxidase-4 and by antioxidants. Wnt3a increased H2O2 levels in endothelial cells and impaired endothelium-dependent vasorelaxation in mouse aortas, both of which were rescued by p66(Shc) knockdown. P66(Shc) knockdown also inhibited adhesion of monocytes to Wnt3a-stimulated endothelial cells. Furthermore, constitutively active ß-catenin expression in the endothelium increased vascular reactive oxygen species and impaired endothelium-dependent vasorelaxation. In vivo, high-fat diet feeding-induced endothelial dysfunction in mice was associated with increased endothelial Wnt3a, dephosphorylated ß-catenin, and phosphorylated p66(Shc). High-fat diet-induced dephosphorylation of endothelial ß-catenin was diminished in mice in which p66(Shc) was knocked down. CONCLUSIONS: p66(Shc) plays a vital part in canonical Wnt signaling in the endothelium and mediates Wnt3a-stimulated endothelial oxidative stress and dysfunction.


Asunto(s)
Células Endoteliales/enzimología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Bovinos , Técnicas de Cocultivo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Hiperlipidemias/enzimología , Hiperlipidemias/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fosforilación , Interferencia de ARN , Proteínas Adaptadoras de la Señalización Shc/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Transfección , Células U937 , Vasodilatación , Vasodilatadores/farmacología , Proteína Wnt3A/genética , beta Catenina/metabolismo
10.
Aust J Rural Health ; 23(3): 155-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25929462

RESUMEN

OBJECTIVE: To examine the timing of operative management and interhospital transfer of emergency general surgical patients in a regional setting. DESIGN: Retrospective cohort study. SETTING: The surgical unit at a major rural referral centre for North-Eastern Victoria servicing a population of 90 000. PARTICIPANTS: General surgical patients (n = 649) admitted via the emergency department at Northeast Health Wangaratta between January 2011 and March 2013 undergoing operative management (n = 608) or transfer to a tertiary centre (n = 44). MAIN OUTCOME MEASURES: Timing of operative management, using appendicectomy as a benchmark operation, was measured as time from presentation to decision to operate, time from decision to surgery, percentage after-hours operating and length of stay (LOS). Time to interhospital transfer was calculated and reasons for delay were sought. RESULTS: Two hundred forty-six appendicectomies were performed. Median time from decision to operate to theatre was 3 hours (interquartile range (IQR) 2-8), and total LOS was 43 hours (IQR: 28-56). Two hundred seventy-two procedures (43%) were performed out-of-hours, including 48% of appendicectomies. Median time from decision making to transfer was 10.3 hours (IQR: 4.7-25). Transfer was less likely to be delayed in trauma patients when compared with urgent non-trauma patients (5.3 versus 10.6 hours; P = 0.04). CONCLUSION: Even in the absence of a strict four-hour rule program and a dedicated emergency surgical unit, main outcome measures appear to be comparatively efficient. However, the duration for transfer of patients is suboptimal because of the lack of established pathways for urgent non-trauma transfer from rural centres and bed availability in tertiary hospitals.


Asunto(s)
Servicios de Salud Rural , Procedimientos Quirúrgicos Operativos , Listas de Espera , Adulto , Anciano , Bases de Datos Factuales , Servicio de Urgencia en Hospital , Femenino , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Asignación de Recursos , Estudios Retrospectivos , Centros Traumatológicos , Victoria
11.
Circulation ; 127(12): 1308-16, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23426106

RESUMEN

BACKGROUND: The incidence of thrombotic events increases during aging, but the mechanisms are not well understood. To investigate the prothrombotic role of oxidative stress during aging, we tested the hypothesis that aged mice overexpressing the antioxidant enzyme glutathione peroxidase-1 (Gpx1) are protected from experimental thrombosis. METHODS AND RESULTS: Susceptibility to carotid artery thrombosis was first examined in wild-type C57BL/6J mice. After photochemical injury of the carotid artery, the time to stable occlusion was significantly shorter in 12- and 18-month-old mice compared with 4-month-old mice (P<0.01). Unlike wild-type mice, transgenic mice overexpressing Gpx1 (Gpx1 Tg) did not exhibit shortened times to occlusion of the carotid artery at 12 or 18 months of age. Wild-type mice also exhibited increased susceptibility to venous thrombosis after inferior vena cava ligation at 12 or 18 months of age (P<0.05 versus 4 months of age). Gpx1 Tg mice were protected from this aging-related enhanced susceptibility to venous thrombosis. Age-dependent platelet hyperactivation, evidenced by increased hydrogen peroxide, fibrinogen binding, and activation of fibrinogen receptor αIIbß3, was observed in thrombin-activated platelets from wild-type but not Gpx1 Tg mice (P<0.05). Enhanced platelet activation responses in aged mice were also prevented by polyethylene glycol-catalase or apocynin, an inhibitor of NADPH oxidase. Aged mice displayed increased intraplatelet expression of p47(phox) and superoxide dismutase-1, suggesting a mechanistic pathway for increased hydrogen peroxide generation. CONCLUSIONS: Our findings demonstrate that hydrogen peroxide is a key mediator of platelet hyperactivity and enhanced thrombotic susceptibility in aged mice.


Asunto(s)
Envejecimiento/metabolismo , Plaquetas/metabolismo , Peróxido de Hidrógeno/metabolismo , Activación Plaquetaria/fisiología , Trombosis/epidemiología , Trombosis/metabolismo , Acetofenonas/farmacología , Animales , Catalasa/farmacología , Femenino , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Estrés Oxidativo/fisiología , Activación Plaquetaria/efectos de los fármacos , Polietilenglicoles/farmacología , Factores de Riesgo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Trombosis/fisiopatología , Glutatión Peroxidasa GPX1
12.
Int J Mol Sci ; 15(7): 11324-49, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24968272

RESUMEN

Chronic inflammatory diseases are associated with accelerated atherosclerosis and increased risk of cardiovascular diseases (CVD). As the pathogenesis of atherosclerosis is increasingly recognized as an inflammatory process, similarities between atherosclerosis and systemic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel diseases, lupus, psoriasis, spondyloarthritis and others have become a topic of interest. Endothelial dysfunction represents a key step in the initiation and maintenance of atherosclerosis and may serve as a marker for future risk of cardiovascular events. Patients with chronic inflammatory diseases manifest endothelial dysfunction, often early in the course of the disease. Therefore, mechanisms linking systemic inflammatory diseases and atherosclerosis may be best understood at the level of the endothelium. Multiple factors, including circulating inflammatory cytokines, TNF-α (tumor necrosis factor-α), reactive oxygen species, oxidized LDL (low density lipoprotein), autoantibodies and traditional risk factors directly and indirectly activate endothelial cells, leading to impaired vascular relaxation, increased leukocyte adhesion, increased endothelial permeability and generation of a pro-thrombotic state. Pharmacologic agents directed against TNF-α-mediated inflammation may decrease the risk of endothelial dysfunction and cardiovascular disease in these patients. Understanding the precise mechanisms driving endothelial dysfunction in patients with systemic inflammatory diseases may help elucidate the pathogenesis of atherosclerosis in the general population.


Asunto(s)
Artritis/metabolismo , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Animales , Artritis/fisiopatología , Aterosclerosis/fisiopatología , LDL-Colesterol/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Enfermedades Inflamatorias del Intestino/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Int J Mol Sci ; 15(4): 6002-8, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24722571

RESUMEN

In the special issue "Oxidative Stress in Cardiovascular Disease" authors were invited to submit papers that investigate key questions in the field of cardiovascular free radical biology. The original research articles included in this issue provide important information regarding novel aspects of reactive oxygen species (ROS)-mediated signaling, which have important implications in physiological and pathophysiological cardiovascular processes. The issue also included a number of review articles that highlight areas of intense research in the fields of free radical biology and cardiovascular medicine.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Estrés Oxidativo , Antioxidantes/uso terapéutico , Biomarcadores/análisis , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/patología , Humanos , Especies Reactivas de Oxígeno , Transducción de Señal
14.
Int J Mol Sci ; 15(11): 20079-100, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25375191

RESUMEN

BACKGROUND: Ventricular tachycardia or fibrillation (VT/VF) of focal origin due to triggered activity (TA) from delayed afterdepolarizations (DADs) is reproducibly inducible after anterior coronary artery occlusion. Both VT/VF and TA can be blocked by reducing reactive oxygen species (ROS). We tested the hypothesis that inhibition of NADPH oxidase and xanthine oxidase would block VT/VF. METHODS: 69 dogs received apocynin (APO), 4 mg/kg intraveneously (IV), oxypurinol (OXY), 4 mg/kg IV, or both APO and OXY (BOTH) agents, or saline 3 h after coronary occlusion. Endocardium from ischemic sites (3-D mapping) was sampled for Rac1 (GTP-binding protein in membrane NADPH oxidase) activation or standard microelectrode techniques. Results (mean±SE, * p<0.05): VT/VF originating from ischemic zones was blocked by APO in 6/10 *, OXY in 4/9 *, BOTH in 5/8 * or saline in 1/27; 11/16 VT/VFs blocked were focal. In isolated myocardium, TA was blocked by APO (10(-6) M) or OXY (10(-8) M). Rac1 levels in ischemic endocardium were decreased by APO or OXY. CONCLUSION: APO and OXY suppressed focal VT/VF due to DADs, but the combination of the drugs was not more effective than either alone. Both drugs inhibited ischemic Rac1 with inhibition by OXY suggesting ROS-induced ROS. The inability to totally prevent VT/VF suggests that other mechanisms also contribute to ischemic VT.


Asunto(s)
Isquemia Miocárdica/enzimología , NADPH Oxidasas/metabolismo , Taquicardia Ventricular/enzimología , Fibrilación Ventricular/enzimología , Xantina Oxidasa/metabolismo , Acetofenonas/farmacología , Acetofenonas/uso terapéutico , Potenciales de Acción/efectos de los fármacos , Animales , Western Blotting , Modelos Animales de Enfermedad , Perros , Femenino , Masculino , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/fisiopatología , NADPH Oxidasas/antagonistas & inhibidores , Oxipurinol/farmacología , Oxipurinol/uso terapéutico , Taquicardia Ventricular/complicaciones , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/fisiopatología , Ultrasonografía , Fibrilación Ventricular/complicaciones , Fibrilación Ventricular/tratamiento farmacológico , Fibrilación Ventricular/fisiopatología , Xantina Oxidasa/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
15.
J Thromb Haemost ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38815756

RESUMEN

BACKGROUND: COVID-19 can cause profound inflammation and coagulopathy, and while many mechanisms have been proposed, there is no known common pathway leading to a prothrombotic state. OBJECTIVES: From the beginning of the COVID-19 pandemic, elevated levels of extracellular histones have been found in plasma of patients infected with SARS-CoV-2. We hypothesized that platelet activation triggered by extracellular histones might represent a unifying mechanism leading to increased thrombin generation and thrombosis. METHODS: We utilized blood samples collected from an early clinical trial of hospitalized COVID-19 patients (NCT04360824) and recruited healthy subjects as controls. Using plasma samples, we measured the procoagulant and prothrombotic potential of circulating extracellular histones and extracellular vesicles (EVs). Platelet prothrombotic activity was assessed via thrombin generation potential and platelet thrombus growth. Circulating EVs were assessed for thrombin generation potential in vitro in plasma and enhancement of thrombotic susceptibility in vivo in mice. RESULTS: Compared with controls, COVID-19 patients had elevated plasma levels of citrullinated histone H3, cell-free DNA, nucleosomes, and EVs. Plasma from COVID-19 patients promoted platelet activation, platelet-dependent thrombin generation, thrombus growth under venous shear stress, and release of platelet-derived EVs. These prothrombotic effects of COVID-19 plasma were inhibited by an RNA aptamer that neutralizes both free and DNA-bound histones. EVs isolated from COVID-19 plasma enhanced thrombin generation in vitro and potentiated venous thrombosis in mice in vivo. CONCLUSION: We conclude that extracellular histones and procoagulant EVs drive the prothrombotic state in COVID-19 and that histone-targeted therapy may prove beneficial.

16.
J Hypertens ; 42(6): 984-999, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690903

RESUMEN

Nox1 signaling is a causal key element in arterial hypertension. Recently, we identified protein disulfide isomerase A1 (PDI) as a novel regulatory protein that regulates Nox1 signaling in VSMCs. Spontaneously hypertensive rats (SHR) have increased levels of PDI in mesenteric resistance arteries compared with Wistar controls; however, its consequences remain unclear. Herein, we investigated the role of PDI in mediating Nox1 transcriptional upregulation and its effects on vascular dysfunction in hypertension. We demonstrate that PDI contributes to the development of hypertension via enhanced transcriptional upregulation of Nox1 in vascular smooth muscle cells (VSMCs). We show for the first time that PDI sulfenylation by hydrogen peroxide contributes to EGFR activation in hypertension via increased shedding of epidermal growth factor-like ligands. PDI also increases intracellular calcium levels, and contractile responses induced by ANG II. PDI silencing or pharmacological inhibition in VSMCs significantly decreases EGFR activation and Nox1 transcription. Overexpression of PDI in VSMCs enhances ANG II-induced EGFR activation and ATF1 translocation to the nucleus. Mechanistically, PDI increases ATF1-induced Nox1 transcription and enhances the contractile responses to ANG II. Herein we show that ATF1 binding to Nox1 transcription putative regulatory regions is augmented by PDI. Altogether, we provide evidence that HB-EGF in SHR resistance vessels promotes the nuclear translocation of ATF1, under the control of PDI, and thereby induces Nox1 gene expression and increases vascular reactivity. Thus, PDI acts as a thiol redox-dependent enhancer of vascular dysfunction in hypertension and could represent a novel therapeutic target for the treatment of this disease.


Asunto(s)
Hipertensión , Músculo Liso Vascular , NADPH Oxidasa 1 , Proteína Disulfuro Isomerasas , Ratas Endogámicas SHR , Regulación hacia Arriba , Animales , Proteína Disulfuro Isomerasas/metabolismo , Proteína Disulfuro Isomerasas/genética , NADPH Oxidasa 1/metabolismo , NADPH Oxidasa 1/genética , Hipertensión/fisiopatología , Hipertensión/genética , Hipertensión/metabolismo , Ratas , Músculo Liso Vascular/metabolismo , Masculino , Miocitos del Músculo Liso/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/genética , Ratas Wistar , Transcripción Genética
17.
Toxicol Appl Pharmacol ; 272(3): 736-45, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23917044

RESUMEN

Most head and neck squamous cell carcinomas (HNSCCs) overexpress epidermal growth factor receptor (EGFR) and EGFR inhibitors are routinely used in the treatment of HNSCC. However, many HNSCC tumors do not respond or become refractory to EGFR inhibitors. Autophagy, which is a stress-induced cellular self-degradation process, has been reported to reduce the efficacy of chemotherapy in various disease models. The purpose of this study is to determine if the efficacy of the EGFR inhibitor erlotinib is reduced by activation of autophagy via NOX4-mediated oxidative stress in HNSCC cells. Erlotinib induced the expression of the autophagy marker LC3B-II and autophagosome formation in FaDu and Cal-27 cells. Inhibition of autophagy by chloroquine and knockdown of autophagy pathway genes Beclin-1 and Atg5 sensitized both cell lines to erlotinib-induced cytotoxicity, suggesting that autophagy may serve as a protective mechanism. Treatment with catalase (CAT) and diphenylene iodonium (DPI) in the presence of erlotinib suppressed the increase in LC3B-II expression in FaDu and Cal-27 cells. Erlotinib increased NOX4 mRNA and protein expression by increasing its promoter activity and mRNA stability in FaDu cells. Knockdown of NOX4 using adenoviral siNOX4 partially suppressed erlotinib-induced LC3B-II expression, while overexpression of NOX4 increased expression of LC3B-II. These studies suggest that erlotinib may activate autophagy in HNSCC cells as a pro-survival mechanism, and NOX4 may play a role in mediating this effect.


Asunto(s)
Autofagia/fisiología , Carcinoma de Células Escamosas/metabolismo , Citoprotección/fisiología , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , NADPH Oxidasas/fisiología , Quinazolinas/farmacología , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/enzimología , Citoprotección/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib , Células HEK293 , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/enzimología , Humanos , NADPH Oxidasa 4 , Quinazolinas/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello , Células Tumorales Cultivadas
18.
FASEB J ; 26(6): 2531-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22375019

RESUMEN

ß-Adrenergic receptor (AR) blockers provide substantial clinical benefits, including improving overall survival and left ventricular (LV) function following myocardial infarction (MI), though the mechanisms remain incompletely defined. The transverse-tubule (T-tubule) system of ventricular myocytes is an important determinant of cardiac excitation-contraction function. T-tubule remodeling occurs early during LV failure. We hypothesized that ß-AR blockers prevent T-tubule remodeling and thereby provide therapeutic benefits. A murine model of MI was utilized to examine the effect of ß-AR blockers on T-tubule remodeling following LV MI. We applied the in situ imaging of T-tubule structure from Langendorff-perfused intact hearts with laser scanning confocal microscopy. We found that MI caused remarkable T-tubule remodeling near the infarction border zone and moderate LV remodeling remote from the MI. Metoprolol and carvedilol administered 6 d after MI for 4 wk each increased the T-tubule integrity at the remote and border zones. At the molecular level, both ß-AR blockers restored border and remote zone expression of junctophilin-2 (JP-2), which is involved in T-tubule organization and formation of the T-tubule/sarcoplasmic reticulum junctions. In contrast, ß-AR blockers had no significant effects on caveolin-3 expression. In summary, our data show that ß-AR antagonists can protect against T-tubule remodeling after MI, suggesting a novel therapeutic mechanism of action for this drug class. Preservation of JP-2 expression may contribute to the beneficial effects of metoprolol and carvedilol on T-tubule remodeling.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Remodelación Ventricular/efectos de los fármacos , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Carbazoles/farmacología , Carvedilol , Caveolina 3/biosíntesis , Masculino , Proteínas de la Membrana/biosíntesis , Metoprolol/farmacología , Ratones , Infarto del Miocardio/tratamiento farmacológico , Propanolaminas/farmacología , Remodelación Ventricular/fisiología
19.
Arterioscler Thromb Vasc Biol ; 32(10): 2452-60, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22879585

RESUMEN

OBJECTIVE: To characterize the relationship between the expression of epidermal growth factor (EGF)-like ligands and vascular nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and activity in a primate model of atherosclerosis. METHODS AND RESULTS: Adult male Cynomolgus monkeys were fed a normal or atherogenic (AS) diet for 45 months, after which animals from the AS group were placed on a normal diet for 8 months (regression). The expression of membrane-associated EGF-like ligands was increased in arteries from animals on the AS diet and normalized in the regression group. EGF-like ligands were distributed throughout atherosclerotic vessels but predominantly colocalized with macrophages. Consistent with ligand shedding, circulating heparin-bound EGF was elevated in the plasma of AS monkeys but not in those on regression diet. Atherosclerosis was associated with the activation of EGF receptor signaling. Expression of NADPH oxidase subunits Nox1 and Nox2 but not Nox4 or Nox5 was increased in arteries from monkeys on the AS diet and returned to normal with regression. Levels of Nox1 and Nox2 positively correlated with EGF-like ligands. In cultured monkey smooth muscle cells, treatment with EGF-like ligands increased Nox1 expression and activity. CONCLUSIONS: These data identify EGF-like ligands as potential modulators of atherogenesis, resulting in part from increased vascular NADPH oxidase activity.


Asunto(s)
Arterias/metabolismo , Aterosclerosis/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , NADPH Oxidasas/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/patología , Células Cultivadas , Dieta Aterogénica/efectos adversos , Modelos Animales de Enfermedad , Ligandos , Macaca fascicularis , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Transducción de Señal
20.
Int J Mol Sci ; 14(10): 20597-613, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24132149

RESUMEN

The epidermal growth factor receptor (EGFR) family and its ligands serve as a switchboard for the regulation of multiple cellular processes. While it is clear that EGFR activity is essential for normal cardiac development, its function in the vasculature and its role in cardiovascular disease are only beginning to be elucidated. In the blood vessel, endothelial cells and smooth muscle cells are both a source and a target of EGF-like ligands. Activation of EGFR has been implicated in blood pressure regulation, endothelial dysfunction, neointimal hyperplasia, atherogenesis, and cardiac remodeling. Furthermore, increased circulating EGF-like ligands may mediate accelerated vascular disease associated with chronic inflammation. Although EGFR inhibitors are currently being used clinically for the treatment of cancer, additional studies are necessary to determine whether abrogation of EGFR signaling is a potential strategy for the treatment of cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Receptores ErbB/metabolismo , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Ligandos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA