Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 14(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38927040

RESUMEN

Metabolic syndrome (MetS) is a cluster of metabolic abnormalities affecting ~25% of adults and is linked to chronic diseases such as cardiovascular disease, cancer, and neurodegenerative diseases. Oxidative stress and inflammation are key drivers of MetS. Hesperidin, a citrus bioflavonoid, has demonstrated antioxidant and anti-inflammatory properties; however, its effects on MetS are not fully established. We aimed to determine the optimal dose of hesperidin required to improve oxidative stress, systemic inflammation, and glycemic control in a novel mouse model of MetS. Male 5-week-old C57BL/6 mice were fed a high-fat, high-salt, high-sugar diet (HFSS; 42% kcal fat content in food and drinking water with 0.9% saline and 10% high fructose corn syrup) for 16 weeks. After 6 weeks of HFSS, mice were randomly allocated to either the placebo group or low- (70 mg/kg/day), mid- (140 mg/kg/day), or high-dose (280 mg/kg/day) hesperidin supplementation for 12 weeks. The HFSS diet induced significant metabolic disturbances. HFSS + placebo mice gained almost twice the weight of control mice (p < 0.0001). Fasting blood glucose (FBG) increased by 40% (p < 0.0001), plasma insulin by 100% (p < 0.05), and HOMA-IR by 150% (p < 0.0004), indicating insulin resistance. Hesperidin supplementation reduced plasma insulin by 40% at 140 mg/kg/day (p < 0.0001) and 50% at 280 mg/kg/day (p < 0.005). HOMA-IR decreased by 45% at both doses (p < 0.0001). Plasma hesperidin levels significantly increased in all hesperidin groups (p < 0.0001). Oxidative stress, measured by 8-OHdG, was increased by 40% in HFSS diet mice (p < 0.001) and reduced by 20% with all hesperidin doses (p < 0.005). In conclusion, hesperidin supplementation reduced insulin resistance and oxidative stress in HFSS-fed mice, demonstrating its dose-dependent therapeutic potential in MetS.


Asunto(s)
Citrus , Suplementos Dietéticos , Modelos Animales de Enfermedad , Hesperidina , Resistencia a la Insulina , Síndrome Metabólico , Ratones Endogámicos C57BL , Estrés Oxidativo , Animales , Hesperidina/farmacología , Estrés Oxidativo/efectos de los fármacos , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Masculino , Ratones , Citrus/química , Relación Dosis-Respuesta a Droga , Glucemia/metabolismo , Glucemia/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Antioxidantes/farmacología
2.
Front Cardiovasc Med ; 11: 1419958, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38883991

RESUMEN

Introduction: Marginal zone and follicular B cells are known to contribute to the development of angiotensin II-induced hypertension in mice, but the effector function(s) mediating this effect (e.g., antigen presentation, antibody secretion and/or cytokine production) are unknown. B cell differentiation into antibody secreting cells (ASCs) requires the transcription factor Blimp-1. Here, we studied mice with a Blimp-1 deficiency in follicular B cells to evaluate whether antibody secretion underlies the pro-hypertensive action of B cells. Methods: 10- to 14-week-old male follicular B cell Blimp-1 knockout (FoB-Blimp-1-KO) and floxed control mice were subcutaneously infused with angiotensin II (0.7 mg/kg/d) or vehicle (0.1% acetic acid in saline) for 28 days. BP was measured by tail-cuff plethysmography or radiotelemetry. Pulse wave velocity was measured by ultrasound. Aortic collagen was quantified by Masson's trichrome staining. Cell types and serum antibodies were quantified by flow cytometry and a bead-based multiplex assay, respectively. Results: In control mice, angiotensin II modestly increased serum IgG3 levels and markedly increased BP, cardiac hypertrophy, aortic stiffening and fibrosis. FoB-Blimp-1-KO mice exhibited impaired IgG1, IgG2a and IgG3 production despite having comparable numbers of B cells and ASCs to control mice. Nevertheless, FoB-Blimp-1-KO mice still developed hypertension, cardiac hypertrophy, aortic stiffening and fibrosis following angiotensin II infusion. Conclusions: Inhibition of follicular B cell differentiation into ASCs did not protect against angiotensin II-induced hypertension or vascular compliance. Follicular B cell functions independent of their differentiation into ASCs and ability to produce high-affinity antibodies, or other B cell subtypes, are likely to be involved in angiotensin II-induced hypertension.

3.
Hypertension ; 66(5): 1023-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351030

RESUMEN

Clinical hypertension is associated with raised serum IgG antibodies. However, whether antibodies are causative agents in hypertension remains unknown. We investigated whether hypertension in mice is associated with B-cell activation and IgG production and moreover whether B-cell/IgG deficiency affords protection against hypertension and vascular remodeling. Angiotensin II (Ang II) infusion (0.7 mg/kg per day; 28 days) was associated with (1) a 25% increase in the proportion of splenic B cells expressing the activation marker CD86, (2) an 80% increase in splenic plasma cell numbers, (3) a 500% increase in circulating IgG, and (4) marked IgG accumulation in the aortic adventitia. In B-cell-activating factor receptor-deficient (BAFF-R(-/-)) mice, which lack mature B cells, there was no evidence of Ang II-induced increases in serum IgG. Furthermore, the hypertensive response to Ang II was attenuated in BAFF-R(-/-) (Δ30±4 mm Hg) relative to wild-type (Δ41±5 mm Hg) mice, and this response was rescued by B-cell transfer. BAFF-R(-/-) mice displayed reduced IgG accumulation in the aorta, which was associated with 80% fewer aortic macrophages and a 70% reduction in transforming growth factor-ß expression. BAFF-R(-/-) mice were also protected from Ang II-induced collagen deposition and aortic stiffening (assessed by pulse wave velocity analysis). Finally, like BAFF-R deficiency, pharmacological depletion of B cells with an anti-CD20 antibody attenuated Ang II-induced hypertension by ≈35%. Hence, these studies demonstrate that B cells/IgGs are crucial for the development of Ang II-induced hypertension and vessel remodeling in mice. Thus, B-cell-targeted therapies-currently used for autoimmune diseases-may hold promise as future treatments for hypertension.


Asunto(s)
Angiotensina II/efectos adversos , Linfocitos B/patología , Linfocitos B/fisiología , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Rigidez Vascular/fisiología , Traslado Adoptivo , Animales , Anticuerpos Antiidiotipos/farmacología , Antígenos CD20/inmunología , Receptor del Factor Activador de Células B/deficiencia , Receptor del Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/metabolismo , Linfocitos B/efectos de los fármacos , Proliferación Celular , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Ratones Noqueados , Bazo/patología , Factor de Crecimiento Transformador beta/metabolismo
4.
Circ Res ; 116(2): 245-54, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25398236

RESUMEN

RATIONALE: CD4(+) natural killer T (NKT) cells augment atherosclerosis in apolipoprotein E-deficient (ApoE)(-/-) mice but their mechanisms of action are unknown. OBJECTIVES: We investigated the roles of bystander T, B, and NK cells; NKT cell-derived interferon-γ, interleukin (IL)-4, and IL-21 cytokines; and NKT cell-derived perforin and granzyme B cytotoxins in promoting CD4(+) NKT cell atherogenicity. METHODS AND RESULTS: Transfer of CD4(+) NKT cells into T- and B-cell-deficient ApoE(-/-)Rag2(-/-) mice augmented aortic root atherosclerosis by ≈75% that was ≈30% of lesions in ApoE(-/-) mice; macrophage accumulation similarly increased. Transferred NKT cells were identified in the liver and atherosclerotic lesions of recipient mice. Transfer of CD4(+) NKT cells into T-, B-cell-deficient, and NK cell-deficient ApoE(-/-)Rag2(-/-)γC(-/-) mice also augmented atherosclerosis. These data indicate that CD4(+) NKT cells can exert proatherogenic effects independent of other lymphocytes. To investigate the role of NKT cell-derived interferon-γ, IL-4, and IL-21 cytokines and perforin and granzyme B cytotoxins, CD4(+) NKT cells from mice deficient in these molecules were transferred into NKT cell-deficient ApoE(-/-)Jα18(-/-) mice. CD4(+) NKT cells deficient in IL-4, interferon-γ, or IL-21 augmented atherosclerosis in ApoE(-/-)Jα18(-/-) mice by ≈95%, ≈80%, and ≈70%, respectively. Transfer of CD4(+) NKT cells deficient in perforin or granzyme B failed to augment atherosclerosis. Apoptotic cells, necrotic cores, and proinflammatory VCAM-1 (vascular cell adhesion molecule) and MCP-1 (monocyte chemotactic protein) were reduced in mice receiving perforin-deficient NKT cells. CD4(+) NKT cells are twice as potent as CD4(+) T cells in promoting atherosclerosis. CONCLUSIONS: CD4(+) NKT cells potently promote atherosclerosis by perforin and granzyme B-dependent apoptosis that increases postapoptotic necrosis and inflammation.


Asunto(s)
Aterosclerosis/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Granzimas/deficiencia , Células T Asesinas Naturales/metabolismo , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Seno Aórtico/metabolismo , Traslado Adoptivo/métodos , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Linfocitos T CD4-Positivos/inmunología , Masculino , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Seno Aórtico/inmunología , Seno Aórtico/patología
6.
Biomed Res Int ; 2014: 504045, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25050352

RESUMEN

It has long been known that circulating levels of IgG and IgM antibodies are elevated in patients with essential and pregnancy-related hypertension. Recent studies indicate these antibodies target, and in many cases activate, G-protein coupled receptors and ion channels. Prominent among these protein targets are AT1 receptors, α1-adrenoceptors, ß1-adrenoceptors, and L-type voltage operated Ca(2+) channels, all of which are known to play key roles in the regulation of blood pressure through modulation of vascular tone, cardiac output, and/or Na(+)/water reabsorption in the kidneys. This suggests that elevated antibody production may be a causal mechanism in at least some cases of hypertension. In this brief review, we will further describe the protein targets of the antibodies that are elevated in individuals with essential and pregnancy-related hypertension and the likely pathophysiological consequences of antibody binding to these targets. We will speculate on the potential mechanisms that underlie elevated antibody levels in hypertensive individuals and, finally, we will outline the therapeutic opportunities that could arise with a better understanding of how and why antibodies are produced in hypertension.


Asunto(s)
Anticuerpos/inmunología , Hipertensión/etiología , Hipertensión/inmunología , Animales , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Humanos , Hipertensión/terapia
8.
Arterioscler Thromb Vasc Biol ; 34(3): 516-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24436372

RESUMEN

OBJECTIVE: Atherosclerosis is driven by inflammatory reactions that are shared with the innate immune system. Toll-like receptor-9 (TLR9) is an intracellular pattern recognition receptor of the innate immune system that is currently under clinical investigation as a therapeutic target in inflammatory diseases. Here, we investigated whether TLR9 has a role in the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Newly generated double-knockout ApoE(-/-):TLR9(-/-) mice and control ApoE(-/-) mice were fed a high-fat diet from 8 weeks and effects on lesion size, cellular composition, inflammatory status, and plasma lipids were assessed after 8, 12, 15, and 20 weeks. All 4 time points demonstrated exacerbated atherosclerotic lesion severity in ApoE(-/-):TLR9(-/-) mice, with a corresponding increase in lipid deposition and accumulation of macrophages, dendritic cells, and CD4(+) T cells. Although ApoE(-/-):TLR9(-/-) mice exhibited an increase in plasma very low-density lipoprotein/low-density-lipoprotein cholesterol, the very low-density lipoprotein/low-density lipoprotein:high-density lipoprotein ratio was unaltered because of a parallel increase in plasma high-density lipoprotein cholesterol. As a potential mechanism accounting for plaque progression in ApoE(-/-):TLR9(-/-) mice, CD4(+) T-cell accumulation was further investigated and depletion of these cells in ApoE(-/-):TLR9(-/-) mice significantly reduced lesion severity. As a final translational approach, administration of a TLR9 agonist (type B CpG oligodeoxynucleotide 1668) to ApoE(-/-) mice resulted in a reduction of lesion severity. CONCLUSIONS: Genetic deletion of the innate immune receptor TLR9 exacerbated atherosclerosis in ApoE(-/-) mice fed a high-fat diet. CD4(+) T cells were identified as potential mediators of this effect. A type B CpG oligodeoxynucleotide TLR9 agonist reduced lesion severity, thus identifying a novel therapeutic approach in atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Receptor Toll-Like 9/fisiología , Animales , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/sangre , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/patología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Grasas de la Dieta/toxicidad , Progresión de la Enfermedad , Lipoproteínas HDL/sangre , Lipoproteínas VLDL/sangre , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos/farmacología , Oligodesoxirribonucleótidos/uso terapéutico , Distribución Aleatoria , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
10.
Circulation ; 126(10): 1256-66, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22851544

RESUMEN

BACKGROUND: CD4+CD25+Foxp3+ regulatory T cells (Tregs) attenuate atherosclerosis, but their therapeutic application by adoptive transfer is limited by the need for their expansion in vitro and limited purity. Recently, an interleukin (IL)-2/anti-IL-2 neutralizing monoclonal antibody (IL-2/anti-IL-2 mAb) complex has been shown to expand these Tregs. We examined the capacity of a modified IL-2/anti-IL-2 mAb treatment to expand Tregs and inhibit both the progression and development of developed atherosclerosis. METHODS AND RESULTS: Six-week old apolipoprotein E-deficient mice fed a high-fat diet for 8 weeks were administered IL-2/anti-IL-2 mAb commencing 2 weeks after starting the diet. Tregs in the spleen, lymph node, and liver were selectively expanded without affecting CD4+, CD8+, or natural killer cells. Tregs were increased in lesions and lesion size reduced. CD4+ T-cells, macrophages, mature dendritic cells, proliferating cell nuclear antigen+ cells, and monocyte chemoattractant protein-1 and vascular cell adhesion molecule-1 were reduced. In anti-CD3-stimulated splenocytes, proliferation and secretion of Th1, Th2, and Th17 (IL-17) cytokines and IL-1ß were reduced. To determine whether treatment attenuated progression of developed atherosclerosis, 6-week-old apolipoprotein E-deficient mice were fed a high-fat diet for 6 weeks, followed by IL-2/anti-IL-2 mAb treatment for 6 weeks while continuing the high-fat diet. Treatment also increased Tregs without affecting CD4+, CD8+, or natural killer cells, suppressed inflammation, and greatly attenuated progression of atherosclerosis. CONCLUSIONS: IL-2/anti-IL-2 mAb treatment in vivo attenuates atherosclerosis via selective Tregs expansion. The findings suggest that cytokine-based IL-2/anti-IL-2 mAb complex therapy could represent an attractive approach for treating atherosclerosis, because it markedly attenuates progression as well as development, by modulating its immunoinflammatory component.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Complejo Antígeno-Anticuerpo/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Interleucina-2/farmacología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Complejo Antígeno-Anticuerpo/inmunología , Apolipoproteínas E/genética , Biomarcadores/metabolismo , Antígenos CD4/metabolismo , División Celular/efectos de los fármacos , División Celular/inmunología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Relación Dosis-Respuesta Inmunológica , Factores de Transcripción Forkhead/metabolismo , Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Ratones , Ratones Mutantes , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
11.
Cardiovasc Res ; 95(1): 77-85, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22492684

RESUMEN

AIMS: The mechanisms underlying cardiac fibrosis in hypertension are yet to be defined, although inflammatory cells, fibroblasts, and cytokines have been implicated. Here, we investigated the role of interleukin-4 (IL-4) in cardiac fibrosis, which is elevated in the hypertensive heart. IL-4 has been shown to be pro-fibrotic in the liver and the lung, but its role in cardiac fibrosis has not been investigated. METHODS AND RESULTS: Cardiac fibrosis was induced in mice by constricting the aorta between the two carotid arteries. Fourteen days later marked left ventricular fibrosis developed together with expression of IL-4. Anti-IL-4 neutralizing antibodies attenuated this fibrosis without affecting blood pressure or expression of the transforming growth factor-beta system. The reduction in fibrosis was associated with reductions in interstitial fibroblasts and macrophages together with reductions in proliferating cells and expression of monocyte chemoattractant protein-1 (MCP-1). Since mast cells are a source of IL-4, we also assessed their role in fibrosis. Cromolyn, a mast cell inhibitor attenuated mast cell degranulation as well as IL-4 mRNA expression and cardiac fibrosis without affecting blood pressure. Treatment with Cromolyn also reduced interstitial fibroblasts and macrophages in regions of developing fibrosis as well MCP-1 expression. CONCLUSION: This study demonstrates for the first time that IL-4, most likely produced by mast cells in the heart during pressure overload, is a significant contributor to cardiac fibrosis. Targeting this cytokine may be a useful therapeutic strategy to limit cardiac fibrosis.


Asunto(s)
Hipertensión/patología , Interleucina-4/fisiología , Miocardio/patología , Animales , Coartación Aórtica/etiología , Proliferación Celular , Fibrosis , Macrófagos/fisiología , Masculino , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/fisiología
13.
J Hypertens ; 29(9): 1820-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21785365

RESUMEN

BACKGROUND: CD4⁺CD25⁺Foxp3⁺ regulatory T cells (Tregs) are potent inhibitors of inflammation and autoimmune diseases. Because inflammation has been associated with development of cardiac fibrosis in experimental hypertension, here we investigated whether adoptively transferred Tregs would inhibit development of cardiac fibrosis initiated by elevating blood pressure. METHODS: Cardiac fibrosis was induced in mice by constricting the aorta between the two carotid arteries. Immediately after the operation mice received either vehicle or purified, cultured Tregs (1.5 × 106). Fourteen days later we assessed effects on developing left ventricular fibrosis, blood pressure, inflammation, myofibroblasts and the transforming growth factor-beta1 (TGF-ß1) system. RESULTS: Fourteen days after aortic constriction, marked left-ventricular fibrosis was apparent and this was greatly reduced in mice receiving adoptively transferred Tregs. This reduction in fibrosis was associated with attenuated inflammatory cell numbers, reduced interstitial myofibroblast numbers and attenuated activity of the TGF-ß1 system, indicated by reductions in the expression of TGF-ß1 and its receptors activin-like kinase-5 and type II TGF-ß receptor. Adoptively transferred Tregs did not affect blood pressure and exerted only a small effect on left-ventricular hypertrophy. CONCLUSIONS: These data indicate that Tregs attenuate cardiac fibrosis associated with hypertensive heart disease by suppressing inflammation.


Asunto(s)
Antígenos CD4/inmunología , Factores de Transcripción Forkhead/inmunología , Corazón , Hipertensión/patología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Linfocitos T Reguladores/inmunología , Animales , Fibrosis , Hipertensión/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Br J Pharmacol ; 160(8): 2085-96, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20649603

RESUMEN

BACKGROUND AND PURPOSE: Darbepoetin, a long-acting erythropoietin derivative, attenuates cardiomyocyte apoptosis and improves short-term (3 days) cardiac function, but the mechanisms responsible are unknown. We investigated potential mechanisms by which darbepoetin exerts cardioprotection following myocardial infarction in mice and the significance of the erythropoietin receptor (EPOR)-common beta-chain (c-beta-chain) heteroreceptor. EXPERIMENTAL APPROACH: Mice underwent 60 min coronary occlusion followed by treatment with vehicle or a single dose of darbepoetin. Effects on gene expression, apoptosis and neutrophil accumulation in infarcted left ventricle were assessed 24 h later. Cardiac function, effects on vascularization and fibrosis were assessed 28 days later. The significance of EPOR-c-beta-chain heteroreceptor was examined 28 days after infarction using mice deficient in c-beta-chain. KEY RESULTS: Twenty-four hours after darbepoetin, mRNAs encoding haeme oxygenase-1 (HO-1), iNOS and brain natriuretic peptide (BNP) were markedly elevated only in infarcted regions, and the frequency of apoptotic cells attenuated. Inflammation was also attenuated with reductions in neutrophil numbers. Darbepoetin also elevated mRNAs encoding angiogenic factors: placental growth factor, monocyte chemoattractant protein-1 and interleukin-1beta. Twenty-eight days after treatment, CD31+ vessels in the infarct zone doubled and fibrosis reduced. Cardiac haemodynamics were improved. Darbepoetin also improved cardiac haemodynamics in c-beta-chain-deficient mice, increased HO-1 and iNOS expression and vessel numbers and attenuated fibrosis. CONCLUSIONS AND IMPLICATIONS: Darbepoetin stimulates expression of haeme oxygenase, iNOS, BNP and angiogenic factors specifically in infarcted left ventricle that attenuates inflammation, apoptosis and fibrosis; elevate vessel numbers; and improve cardiac function. The EPOR-c-beta-chain heteroreceptor is not essential for these effects.


Asunto(s)
Cardiotónicos/farmacología , Eritropoyetina/análogos & derivados , Hemodinámica/efectos de los fármacos , Infarto del Miocardio/dietoterapia , Miocardio/patología , Receptores de Eritropoyetina/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Darbepoetina alfa , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Eritropoyetina/farmacología , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Péptido Natriurético Encefálico/genética , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Infiltración Neutrófila/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Receptores de Eritropoyetina/deficiencia , Receptores de Eritropoyetina/genética , Factores de Tiempo
17.
Circ Res ; 103(4): 378-87, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18617696

RESUMEN

Activation transcription factor (ATF)-4 is a member of the ATF/CREB family of basic leucine zipper transcription factors that regulates cellular responses to a variety of stresses. The role of ATF-4 in smooth muscle cells of the vessel wall is completely unknown. Here, we show that ATF-4 expression is induced in smooth muscle cells in response to injury, both in vitro using a model of mechanical injury and in the media of balloon-injured rat carotid arteries. We demonstrate that ATF-4 is activated by fibroblast growth factor (FGF)-2, an injury-induced mitogen, through the phosphatidylinositol 3-kinase pathway. Injury also activates vascular endothelial growth factor (VEGF)-A, whose expression is stimulated by ATF-4 overexpression and exposure to FGF-2. FGF-2 induces ATF-4 binding to a recognition element located in the VEGF-A gene at +1767 bp and luciferase reporter gene expression dependent on this site. Moreover, ATF-4 knockdown with small interfering RNA or ATF-4 deficiency ameliorates FGF-2-inducible VEGF-A expression. Intraluminal delivery of ATF-4 small interfering RNA in rat carotid arteries blocks balloon injury-inducible ATF-4 and VEGF-A expression after 4 hours and intimal thickening after 14 days. These findings reveal, for the first time, the induction of ATF-4 by both vascular injury and FGF-2. ATF-4 serves as a conduit for the inducible expression of 1 growth factor by another during the process of intimal thickening.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Cateterismo/efectos adversos , Endotelio Vascular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Túnica Íntima/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Aorta/lesiones , Aorta/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/lesiones , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/lesiones , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Túnica Íntima/citología , Túnica Íntima/lesiones , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo
18.
Circ Res ; 101(2): 146-55, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17556661

RESUMEN

Vascular injury initiates a cascade of phenotype-altering molecular events. Transcription factor function in this process, particularly that of negative regulators, is poorly understood. We demonstrate here that the forced expression of the injury-inducible GLI-Krüppel zinc finger protein Yin Yang-1 (YY1) inhibits neointima formation in human, rabbit and rat blood vessels. YY1 inhibits p21(WAF1/Cip1) transcription, prevents assembly of a p21(WAF1/Cip1)-cdk4-cyclin D1 complex, and blocks downstream pRb(Ser249/Thr252) phosphorylation and expression of PCNA and TK-1. Conversely, suppression of endogenous YY1 elevates levels of p21(WAF1/Cip1), PCNA, pRb(Ser249/Thr252) and TK-1, and increases intimal thickening. YY1 binds Sp1 and prevents its occupancy of a distinct element in the p21(WAF1/Cip1) promoter without YY1 itself binding the promoter. Additionally, YY1 induces ubiquitination and proteasome-dependent degradation of p53, decreasing p53 immunoreactivity in the artery wall. These findings define a new role for YY1 as both an inducer of p53 instability in smooth muscle cells, and an indirect repressor of p21(WAF1/Cip1) transcription, p21(WAF1/Cip1)-cdk4-cyclin D1 assembly and intimal thickening.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/metabolismo , Complejos Multiproteicos/metabolismo , Miocitos del Músculo Liso/metabolismo , Túnica Íntima/crecimiento & desarrollo , Factor de Transcripción YY1/metabolismo , Animales , Arterias/citología , Arterias/crecimiento & desarrollo , Línea Celular , Ciclina D , Quinasa 4 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Ciclinas/genética , Regulación de la Expresión Génica/fisiología , Humanos , Complejos Multiproteicos/genética , Miocitos del Músculo Liso/citología , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica/fisiología , Conejos , Ratas , Elementos de Respuesta/fisiología , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Túnica Íntima/citología , Factor de Transcripción YY1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA