Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 42(2): 113-126, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34852642

RESUMEN

OBJECTIVE: IGF-1 (insulin-like growth factor 1) exerts pleiotropic effects including promotion of cellular growth, differentiation, survival, and anabolism. We have shown that systemic IGF-1 administration reduced atherosclerosis in Apoe-/- (apolipoprotein E deficient) mice, and this effect was associated with a reduction in lesional macrophages and a decreased number of foam cells in the plaque. Almost all cell types secrete IGF-1, but the effect of macrophage-derived IGF-1 on the pathogenesis of atherosclerosis is poorly understood. We hypothesized that macrophage-derived IGF-1 will reduce atherosclerosis. Approach and Results: We created macrophage-specific IGF-1 overexpressing mice on an Apoe-/- background. Macrophage-specific IGF-1 overexpression reduced plaque macrophages, foam cells, and atherosclerotic burden and promoted features of stable atherosclerotic plaque. Macrophage-specific IGF1 mice had a reduction in monocyte infiltration into plaque, decreased expression of CXCL12 (CXC chemokine ligand 12), and upregulation of ABCA1 (ATP-binding cassette transporter 1), a cholesterol efflux regulator, in atherosclerotic plaque and in peritoneal macrophages. IGF-1 prevented oxidized lipid-induced CXCL12 upregulation and foam cell formation in cultured THP-1 macrophages and increased lipid efflux. We also found an increase in cholesterol efflux in macrophage-specific IGF1-derived peritoneal macrophages. CONCLUSIONS: Macrophage IGF-1 overexpression reduced atherosclerotic burden and increased features of plaque stability, likely via a reduction in CXCL12-mediated monocyte recruitment and an increase in ABCA1-dependent macrophage lipid efflux.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Quimiocina CXCL12/sangre , Factor I del Crecimiento Similar a la Insulina/genética , Macrófagos/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/patología , Quimiocina CXCL12/análisis , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Ratas , Células THP-1 , Regulación hacia Arriba
2.
Am J Physiol Heart Circ Physiol ; 319(4): H730-H743, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32795184

RESUMEN

Insulin-like growth factor-1 (IGF-1) decreases atherosclerosis in apolipoprotein E (Apoe)-deficient mice when administered systemically. However, mechanisms for its atheroprotective effect are not fully understood. We generated endothelium-specific IGF-1 receptor (IGF1R)-deficient mice on an Apoe-deficient background to assess effects of IGF-1 on the endothelium in the context of hyperlipidemia-induced atherosclerosis. Endothelial deficiency of IGF1R promoted atherosclerotic burden, when animals were fed on a high-fat diet for 12 wk or normal chow for 12 mo. Under the normal chow feeding condition, the vascular relaxation response to acetylcholine was increased in the endothelial IGF1R-deficient aorta; however, feeding of a high-fat diet substantially attenuated the relaxation response, and there was no difference between endothelial IGF1R-deficient and control mice. The endothelium and its intercellular junctions provide a barrier function to the vasculature. In human aortic endothelial cells, IGF-1 upregulated occludin, claudin 5, VE-cadherin, JAM-A, and CD31 expression levels, and vice versa, specific IGF1R inhibitor, picropodophyllin, an IGF1R-neutralizing antibody (αIR3), or siRNA to IGF1R abolished the IGF-1 effects on junction and adherens proteins, suggesting that IGF-1 promoted endothelial barrier function. Moreover, endothelial transwell permeability assays indicated that inhibition of IGF-1 signaling elevated solute permeability through the monolayer of human aortic endothelial cells. In summary, endothelial IGF1R deficiency increases atherosclerosis, and IGF-1 positively regulates tight junction protein and adherens junction protein levels and endothelial barrier function. Our findings suggest that the elevation of the endothelial junction protein level is, at least in part, the mechanism for antiatherogenic effects of IGF-1.NEW & NOTEWORTHY Endothelial insulin-like growth factor-1 (IGF-1) receptor deficiency significantly elevated atherosclerotic burden in apolipoprotein E-deficient mice, mediated at least in part by downregulation of intercellular junction proteins and, thus, elevated endothelial permeability. This study revealed a novel role for IGF-1 in supporting endothelial barrier function. These findings suggest that IGF-1's ability to promote endothelial barrier function may offer a novel therapeutic strategy for vascular diseases such as atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Receptor IGF Tipo 1/deficiencia , Animales , Antígenos CD/metabolismo , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/patología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Placa Aterosclerótica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Células THP-1 , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
3.
J Cell Physiol ; 234(12): 22242-22259, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31074012

RESUMEN

Sustained inflammation and matrix metalloproteinase (MMP) activation contribute to vascular occlusive/proliferative disorders. Interleukin-17 (IL-17) is a proinflammatory cytokine that signals mainly via TRAF3 Interacting Protein 2 (TRAF3IP2), an upstream regulator of various critical transcription factors, including AP-1 and NF-κB. Reversion inducing cysteine rich protein with kazal motifs (RECK) is a membrane-anchored MMP inhibitor. Here we investigated whether IL-17A/TRAF3IP2 signaling promotes MMP-13-dependent human aortic smooth muscle cell (SMC) proliferation and migration, and determined whether RECK overexpression blunts these responses. Indeed, IL-17A treatment induced (a) JNK, p38 MAPK, AP-1, NF-κB, and CREB activation, (b) miR-21 induction, (c) miR-27b and miR-320 inhibition, (d) MMP-13 expression and activation, (e) RECK suppression, and (f) SMC migration and proliferation, all in a TRAF3IP2-dependent manner. In fact, gain of TRAG3IP2 function, by itself, induced MMP-13 expression and activation, and RECK suppression. Furthermore, treatment with recombinant MMP-13 stimulated SMC migration in part via ERK activation. Importantly, RECK gain-of-function attenuated MMP-13 activity without affecting its mRNA or protein levels, and inhibited IL-17A- and MMP-13-induced SMC migration. These results indicate that increased MMP-13 and decreased RECK contribute to IL-17A-induced TRAF3IP2-dependent SMC migration and proliferation, and suggest that TRAF3IP2 inhibitors or RECK inducers have the potential to block the progression of neointimal thickening in hyperplastic vascular diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aorta/citología , Movimiento Celular , Proteínas Ligadas a GPI/metabolismo , Interleucina-17/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proliferación Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Proteínas Recombinantes/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transducción de Señal , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
4.
Arterioscler Thromb Vasc Biol ; 38(10): 2306-2317, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354209

RESUMEN

Objective- IGF-1 (insulin-like growth factor 1) is a major autocrine/paracrine growth factor, which promotes cell proliferation, migration, and survival. We have shown previously that IGF-1 reduced atherosclerosis and promoted features of stable atherosclerotic plaque in Apoe-/- mice-an animal model of atherosclerosis. The aim of this study was to assess effects of smooth muscle cell (SMC) IGF-1 signaling on the atherosclerotic plaque. Approach and Results- We generated Apoe-/- mice with IGF1R (IGF-1 receptor) deficiency in SMC and fibroblasts (SM22α [smooth muscle protein 22 α]-CreKI/IGF1R-flox mice). IGF1R was decreased in the aorta and adventitia of SM22α-CreKI/IGF1R-flox mice and also in aortic SMC, embryonic, skin, and lung fibroblasts isolated from SM22α-CreKI/IGF1R-flox mice. IGF1R deficiency downregulated collagen mRNA-binding protein LARP6 (La ribonucleoprotein domain family, member 6) and vascular collagen, and mice exhibited growth retardation. The high-fat diet-fed SM22α-CreKI/IGF1R-flox mice had increased atherosclerotic burden and inflammatory responses. α-SMA (α-smooth muscle actin)-positive plaque cells had reduced proliferation and elevated apoptosis. SMC/fibroblast-targeted decline in IGF-1 signaling decreased atherosclerotic plaque SMC, markedly depleted collagen, reduced plaque fibrous cap, and increased plaque necrotic cores. Aortic SMC isolated from SM22α-CreKI/IGF1R-flox mice had decreased cell proliferation, migration, increased sensitivity to apoptosis, and these effects were associated with disruption of IGF-1-induced Akt signaling. Conclusions- IGF-1 signaling in SMC and in fibroblast is a critical determinant of normal vascular wall development and atheroprotection.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica , Regiones Promotoras Genéticas , Receptor IGF Tipo 1/deficiencia , Actinas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Autoantígenos/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibrosis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Ribonucleoproteínas/metabolismo , Transducción de Señal , Antígeno SS-B
5.
FASEB J ; 31(7): 3179-3192, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28404743

RESUMEN

Atherosclerotic plaque destabilization is the major determinant of most acute coronary events. Smooth muscle cell (SMC) death contributes to plaque destabilization. Here, we describe a novel antiapoptotic mechanism in vascular SMCs that involves interaction of nuclear glyceraldehyde-3-phosphate dehydrogenase (GAPDH) with apurinic/apyrimidinic endonuclease 1 (Ape1), the major oxidized DNA repair enzyme. GAPDH down-regulation potentiated H2O2-induced DNA damage and SMC apoptosis. Conversely, GAPDH overexpression decreased DNA damage and protected SMCs against apoptosis. Ape1 down-regulation reversed the resistance of GAPDH-overexpressing cells to DNA damage and apoptosis, which indicated that Ape1 is indispensable for GAPDH-dependent protective effects. GAPDH bound Ape1 in the SMC nucleus, and blocking (or oxidation) of GAPDH active site cysteines suppressed GAPDH/Ape1 interaction and potentiated apoptosis. GAPDH up-regulated Ape1 via a transcription factor homeobox protein Hox-A5-dependent mechanism. GAPDH levels were reduced in atherosclerotic plaque SMCs, and this effect correlated with oxidative stress and SMC apoptosis. Thus, we demonstrated that nuclear GAPDH/Ape1 interaction preserved Ape1 activity, reduced DNA damage, and prevented SMC apoptosis. Suppression of SMC apoptosis by maintenance of nuclear GAPDH/Ape1 interactions may be a novel therapy to increase atherosclerotic plaque stability.-Hou, X., Snarski, P., Higashi, Y., Yoshida, T., Jurkevich, A., Delafontaine, P., Sukhanov, S. Nuclear complex of glyceraldehyde-3-phosphate dehydrogenase and DNA repair enzyme apurinic/apyrimidinic endonuclease I protect smooth muscle cells against oxidant-induced cell death.


Asunto(s)
Muerte Celular/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Transporte Activo de Núcleo Celular , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Núcleo Celular/enzimología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Regulación Enzimológica de la Expresión Génica , Peróxido de Hidrógeno , Ratones , Ratones Noqueados , Ratas
6.
J Biol Chem ; 291(37): 19425-36, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27466370

RESUMEN

TRAF3IP2 (TRAF3 interacting protein 2; previously known as CIKS or Act1) is a key intermediate in the normal inflammatory response and the pathogenesis of various autoimmune and inflammatory diseases. Induction of TRAF3IP2 activates IκB kinase (IKK)/NF-κB, JNK/AP-1, and c/EBPß and stimulates the expression of various inflammatory mediators with negative myocardial inotropic effects. To investigate the role of TRAF3IP2 in heart disease, we generated a transgenic mouse model with cardiomyocyte-specific TRAF3IP2 overexpression (TRAF3IP2-Tg). Echocardiography, magnetic resonance imaging, and pressure-volume conductance catheterization revealed impaired cardiac function in 2-month-old male transgenic (Tg) mice as evidenced by decreased ejection fraction, stroke volume, cardiac output, and peak ejection rate. Moreover, the male Tg mice spontaneously developed myocardial hypertrophy (increased heart/body weight ratio, cardiomyocyte cross-sectional area, GATA4 induction, and fetal gene re-expression). Furthermore, TRAF3IP2 overexpression resulted in the activation of IKK/NF-κB, JNK/AP-1, c/EBPß, and p38 MAPK and induction of proinflammatory cytokines, chemokines, and extracellular matrix proteins in the heart. Although myocardial hypertrophy decreased with age, cardiac fibrosis (increased number of myofibroblasts and enhanced expression and deposition of fibrillar collagens) increased progressively. Despite these adverse changes, TRAF3IP2 overexpression did not result in cell death at any time period. Interestingly, despite increased mRNA expression, TRAF3IP2 protein levels and activation of its downstream signaling intermediates remained unchanged in the hearts of female Tg mice. The female Tg mice also failed to develop myocardial hypertrophy. In summary, these results demonstrate that overexpression of TRAF3IP2 in male mice is sufficient to induce myocardial hypertrophy, cardiac fibrosis, and contractile dysfunction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Sistema de Señalización de MAP Quinasas , Miocardio/metabolismo , Volumen Sistólico , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Quimiocinas/genética , Quimiocinas/metabolismo , Colágeno/biosíntesis , Colágeno/genética , Modelos Animales de Enfermedad , Femenino , Fibrosis , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocardio/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Caracteres Sexuales , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Circulation ; 133(23): 2263-78, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27154724

RESUMEN

BACKGROUND: We have previously shown that systemic infusion of insulin-like growth factor-1 (IGF-1) exerts anti-inflammatory and antioxidant effects and reduces atherosclerotic burden in apolipoprotein E (Apoe)-deficient mice. Monocytes/macrophages express high levels of IGF-1 receptor (IGF1R) and play a pivotal role in atherogenesis, but the potential effects of IGF-1 on their function are unknown. METHODS AND RESULTS: To determine mechanisms whereby IGF-1 reduces atherosclerosis and to explore the potential involvement of monocytes/macrophages, we created monocyte/macrophage-specific IGF1R knockout (MΦ-IGF1R-KO) mice on an Apoe(-/-) background. We assessed atherosclerotic burden, plaque features of stability, and monocyte recruitment to atherosclerotic lesions. Phenotypic changes of IGF1R-deficient macrophages were investigated in culture. MΦ-IGF1R-KO significantly increased atherosclerotic lesion formation, as assessed by Oil Red O staining of en face aortas and aortic root cross-sections, and changed plaque composition to a less stable phenotype, characterized by increased macrophage and decreased α-smooth muscle actin-positive cell population, fibrous cap thinning, and decreased collagen content. Brachiocephalic artery lesions of MΦ-IGF1R-KO mice had histological features implying plaque vulnerability. Macrophages isolated from MΦ-IGF1R-KO mice showed enhanced proinflammatory responses on stimulation by interferon-γ and oxidized low-density lipoprotein and elevated antioxidant gene expression levels. Moreover, IGF1R-deficient macrophages had decreased expression of ABCA1 and ABCG1 and reduced lipid efflux. CONCLUSIONS: Our data indicate that macrophage IGF1R signaling suppresses macrophage and foam cell accumulation in lesions and reduces plaque vulnerability, providing a novel mechanism whereby IGF-1 exerts antiatherogenic effects.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica , Receptor IGF Tipo 1/deficiencia , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Plasticidad de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Interferón gamma/farmacología , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Noqueados , Fenotipo , Receptor IGF Tipo 1/genética , Rotura Espontánea
8.
J Biol Chem ; 289(11): 7264-74, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24469459

RESUMEN

Collagen content in atherosclerotic plaque is a hallmark of plaque stability. Our earlier studies showed that insulin-like growth factor-1 (IGF-1) increases collagen content in atherosclerotic plaques of Apoe(-/-) mice. To identify mechanisms we investigated the effect of IGF-1 on the la ribonucleoprotein domain family member 6 (LARP6). LARP6 binds a stem-loop motif in the 5'-UTR of the mRNAs encoding the collagen type I α-subunits (α1(I) and α2(I)), and coordinates their translation into the heterotrimeric collagen type I molecule. In human aortic smooth muscle cells (SMCs), IGF-1 rapidly increased LARP6 expression and the rate of collagen synthesis and extracellular accumulation. IGF-1 increased both LARP6 and collagen type I expression via a post-transcriptional and translation-dependent mechanism involving PI3K/Akt/p70S6k-signaling. Immunoprecipitation of LARP6, followed by qPCR indicated that IGF-1 increased the level of COL1a1 and COL1a2 mRNA bound to LARP6. Mutation of the 5' stem-loop of Col1a1 mRNA, which inhibits binding of LARP6, abolished the ability of IGF-1 to increase synthesis of collagen type I. Furthermore, overexpression of a 5' stem-loop RNA molecular decoy that sequesters LARP6, prevented the ability of IGF-1 to increase pro-α1(I) and mature α1(I) expression in cultured medium. IGF-1 infusion in Apoe(-/-) mice increased expression of LARP6 and pro-α1(I) in aortic lysates, and SMC-specific IGF-1-overexpression robustly increased collagen fibrillogenesis in atherosclerotic plaque. In conclusion, we identify LARP6 as a critical mediator by which IGF-1 augments synthesis of collagen type I in vascular smooth muscle, which may play an important role in promoting atherosclerotic plaque stability.


Asunto(s)
Autoantígenos/metabolismo , Colágeno Tipo I/metabolismo , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Aorta/citología , Aterosclerosis/metabolismo , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/citología , Mutación , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo , Placa Aterosclerótica/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Antígeno SS-B
9.
J Biol Chem ; 288(33): 23823-32, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23831688

RESUMEN

Cachexia is a serious complication of many chronic diseases, such as congestive heart failure (CHF) and chronic kidney disease (CKD). Although patients with advanced CHF or CKD often have increased angiotensin II (Ang II) levels and cachexia and Ang II causes skeletal muscle wasting in rodents, the potential effects of Ang II on muscle regeneration are unknown. Muscle regeneration is highly dependent on the ability of a pool of muscle stem cells (satellite cells) to proliferate and to repair damaged myofibers or form new myofibers. Here we show that Ang II reduced skeletal muscle regeneration via inhibition of satellite cell (SC) proliferation. Ang II reduced the number of regenerating myofibers and decreased expression of SC proliferation/differentiation markers (MyoD, myogenin, and active-Notch) after cardiotoxin-induced muscle injury in vivo and in SCs cultured in vitro. Ang II depleted the basal pool of SCs, as detected in Myf5(nLacZ/+) mice and by FACS sorting, and this effect was inhibited by Ang II AT1 receptor (AT1R) blockade and in AT1aR-null mice. AT1R was highly expressed in SCs, and Notch activation abrogated the AT1R-mediated antiproliferative effect of Ang II in cultured SCs. In mice that developed CHF postmyocardial infarction, there was skeletal muscle wasting and reduced SC numbers that were inhibited by AT1R blockade. Ang II inhibition of skeletal muscle regeneration via AT1 receptor-dependent suppression of SC Notch and MyoD signaling and proliferation is likely to play an important role in mechanisms leading to cachexia in chronic disease states such as CHF and CKD.


Asunto(s)
Angiotensina II/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/patología , Angiotensina II/administración & dosificación , Animales , Recuento de Células , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ratones , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Notch/metabolismo , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal/efectos de los fármacos , Síndrome Debilitante/complicaciones , Síndrome Debilitante/metabolismo , Síndrome Debilitante/patología , Síndrome Debilitante/fisiopatología
10.
JCI Insight ; 8(4)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36602878

RESUMEN

Although murine models of coronary atherosclerotic disease have been used extensively to determine mechanisms, limited new therapeutic options have emerged. Pigs with familial hypercholesterolemia (FH pigs) develop complex coronary atheromas that are almost identical to human lesions. We reported previously that insulin-like growth factor 1 (IGF-1) reduced aortic atherosclerosis and promoted features of stable plaque in a murine model. We administered human recombinant IGF-1 or saline (control) in atherosclerotic FH pigs for 6 months. IGF-1 decreased relative coronary atheroma in vivo (intravascular ultrasound) and reduced lesion cross-sectional area (postmortem histology). IGF-1 increased plaque's fibrous cap thickness, and reduced necrotic core, macrophage content, and cell apoptosis, consistent with promotion of a stable plaque phenotype. IGF-1 reduced circulating triglycerides, markers of systemic oxidative stress, and CXCL12 chemokine levels. We used spatial transcriptomics (ST) to identify global transcriptome changes in advanced plaque compartments and to obtain mechanistic insights into IGF-1 effects. ST analysis showed that IGF-1 suppressed FOS/FOSB factors and gene expression of MMP9 and CXCL14 in plaque macrophages, suggesting possible involvement of these molecules in IGF-1's effect on atherosclerosis. Thus, IGF-1 reduced coronary plaque burden and promoted features of stable plaque in a pig model, providing support for consideration of clinical trials.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Hiperlipoproteinemia Tipo II , Placa Aterosclerótica , Ratones , Humanos , Animales , Porcinos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Aterosclerosis/patología , Placa Aterosclerótica/patología
11.
Am J Physiol Heart Circ Physiol ; 300(5): H1898-906, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21335474

RESUMEN

Some clinical studies have suggested that lower IGF-I levels may be associated with an increased risk of ischemic heart disease. We generated atherosclerosis-prone apolipoprotein E-deficient (ApoE(-/-)) mice with 6T alleles (6T/ApoE(-/-) mice) with a 20% decline in circulating IGF-I and fed these mice and control ApoE(-/-) mice with normal chow or a Western diet for 12 wk to evaluate the effect of low serum IGF-I on atherosclerosis progression. We found that the 6T/ApoE(-/-) phenotype was characterized by an increased atherosclerotic burden, elevated plaque macrophages, and increased proinflammatory cytokine TNF-α levels compared with ApoE(-/-) controls. 6T/ApoE(-/-) mice had similar body weight, blood pressure, serum total cholesterol levels, total plaque and smooth muscle cell apoptosis rates, and circulating levels of endothelial progenitor cells as ApoE(-/-) mice. 6T/ApoE(-/-) mice fed with normal chow had reduced vascular endothelial nitric oxide synthase mRNA levels and a trend to increased aortic expression of chemokine (C-C motif) receptor (CCR)1, CCR2, and monocyte chemoattractant protein-1/chemokine (C-C motif) ligand 2. Western diet-fed 6T/ApoE(-/-) mice had a trend to increased expression of macrophage scavenger receptor-1/scavenger receptor-A, osteopontin, ATP-binding cassette (subfamily A, member 1), and angiotensin-converting enzyme and elevated circulating levels of the neutrophil chemoattractant chemokine (C-X-C motif) ligand 1 (KC). Our data establish a link between lower circulating IGF-I and increased atherosclerosis that has important clinical implications.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/epidemiología , Aterosclerosis/fisiopatología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Presión Sanguínea/fisiología , Peso Corporal/fisiología , Colesterol/sangre , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Factores de Riesgo , Factor de Necrosis Tumoral alfa/sangre
12.
Biochem Biophys Res Commun ; 409(2): 217-21, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21570954

RESUMEN

Angiotensin II (Ang II) causes skeletal muscle wasting via an increase in muscle catabolism. To determine whether the wasting effects of Ang II were related to its ability to increase NADPH oxidase-derived reactive oxygen species (ROS) we infused wild-type C57BL/6J or p47(phox)(-/-) mice with vehicle or Ang II for 7days. Superoxide production was increased 2.4-fold in the skeletal muscle of Ang II infused mice, and this increase was prevented in p47(phox)(-/-) mice. Apocynin treatment prevented Ang II-induced superoxide production in skeletal muscle, consistent with Ang II increasing NADPH oxidase derived ROS. Ang II induced loss of body and skeletal muscle weight in C57BL/6J mice, whereas the reduction was significantly attenuated in p47(phox)(-/-) animals. The reduction of skeletal muscle weight caused by Ang II was associated with an increase of proteasome activity, and this increase was completely prevented in the skeletal muscle of p47(phox)(-/-) mice. In conclusion, Ang II-induced skeletal muscle wasting is in part dependent on NADPH oxidase derived ROS.


Asunto(s)
Angiotensina II/efectos adversos , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo , Síndrome Debilitante/inducido químicamente , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , NADPH Oxidasas/genética , Oxidación-Reducción , Complejo de la Endopetidasa Proteasomal/metabolismo , Superóxidos/metabolismo , Síndrome Debilitante/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 30(10): 1916-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20671230

RESUMEN

OBJECTIVE: Growth factors may play a permissive role in atherosclerosis initiation and progression, in part via their promotion of vascular smooth muscle cell (VSMC) accumulation in plaques. However, unstable human plaques often have a relative paucity of VSMC, which has been suggested to contribute to plaque rupture and erosion and to clinical events. Insulin-like growth factor-1 (IGF-1) is an endocrine and autocrine/paracrine growth factor that is a mitogen for VSMC, but when infused into Apoe(-/-) mice it paradoxically reduces atherosclerosis burden. METHODS AND RESULTS: To determine the effect of stimulation of VSMC growth on atherosclerotic plaque development and to understand mechanisms of IGF-1's atheroprotective effect, we assessed atherosclerotic plaques in mice overexpressing IGF-1 in smooth muscle cells (SMC) under the control of the α-smooth muscle actin promoter, after backcrossing to the Apoe(-/-) background (SMP8/Apoe(-/-)). Compared with Apoe(-/-) mice, these SMP8/Apoe(-/-) mice developed a comparable plaque burden after 12 weeks on a Western diet, suggesting that the ability of increased circulating IGF-1 to reduce plaque burden was mediated in large part via non-SMC target cells. However, advanced plaques in SMP8/Apoe(-/-) mice displayed several features of plaque stability, including increased fibrous cap area, α-smooth muscle actin-positive SMC and collagen content, and reduced necrotic cores. CONCLUSIONS: These findings indicate that stimulation of VSMC IGF-1 signaling does not alter total atherosclerotic plaque burden and may improve atherosclerotic plaque stability.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Factor I del Crecimiento Similar a la Insulina/fisiología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Animales , Aorta/patología , Aorta/fisiopatología , Apolipoproteínas E/genética , Aterosclerosis/genética , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Hipertrofia , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Cell Signal ; 77: 109825, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33160017

RESUMEN

Chronic inflammation and persistent oxidative stress contribute to the development and progression of vascular proliferative diseases. We hypothesized that the proinflammatory cytokine interleukin (IL)-17A induces oxidative stress and amplifies inflammatory signaling in human aortic smooth muscle cells (SMC) via TRAF3IP2-mediated NLRP3/caspase-1-dependent mitogenic and migratory proinflammatory cytokines IL-1ß and IL-18. Further, we hypothesized that these maladaptive changes are prevented by empagliflozin (EMPA), an SGLT2 (Sodium/Glucose Cotransporter 2) inhibitor. Supporting our hypotheses, exposure of cultured SMC to IL-17A promoted proliferation and migration via TRAF3IP2, TRAF3IP2-dependent superoxide and hydrogen peroxide production, NLRP3 expression, caspase-1 activation, and IL-1ß and IL-18 secretion. Furthermore, NLRP3 knockdown, caspase-1 inhibition, and pretreatment with IL-1ß and IL-18 neutralizing antibodies and IL-18BP, each attenuated IL-17A-induced SMC migration and proliferation. Importantly, SMC express SGLT2, and pre-treatment with EMPA attenuated IL-17A/TRAF3IP2-dependent oxidative stress, NLRP3 expression, caspase-1 activation, IL-1ß and IL-18 secretion, and SMC proliferation and migration. Importantly, silencing SGLT2 attenuated EMPA-mediated inhibition of IL-17A-induced cytokine secretion and SMC proliferation and migration. EMPA exerted these beneficial antioxidant, anti-inflammatory, anti-mitogenic and anti-migratory effects under normal glucose conditions and without inducing cell death. These results suggest the therapeutic potential of EMPA in vascular proliferative diseases.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Caspasa 1/metabolismo , Proliferación Celular/efectos de los fármacos , Glucósidos/farmacología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , ARN/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Movimiento Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-17/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN/antagonistas & inhibidores , ARN/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Am J Physiol Heart Circ Physiol ; 298(5): H1565-70, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20228261

RESUMEN

Congestive heart failure is associated with activation of the renin-angiotensin system and skeletal muscle wasting. Angiotensin II (ANG II) has been shown to increase muscle proteolysis and decrease circulating and skeletal muscle IGF-1. We have shown previously that skeletal muscle-specific overexpression of IGF-1 prevents proteolysis and apoptosis induced by ANG II. These findings indicated that downregulation of IGF-1 signaling in skeletal muscle played an important role in the wasting effect of ANG II. However, the signaling pathways and mechanisms whereby IGF-1 prevents ANG II-induced skeletal muscle atrophy are unknown. Here we show ANG II-induced transcriptional regulation of two ubiquitin ligases atrogin-1 and muscle ring finger-1 (MuRF-1) that precedes the reduction of skeletal muscle IGF-1 expression, suggesting that activation of atrogin-1 and MuRF-1 is an initial mechanism leading to skeletal muscle atrophy in response to ANG II. IGF-1 overexpression in skeletal muscle prevented ANG II-induced skeletal muscle wasting and the expression of atrogin-1, but not MuRF-1. Dominant-negative Akt and constitutively active Foxo-1 blocked the ability of IGF-1 to prevent ANG II-mediated upregulation of atrogin-1 and skeletal muscle wasting. Our findings demonstrate that the ability of IGF-1 to prevent ANG II-induced skeletal muscle wasting is mediated via an Akt- and Foxo-1-dependent signaling pathway that results in inhibition of atrogin-1 but not MuRF-1 expression. These data suggest strongly that atrogin-1 plays a critical role in mechanisms of ANG II-induced wasting in vivo.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Angiotensina II/toxicidad , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas Musculares/biosíntesis , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Ligasas SKP Cullina F-box/biosíntesis , Animales , Atrofia , Electroporación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba/efectos de los fármacos
16.
J Aging Res ; 2020: 4939310, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32802505

RESUMEN

Mitochondrial dysfunction is a hallmark of cellular aging. Mitophagy is a critical mitochondrial quality control mechanism that removes dysfunctional mitochondria and contributes to cell survival. Insulin-like growth factor 1 (IGF-1) promotes survival of smooth muscle cells (SMCs), but its potential effect on cellular aging is unknown yet. We found that IGF-1 decreased cell senescence, prevented DNA telomere shortening, increased mitochondrial membrane potential, activated cytochrome C oxidase, and reduced mitochondrial DNA damage in long-term cultured (aged) aortic SMC, suggesting an antiaging effect. IGF-1 increased mitophagy in aged cells, and this was associated with decreased expression of cyclin-dependent kinase inhibitors p16 and p21 and elevated levels of Nrf2 and Sirt3, regulators of mitophagy and mitochondrial biogenesis. SiRNA-induced inhibition of either Nrf2 or Sirt3 blocked IGF-1-induced upregulation of mitophagy, suggesting that the Nrf2/Sirt3 pathway was required for IGF-1's effect on mitophagy. PINK1 is a master regulator of mitophagy. PINK1 silencing suppressed mitophagy and inhibited IGF-1-induced antiaging effects in aged SMC, consistent with an essential role of mitophagy in IGF-1's effect on cellular aging. Thus, IGF-1 inhibited cellular aging via Nrf2/Sirt3-dependent activation of mitophagy. Our data suggest that activation of IGF-1 signaling is a novel potential strategy to activate mitophagy and slow cellular aging.

17.
Am J Transl Res ; 12(10): 6559-6568, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194052

RESUMEN

BACKGROUND: Macrophage activation and massive foam cell formation are key events in the development of Atherosclerosis (AS). Apurinic apyrimidinic endonuclease 1/Redox factor-1 (APE1) is an enzyme responsible for DNA repair and redox regulation. Recent studies indicate that APE1 is also involved in inflammatory response. We sought to explore its effect on oxidized low-density lipoprotein (oxLDL) induced macrophage activation and foam cell formation. METHODS: Human macrophage cell line THP-1 cells were cultured and treated with oxLDL. The mRNA and protein levels of inflammatory markers for macrophage activation were measured. Foam cell formation was detected by Oil red O staining. Meanwhile the major cellular receptors responsible for oxLDL uptake and efflux were detected. Chromatin immunoprecipitation-quantitative real time PCR (ChIP-qPCR) and dual luciferase reporter assays were performed to identify the molecular mechanisms through which APE1 affects macrophage activation and foam cell formation. RESULTS: Aberrant APE1 expression dramatically decreases the mRNA and protein of oxLDL-induced inflammatory molecules in THP-1 cells, accompanied by significantly inhibited foam cell formation. Western blot assay showed that down-regulation of LOX1, a receptor of oxLDL, is responsible for the inhibitory effect of APE1 on oxLDL induced macrophage inflammation. ChIP-qPCR assay showed that APE1 inhibits binding of the LOX1 promoter to its transcription factor Oct1, leading to suppression of LOX1. CONCLUSION: Our data confirm the anti-inflammatory properties of APE1 and for the first-time report that APE1 suppresses foam cell formation from macrophages via the oxLDL receptor LOX1. This finding indicates that APE1 can be a therapeutic target for AS prevention.

18.
Mol Cell Biochem ; 330(1-2): 1-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19360379

RESUMEN

Diabetes and impaired glucose tolerance are associated with increased cardiovascular disease morbidity and mortality particularly after vascular injury. Since insulin is frequently used in such patients, the effect of glulisine (short acting) and glargine (long acting) were tested in Zucker fatty rat carotid artery subjected to balloon catheter injury. Insulin-resistant Zucker fatty rats were sc injected 0.45 mg/kg/d of glargine (once) or glulisine (twice) for 1 week before, and 3 weeks after balloon injury. Fasting and postprandial glucose was measured twice weekly. Injured and uninjured carotid arteries, liver, and aorta were harvested after 3 weeks of injury. Carotid sections were H&E stained for measuring intima/media ratio or immunostained for nitrotyrosine. Serum and aortic protein were analyzed for IGF-1 and 8-isoprostane, respectively. Carotid intima/media ratio was significantly reduced in the glargine group [0.9 +/- 0.1-control; 0.6 +/- 0.1-glulisine; 0.4 +/- 0.1-glargine, P < 0.05]. Serum IGF-1 levels were higher in both insulins, but significant only in glargine group [567 +/- 121 (ng/ml)-control; 1059 +/- 150 (ng/ml)-glargine; P < 0.05]. The aortic 8-isoprostane levels decreased significantly in the glargine group [(921 vs. 2566 pg/mg protein; P < 0.05]. Compared to control nitrotyrosine staining intensity was significantly lower in both groups of insulin-treated rats; the lowest level was in the glargine group. Insulin glargine attenuates carotid intimal hyperplasia in nondiabetic Zucker fatty rat independent of glucose levels and support a valuable function for insulin in vascular disease that merits additional investigations.


Asunto(s)
Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Cateterismo/efectos adversos , Hiperplasia/tratamiento farmacológico , Insulina/análogos & derivados , Estrés Oxidativo , Túnica Íntima/patología , Animales , Glucemia/análisis , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Hiperplasia/etiología , Hipoglucemiantes , Insulina/farmacología , Insulina/uso terapéutico , Insulina Glargina , Resistencia a la Insulina , Insulina de Acción Prolongada , Ratas , Ratas Zucker , Resultado del Tratamiento
19.
Growth Horm IGF Res ; 45: 6-16, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30735831

RESUMEN

Atherosclerosis is an inflammatory arterial pathogenic condition, which leads to ischemic cardiovascular diseases, such as coronary artery disease and myocardial infarction, stroke, and peripheral arterial disease. Atherosclerosis is a multifactorial disorder and its pathophysiology is highly complex. Changes in expression of multiple genes coupled with environmental and lifestyle factors initiate cascades of adverse events involving multiple types of cells (e.g. vascular endothelial cells, smooth muscle cells, and macrophages). IGF-1 is a pleiotropic factor, which is found in the circulation (endocrine IGF-1) and is also produced locally in arteries (endothelial cells and smooth muscle cells). IGF-1 exerts a variety of effects on these cell types in the context of the pathogenesis of atherosclerosis. In fact, there is an increasing body of evidence suggesting that IGF-1 has beneficial effects on the biology of atherosclerosis. This review will discuss recent findings relating to clinical investigations on the relation between IGF-1 and cardiovascular disease and basic research using animal models of atherosclerosis that have elucidated some of the mechanisms underlying atheroprotective effects of IGF-1.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Humanos
20.
Cell Signal ; 57: 10-20, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30716386

RESUMEN

Minocycline, a tetracycline antibiotic, is known to exert vasculoprotective effects independent of its anti-bacterial properties; however the underlying molecular mechanisms are not completely understood. Reversion Inducing Cysteine Rich Protein with Kazal Motifs (RECK) is a cell surface expressed, membrane anchored protein, and its overexpression inhibits cancer cell migration. We hypothesized that minocycline inhibits platelet-derived growth factor (PDGF)-induced human aortic smooth muscle cell (SMC) proliferation and migration via RECK upregulation. Our data show that the BB homodimer of recombinant PDGF (PDGF-BB) induced SMC migration and proliferation, effects significantly blunted by pre-treatment with minocycline. Further investigations revealed that PDGF-BB induced PI3K-dependent AKT activation, ERK activation, reactive oxygen species generation, Nuclear Factor-κB and Activator Protein-1 activation, microRNA (miR)-221 and miR-222 induction, RECK suppression, and matrix metalloproteinase (MMP2 and 9) activation, effects that were reversed by minocycline. Notably, minocycline induced RECK expression dose-dependently within the therapeutic dose of 1-100 µM, and silencing RECK partially reversed the inhibitory effects of minocycline on PDGF-BB-induced MMP activation, and SMC proliferation and migration. Further, targeting MMP2 and MMP9 blunted PDGF-BB-induced SMC migration. Together, these results demonstrate that minocycline inhibits PDGF-BB-induced SMC proliferation and migration by restoring RECK, an MMP inhibitor. These results indicate that the induction of RECK is one of the mechanisms by which minocycline exerts vasculoprotective effects.


Asunto(s)
Proteínas Ligadas a GPI/efectos de los fármacos , MicroARNs/genética , Minociclina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Ligadas a GPI/genética , Humanos , MicroARNs/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA