Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Med ; 30(1): 21, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38317079

RESUMEN

BACKGROUND: Pericytes are a vital component of the blood-brain barrier, and their involvement in acute inflammation was recently suggested. However, it remains unclear whether pericytes contribute to hypothalamic chronic inflammation and energy metabolism in obesity. The present study investigated the impact of pericytes on the pathophysiology of obesity by focusing on platelet-derived growth factor (PDGF) signaling, which regulates pericyte functions. METHODS: Tamoxifen-inducible systemic conditional PDGF receptor ß knockout mice (Pdgfrb∆SYS-KO) and Calcium/calmodulin-dependent protein kinase type IIa (CaMKIIa)-positive neuron-specific PDGF receptor ß knockout mice (Pdgfrb∆CaMKII-KO) were fed a high-fat diet, and metabolic phenotypes before and 3 to 4 weeks after dietary loading were examined. Intracellular energy metabolism and relevant signal transduction in lipopolysaccharide- and/or platelet-derived growth factor-BB (PDGF-BB)-stimulated human brain pericytes (HBPCs) were assessed by the Seahorse XFe24 Analyzer and Western blotting. The pericyte secretome in conditioned medium from HBPCs was studied using cytokine array kit, and its impact on polarization was examined in bone marrow-derived macrophages (BMDMs), which are microglia-like cells. RESULTS: Energy consumption increased and body weight gain decreased after high-fat diet loading in Pdgfrb∆SYS-KO mice. Cellular oncogene fos (cFos) expression increased in proopiomelanocortin (POMC) neurons, whereas microglial numbers and inflammatory gene expression decreased in the hypothalamus of Pdgfrb∆SYS-KO mice. No significant changes were observed in Pdgfrb∆CaMKII-KO mice. In HBPCs, a co-stimulation with lipopolysaccharide and PDGF-BB shifted intracellular metabolism towards glycolysis, activated mitogen-activated protein kinase (MAPK), and modulated the secretome to the inflammatory phenotype. Consequently, the secretome showed an increase in various proinflammatory chemokines and growth factors including Epithelial-derived neutrophil-activating peptide 78 (C-X-C motif chemokine ligand (CXCL)5), Thymus and activation-regulated chemokine (C-C motif chemokine (CCL)17), Monocyte chemoattractant protein 1 (CCL2), and Growth-regulated oncogene α (CXCL1). Furthermore, conditioned medium from HBPCs stimulated the inflammatory priming of BMDMs, and this change was abolished by the C-X-C motif chemokine receptor (CXCR) inhibitor. Consistently, mRNA expression of CXCL5 was elevated by lipopolysaccharide and PDGF-BB treatment in HBPCs, and the expression was significantly lower in the hypothalamus of Pdgfrb∆SYS-KO mice than in control Pdgfrbflox/flox mice (FL) following 4 weeks of HFD feeding. CONCLUSIONS: PDGF receptor ß signaling in hypothalamic pericytes promotes polarization of macrophages by changing their secretome and contributes to the progression of obesity.


Asunto(s)
Pericitos , Factor de Crecimiento Derivado de Plaquetas , Ratones , Humanos , Animales , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Pericitos/metabolismo , Becaplermina/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Medios de Cultivo Condicionados/metabolismo , Lipopolisacáridos , Transducción de Señal , Inflamación/metabolismo , Ratones Noqueados , Obesidad/metabolismo , Hipotálamo , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo
2.
J Cachexia Sarcopenia Muscle ; 11(1): 241-258, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32003547

RESUMEN

BACKGROUND: Skeletal muscle is mainly responsible for insulin-stimulated glucose disposal. Dysfunction in skeletal muscle metabolism especially during obesity contributes to the insulin resistance. Astaxanthin (AX), a natural antioxidant, has been shown to ameliorate hepatic insulin resistance in obese mice. However, its effects in skeletal muscle are poorly understood. The current study aimed to investigate the molecular target of AX in ameliorating skeletal muscle insulin resistance. METHODS: We fed 6-week-old male C57BL/6J mice with normal chow (NC) or NC supplemented with AX (NC+AX) and high-fat-diet (HFD) or HFD supplemented with AX for 24 weeks. We determined the effect of AX on various parameters including insulin sensitivity, glucose uptake, inflammation, kinase signaling, gene expression, and mitochondrial function in muscle. We also determined energy metabolism in intact C2C12 cells treated with AX using the Seahorse XFe96 Extracellular Flux Analyzer and assessed the effect of AX on mitochondrial oxidative phosphorylation and mitochondrial biogenesis. RESULTS: AX-treated HFD mice showed improved metabolic status with significant reduction in blood glucose, serum total triglycerides, and cholesterol (p< 0.05). AX-treated HFD mice also showed improved glucose metabolism by enhancing glucose incorporation into peripheral target tissues, such as the skeletal muscle, rather than by suppressing gluconeogenesis in the liver as shown by hyperinsulinemic-euglycemic clamp study. AX activated AMPK in the skeletal muscle of the HFD mice and upregulated the expressions of transcriptional factors and coactivator, thereby inducing mitochondrial remodeling, including increased mitochondrial oxidative phosphorylation component and free fatty acid metabolism. We also assessed the effects of AX on mitochondrial biogenesis in the siRNA-mediated AMPK-depleted C2C12 cells and showed that the effect of AX was lost in the genetically AMPK-depleted C2C12 cells. Collectively, AX treatment (i) significantly ameliorated insulin resistance and glucose intolerance through regulation of AMPK activation in the muscle, (ii) stimulated mitochondrial biogenesis in the muscle, (iii) enhanced exercise tolerance and exercise-induced fatty acid metabolism, and (iv) exerted antiinflammatory effects via its antioxidant activity in adipose tissue. CONCLUSIONS: We concluded that AX treatment stimulated mitochondrial biogenesis and significantly ameliorated insulin resistance through activation of AMPK pathway in the skeletal muscle.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fibrinolíticos/uso terapéutico , Resistencia a la Insulina/fisiología , Mitocondrias Musculares/metabolismo , Animales , Fibrinolíticos/farmacología , Humanos , Masculino , Ratones , Biogénesis de Organelos , Xantófilas/farmacología , Xantófilas/uso terapéutico
3.
Pharmacol Ther ; 186: 25-44, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29289556

RESUMEN

Sleep, a mysterious behavior, has recently been recognized as a crucial factor for health and longevity. The daily sleep/wake cycle provides the basis of biorhythms controlling whole-body homeostasis and homeodynamics; therefore, disruption of sleep causes several physical and psychological disorders, including cardiovascular disease, obesity, diabetes, cancer, anxiety, depression, and cognitive dysfunction. However, the mechanism linking sleep disturbances and sleep-related disorders remains unknown. Orexin (also known as hypocretin) is a neuropeptide produced in the hypothalamus. Central levels of orexin oscillate with the daily rhythm and peak at the awake phase. Orexin plays a major role in stabilizing the wakefulness state. Orexin deficiency causes sleep/wake-state instability, resulting in narcolepsy. Hyper-activation of the orexin system also causes sleep disturbances, such as insomnia, and hence, suvorexant, an orexin receptor antagonist, has been clinically used to treat insomnia. Importantly, central actions of orexin regulate motivated behaviors, stress response, and energy/glucose metabolism by coordinating the central-autonomic nervous systems and endocrine systems. These multiple actions of orexin maintain survival. However, it remains unknown whether chronopharmacological interventions targeting the orexin system ameliorate sleep-related disorders as well as sleep in humans. To understand the significance of adequate orexin action for prevention of these disorders, this review summarizes the physiological functions of daily orexin action and pathological implications of its mistimed or reduced action in sleep disturbances and sleep-related disorders (lifestyle-related physical and neurological disorders in particular). Timed administration of drugs targeting the orexin system may prevent lifestyle-related diseases by improving the quality of life in patients with sleep disturbances.


Asunto(s)
Azepinas/uso terapéutico , Ritmo Circadiano/efectos de los fármacos , Estilo de Vida , Antagonistas de los Receptores de Orexina/uso terapéutico , Orexinas/metabolismo , Trastornos del Sueño-Vigilia , Triazoles/uso terapéutico , Azepinas/administración & dosificación , Humanos , Antagonistas de los Receptores de Orexina/administración & dosificación , Receptores de Orexina/metabolismo , Calidad de Vida/psicología , Fármacos Inductores del Sueño/administración & dosificación , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/metabolismo , Triazoles/administración & dosificación
4.
J Endocrinol ; 235(3): 179-191, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28855315

RESUMEN

Obesity-associated activation of the renin-angiotensin-aldosterone system is implicated in the pathogenesis of insulin resistance; however, influences of mineralocorticoid receptor (MR) inhibition remain unclear. Therefore, we aimed to clarify the anti-inflammatory mechanisms of MR inhibition using eplerenone, a selective MR antagonist, in C57BL/6 mice fed a high-fat diet (HFD) for 12 weeks. Eplerenone prevented excessive body weight gain and fat accumulation, ameliorated glucose intolerance and insulin resistance and enhanced energy metabolism. In the epididymal white adipose tissue (eWAT), eplerenone prevented obesity-induced accumulation of F4/80+CD11c+CD206--M1-adipose tissue macrophage (ATM) and reduction of F4/80+CD11c-CD206+-M2-ATM. Interestingly, M1-macrophage exhibited lower expression levels of MR, compared with M2-macrophage, in the ATM of eWAT and in vitro-polarized bone marrow-derived macrophages (BMDM). Importantly, eplerenone and MR knockdown attenuated the increase in the expression levels of proIl1b, Il6 and Tnfa, in the eWAT and liver of HFD-fed mice and LPS-stimulated BMDM. Moreover, eplerenone suppressed IL1b secretion from eWAT of HFD-fed mice. To reveal the anti-inflammatory mechanism, we investigated the involvement of NLRP3-inflammasome activation, a key process of IL1b overproduction. Eplerenone suppressed the expression of the inflammasome components, Nlrp3 and Caspase1, in the eWAT and liver. Concerning the second triggering factors, ROS production and ATP- and nigericin-induced IL1b secretion were suppressed by eplerenone in the LPS-primed BMDM. These results indicate that eplerenone inhibited both the priming and triggering signals that promote NLRP3-inflammasome activation. Therefore, we consider MR to be a crucial target to prevent metabolic disorders by suppressing inflammasome-mediated chronic inflammation in the adipose tissue and liver under obese conditions.


Asunto(s)
Intolerancia a la Glucosa/prevención & control , Inflamación/prevención & control , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Obesidad/complicaciones , Espironolactona/análogos & derivados , Tejido Adiposo Blanco/patología , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Eplerenona , Intolerancia a la Glucosa/etiología , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/etiología , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Antagonistas de Receptores de Mineralocorticoides/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/patología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Mineralocorticoides/metabolismo , Espironolactona/farmacología , Espironolactona/uso terapéutico
5.
Endocrinology ; 157(1): 195-206, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26492471

RESUMEN

Nicotine is known to affect the metabolism of glucose; however, the underlying mechanism remains unclear. Therefore, we here investigated whether nicotine promoted the central regulation of glucose metabolism, which is closely linked to the circadian system. The oral intake of nicotine in drinking water, which mainly occurred during the nighttime active period, enhanced daily hypothalamic prepro-orexin gene expression and reduced hyperglycemia in type 2 diabetic db/db mice without affecting body weight, body fat content, and serum levels of insulin. Nicotine administered at the active period appears to be responsible for the effect on blood glucose, because nighttime but not daytime injections of nicotine lowered blood glucose levels in db/db mice. The chronic oral treatment with nicotine suppressed the mRNA levels of glucose-6-phosphatase, the rate-limiting enzyme of gluconeogenesis, in the liver of db/db and wild-type control mice. In the pyruvate tolerance test to evaluate hepatic gluconeogenic activity, the oral nicotine treatment moderately suppressed glucose elevations in normal mice and mice lacking dopamine receptors, whereas this effect was abolished in orexin-deficient mice and hepatic parasympathectomized mice. Under high-fat diet conditions, the oral intake of nicotine lowered blood glucose levels at the daytime resting period in wild-type, but not orexin-deficient, mice. These results indicated that the chronic daily administration of nicotine suppressed hepatic gluconeogenesis via the hypothalamic orexin-parasympathetic nervous system. Thus, the results of the present study may provide an insight into novel chronotherapy for type 2 diabetes that targets the central cholinergic and orexinergic systems.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Cronoterapia de Medicamentos , Gluconeogénesis/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hígado/efectos de los fármacos , Nicotina/administración & dosificación , Orexinas/agonistas , Animales , Cruzamientos Genéticos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Hiperglucemia/prevención & control , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/uso terapéutico , Hipotálamo/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Nicotina/uso terapéutico , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/uso terapéutico , Obesidad/complicaciones , Obesidad/etiología , Orexinas/genética , Orexinas/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo
6.
Diabetes ; 64(2): 459-70, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25249578

RESUMEN

Circadian rhythm is crucial for preventing hepatic insulin resistance, although the mechanism remains uncovered. Here we report that the wake-active hypothalamic orexin system plays a key role in this regulation. Wild-type mice showed that a daily rhythm in blood glucose levels peaked at the awake period; however, the glucose rhythm disappeared in orexin knockout mice despite normal feeding rhythm. Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. The glucose-elevating and -lowering effects of orexin A were suppressed by adrenergic antagonists and hepatic parasympathectomy, respectively. Moreover, the expression levels of hepatic gluconeogenic genes, including Pepck, were increased and decreased by orexin A at nanomolar and femtomolar doses, respectively. These results indicate that orexin can bidirectionally regulate hepatic gluconeogenesis via control of autonomic balance, leading to generation of the daily blood glucose oscillation. Furthermore, during aging, orexin deficiency enhanced endoplasmic reticulum (ER) stress in the liver and caused impairment of hepatic insulin signaling and abnormal gluconeogenic activity in pyruvate tolerance test. Collectively, the daily glucose rhythm under control of orexin appears to be important for maintaining ER homeostasis, thereby preventing insulin resistance in the liver.


Asunto(s)
Sistema Nervioso Autónomo/efectos de los fármacos , Ritmo Circadiano , Hipotálamo/metabolismo , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular/farmacología , Hígado/efectos de los fármacos , Neuropéptidos/farmacología , Animales , Glucemia , Epinefrina/farmacología , Genes Transgénicos Suicidas , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/genética , Neuropéptidos/metabolismo , Neurotransmisores/farmacología , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
7.
Neuropeptides ; 47(3): 213-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23510906

RESUMEN

Depression is associated with insulin resistance and type 2 diabetes, although the molecular mechanism behind the pathological link remains unclear. Orexin, a hypothalamic neuropeptide regulating energy and glucose homeostasis, has been implicated in the endogenous antidepressant mechanism. To clarify whether orexin is involved in the coordination between mental and metabolic functions, we investigated the influence of orexin deficiency on social interaction behavior and glucose metabolism in mice subjected to chronic social defeat stress. Chronic stress-induced glucose intolerance and systemic insulin resistance as well as social avoidance were ameliorated by calorie restriction in an orexin-dependent manner. Moreover, orexin-deficient mice maintained under ad libitum-fed conditions after defeat stress exhibited hyperinsulinemia and elevated HOMA-IR (homeostasis model assessment for insulin resistance), despite normal fasting blood glucose levels. In a pyruvate tolerance test to evaluate hepatic insulin sensitivity, chronic stress-induced abnormal glucose elevation was observed in orexin-deficient but not wild-type mice, although both types of mice were susceptible to chronic stress. In addition, insulin-induced phosphorylation of Akt in the liver was impaired in orexin-deficient but not wild-type mice after chronic stress. These results demonstrate that the central physiological actions of orexin under ad libitum-fed conditions are required for the adaptive response to chronic defeat stress, which can prevent the development of hepatic insulin resistance but not social avoidance behavior. Moreover, calorie restriction, a paradigm to strongly activate orexin neurons, appears to prevent the persistence of depression-like behavior per se, leading to the amelioration of impaired glucose metabolism after chronic stress; therefore, we suggest that hypothalamic orexin system is the key for inhibiting the exacerbating link between depression and type 2 diabetes.


Asunto(s)
Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Hígado/efectos de los fármacos , Neuropéptidos/fisiología , Predominio Social , Estrés Psicológico/fisiopatología , Animales , Western Blotting , Corticosterona/sangre , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/genética , Orexinas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ácido Pirúvico/metabolismo , Estrés Psicológico/metabolismo
8.
Endocr J ; 59(5): 365-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22293586

RESUMEN

Hypothalamic orexin neurons are known to regulate sleep/wake stability, feeding behavior, emotions, autonomic nerve activity, and whole-body energy metabolism. In addition, emerging evidence indicates that orexin contributes to central regulation of glucose homeostasis. Intriguingly, central administration of orexin is reported to cause blood glucose-elevating effect or blood glucose-lowering effect in rodents, depending on the experimental conditions. Here we reviewed the recent reports regarding the mode and mechanism of actions of orexin on these two opposing effects, and discuss the functional significance for the maintenance of glucose homeostasis. The fact that orexin exhibits biphasic effects on autonomic nerve activity and lipolysis suggests that orexin dually regulates the glucose appearance. In fact, orexin neurons are activated not only depending on the demand for glucose but also according to a circadian rhythm in the suprachiasmatic nucleus. The excited orexin neurons appear to alter the sympathetic or parasympathetic outflow to the periphery, and modulate the glucose production and utilization. Furthermore, deficiency of orexin action, particularly reduction of orexin 2 receptor-signaling, disrupts the mechanism for protection against insulin resistance associated with aging or induced by chronic high fat feeding in mice. Taken together, hypothalamic orexin system may manage multiple tasks to coordinate the interconnection among the arousal, feeding, circadian, and glucose homeostasis pathways.


Asunto(s)
Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Transducción de Señal , Animales , Glucemia/metabolismo , Ritmo Circadiano , Humanos , Hipotálamo/metabolismo , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Neuronas/metabolismo , Neuropéptidos/deficiencia , Receptores de Orexina , Orexinas , Isoformas de Proteínas/metabolismo
9.
J Biol Chem ; 279(15): 14835-43, 2004 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-14744864

RESUMEN

SH2-containing inositol phosphatase 2 (SHIP2) is a physiologically important negative regulator of insulin signaling by hydrolyzing the phosphatidylinositol (PI) 3-kinase product PI 3,4,5-trisphosphate in the target tissues of insulin. Targeted disruption of the SHIP2 gene in mice resulted in increased insulin sensitivity without affecting biological systems other than insulin signaling. Therefore, we investigated the molecular mechanisms by which SHIP2 specifically regulates insulin-induced metabolic signaling in 3T3-L1 adipocytes. Insulin-induced phosphorylation of Akt, one of the molecules downstream of PI3-kinase, was inhibited by expression of wild-type SHIP2, whereas it was increased by expression of 5'-phosphatase-defective (DeltaIP) SHIP2 in whole cell lysates. The regulatory effect of SHIP2 was mainly seen in the plasma membrane (PM) and low density microsomes but not in the cytosol. In this regard, following insulin stimulation, a proportion of Akt2, and not Akt1, appeared to redistribute from the cytosol to the PM. Thus, insulin-induced phosphorylation of Akt2 at the PM was predominantly regulated by SHIP2, whereas the phosphorylation of Akt1 was only minimally affected. Interestingly, insulin also elicited a subcellular redistribution of both wild-type and DeltaIP-SHIP2 from the cytosol to the PM. The degree of this redistribution was inhibited in part by pretreatment with PI3-kinase inhibitor. Although the expression of a constitutively active form of PI3-kinase myr-p110 also elicited a subcellular redistribution of SHIP2 to the PM, expression of SHIP2 appeared to affect the myr-p110-induced phosphorylation, and not the translocation, of Akt2. Furthermore, insulin-induced phosphorylation of Akt was effectively regulated by SHIP2 in embryonic fibroblasts derived from knockout mice lacking either insulin receptor substrate-1 or insulin receptor substrate-2. These results indicate that insulin specifically stimulates the redistribution of SHIP2 from the cytosol to the PM independent of 5'-phosphatase activity, thereby regulating the insulin-induced translocation and phosphorylation of Akt2 at the PM.


Asunto(s)
Membrana Celular/metabolismo , Insulina/metabolismo , Monoéster Fosfórico Hidrolasas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células 3T3-L1 , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Western Blotting , Cromonas/farmacología , Citosol/metabolismo , ADN Complementario/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Microsomas/metabolismo , Modelos Genéticos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Pruebas de Precipitina , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-akt , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA