Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nano Lett ; 22(19): 7944-7951, 2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36129470

RESUMEN

In this study, facile salt-assisted chemical vapor deposition (CVD) was used to synthesize ultrathin non-van der Waals chromium sulfide (Cr2S3) with a thickness of ∼1.9 nm. The structural transformation of as-grown Cr2S3 was studied using advanced in situ heating techniques combined with transmission electron microscopy (TEM). Two-dimensional (2D) and quasi-one-dimensional (1D) samples were fabricated to investigate the connection between specific planes and the dynamic behavior of the structural variation. The rearrangement of atoms during the phase transition was driven by the loss of sulfur atoms at elevated temperatures, resulting in increased free energy. A decrease in the ratio of the (001) plane led to an overall increase in surface energy, thus lowering the critical phase transition temperature. Our study provides detailed insight into the mechanism of structural transformation and the critical factors governing transition temperature, thus paving the way for future studies on intriguing Cr-S compounds.

2.
Small ; 18(7): e2106411, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34995002

RESUMEN

2D materials have great potential for not only device scaling but also various applications. To prompt the development of 2D electronics and optoelectronics, a better understanding of the limitation of materials is essential. Material failure caused by bias can lead to variations in device behavior and even electrical breakdown. In this study, the structural evolution of monolayer MoS2 with high bias is revealed via in situ transmission electron microscopy at the atomic scale. The biasing process is recorded and studied with the aid of aberration-corrected scanning transmission electron microscopy. The effects of electron beam irradiation and biasing are also discussed through the combination of experiments and theory. It is found that the Mo nanoclusters result from disintegration of MoS2 and sulfur depletion, which are induced by Joule heating. The thermal stress can also damage the MoS2 layer and form long cracks in both in situ and ex situ biasing cases. Investigation of the results obtained with different applied voltages helps to further verify the mechanism of evolution and provide a comprehensive study of the function of biasing.

3.
Small ; 16(3): e1905516, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31825564

RESUMEN

Layered MoS2 is a prospective candidate for use in energy harvesting, valleytronics, and nanoelectronics. Its properties strongly related to its stacking configuration and the number of layers. Due to its atomically thin nature, understanding the atomic-level and structural modifications of 2D transition metal dichalcogenides is still underdeveloped, particularly the spatial control and selective precision. Therefore, the development of nanofabrication techniques is essential. Here, an atomic-scale approach used to sculpt 2D few-layer MoS2 into lateral heterojunctions via in situ scanning/transmission electron microscopy (STEM/TEM) is developed. The dynamic evolution is tracked using ultrafast and high-resolution filming equipment. The assembly behaviors inherent to few-layer 2D-materials are observed during the process and included the following: scrolling, folding, etching, and restructuring. Atomic resolution STEM is employed to identify the layer variation and stacking sequence for this new 2D-architecture. Subsequent energy-dispersive X-ray spectroscopy and electron energy loss spectroscopy analyses are performed to corroborate the elemental distribution. This sculpting technique that is established allows for the formation of sub-10 nm features, produces diverse nanostructures, and preserves the crystallinity of the material. The lateral heterointerfaces created in this study also pave the way for the design of quantum-relevant geometries, flexible optoelectronics, and energy storage devices.

4.
Am J Physiol Heart Circ Physiol ; 313(2): H283-H292, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550180

RESUMEN

PRKAG2 encodes the γ2-subunit isoform of 5'-AMP-activated protein kinase (AMPK), a heterotrimeric enzyme with major roles in the regulation of energy metabolism in response to cellular stress. Mutations in PRKAG2 have been implicated in a unique hypertrophic cardiomyopathy (HCM) characterized by cardiac glycogen overload, ventricular preexcitation, and hypertrophy. We identified a novel, de novo PRKAG2 mutation (K475E) in a neonate with prenatal onset of HCM. We aimed to investigate the cellular impact, signaling pathways involved, and therapeutic options for K475E mutation using cells stably expressing human wild-type (WT) or the K475E mutant. In human embryonic kidney-293 cells, the K475E mutation induced a marked increase in the basal phosphorylation of T172 and AMPK activity, reduced sensitivity to AMP in allosteric activation, and a loss of response to phenformin. In H9c2 cardiomyocytes, the K475E mutation induced inhibition of AMPK and reduced the response to phenformin and increases in the phosphorylation of p70S6 kinase (p70S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). Primary fibroblasts from the patient with the K475E mutation also showed marked increases in the phosphorylation of p70S6K and 4E-BP1 compared with those from age-matched, nondiseased controls. Moreover, overexpression of K475E induced hypertrophy in H9c2 cells, which was effectively reversed by treatment with rapamycin. Taken together, we have identified a novel, de novo infantile-onset PRKAG2 mutation causing HCM. Our study suggests the K475E mutation induces alteration in basal AMPK activity and results in a hypertrophy phenotype involving the mechanistic target of rapamycin signaling pathway, which can be reversed with rapamycin.NEW & NOTEWORTHY We identified a novel, de novo PRKAG2 mutation (K475E) in the cystathionine ß-synthase 3 repeat, a region critical for AMP binding but with no previous reported mutation. Our data suggest the mutation affects AMP-activated protein kinase activity, activates cell growth pathways, and results in cardiac hypertrophy, which can be reversed with rapamycin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Cardiomiopatía Hipertrófica/genética , Mutación Missense , Miocitos Cardíacos/enzimología , Transducción de Señal , Proteínas Quinasas Activadas por AMP/química , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato/metabolismo , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Análisis Mutacional de ADN , Activación Enzimática , Fibroblastos/enzimología , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular , Modelos Moleculares , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenformina/farmacología , Fenotipo , Fosfoproteínas/metabolismo , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transfección
5.
J Mol Cell Cardiol ; 53(4): 532-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22824041

RESUMEN

In contrast with pathological hypertrophy, exercise-induced physiological hypertrophy is not associated with electrical abnormalities or increased arrhythmia risk. Recent studies have shown that increased cardiac-specific expression of phosphoinositide-3-kinase-α (PI3Kα), the key mediator of physiological hypertrophy, results in transcriptional upregulation of ion channel subunits in parallel with the increase in myocyte size (cellular hypertrophy) and the maintenance of myocardial excitability. The experiments here were undertaken to test the hypothesis that Akt1, which underlies PI3Kα-induced cellular hypertrophy, mediates the effects of augmented PI3Kα signaling on the transcriptional regulation of cardiac ion channels. In contrast to wild-type animals, chronic exercise (swim) training of mice (Akt1(-/-)) lacking Akt1 did not result in ventricular myocyte hypertrophy. Ventricular K(+) current amplitudes and the expression of K(+) channel subunits, however, were increased markedly in Akt1(-/-) animals with exercise training. Expression of the transcripts encoding inward (Na(+) and Ca(2+)) channel subunits were also increased in Akt1(-/-) ventricles following swim training. Additional experiments in a transgenic mouse model of inducible cardiac-specific expression of constitutively active PI3Kα (icaPI3Kα) revealed that short-term activation of PI3Kα signaling in the myocardium also led to the transcriptional upregulation of ion channel subunits. Inhibition of cardiac Akt activation with triciribine in this (inducible caPI3Kα expression) model did not prevent the upregulation of myocardial ion channel subunits. These combined observations demonstrate that chronic exercise training and enhanced PI3Kα expression/activity result in transcriptional upregulation of myocardial ion channel subunits independent of cellular hypertrophy and Akt signaling.


Asunto(s)
Miocardio/metabolismo , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasa/metabolismo , Condicionamiento Físico Animal , Canales de Potasio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Cardiomegalia , Corazón/fisiología , Ventrículos Cardíacos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/biosíntesis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Ribonucleósidos/farmacología , Regulación hacia Arriba
6.
Cell Commun Signal ; 10(1): 29, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23061721

RESUMEN

BACKGROUND: Insulin receptor (InsR) and insulin signaling proteins are widely distributed throughout the kidney cortex. Insulin signaling can act in the kidney in multiple ways, some of which may be totally independent of its primary role of the maintenance of whole-body glucose homeostasis. However, descriptions of the insulin signaling in renal glomerular mesangial cells (MCs) are quite limited and the roles of insulin signaling in MC functions have not been sufficiently elucidated. RESULTS: InsR silencing induced a unique phenotype of reduced fibronectin (FN) accumulation in renal glomerular MCs. Transcription level of FN was not significantly changed in the InsR silenced cells, suggesting the phenotype switching was caused by post-transcriptional modification. The decreased expression of InsR was associated with enhanced activity of insulin-like growth factor-1 receptor (IGF-1R)/PI3K/Akt signaling pathway which contributed in part to the attenuation of cellular FN accumulation. Formation of IGF-1R homodimer was increased in the InsR silenced cells. The InsR silenced cells also showed increased sensitivity to exogenous IGF-1, and increased PI3K activity was reversed significantly by incubating cells with IGF-1R specific antagonist, AG538. PI3K/Akt dependent activation of cAMP responsive element-binding protein (CREB)-1 induced expression of matrix metalloproteinase (MMP)-9 and suppressing MMP activity by doxycycline partially reversed FN accumulation in the InsR silenced cells. CONCLUSIONS: The effects of InsR silencing on cellular FN accumulation in vitro are, at least partially, mediated by increased degradation of FN by MMPs which is induced by enhanced signaling sequence of IGF-1R/PI3K/Akt/CREB-1.

7.
ACS Appl Mater Interfaces ; 14(36): 41156-41164, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36037311

RESUMEN

Contact engineering of two-dimensional semiconductors is a central issue for performance improvement of micro-/nanodevices based on these materials. Unfortunately, the various methods proposed to improve the Schottky barrier height normally require the use of high temperatures, chemical dopants, or complex processes. This work demonstrates that diffused electron beam energy (DEBE) treatment can simultaneously reduce the Schottky barrier height and enable the direct writing of electrical circuitry on van der Waals semiconductors. The electron beam energy projected into the region outside the electrode diffuses into the main channel, producing selective-area n-type doping in a layered MoTe2 (or MoS2) field-effect transistor. As a result, the Schottky barrier height at the interface between the electrode and the DEBE-treated MoTe2 channel is as low as 12 meV. Additionally, because selective-area doping is possible, DEBE can allow the formation of both n- and p-type doped channels within the same atomic plane, which enables the creation of a nonvolatile and homogeneous MoTe2 p-n rectifier with an ideality factor of 1.1 and a rectification ratio of 1.3 × 103. These results indicate that the DEBE method is a simple, efficient, mask-free, and chemical dopant-free approach to selective-area doping for the development of van der Waals electronics with excellent device performances.

8.
Biochem J ; 423(1): 129-43, 2009 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-19604148

RESUMEN

Diabetic nephropathy is associated with mesangial ECM (extracellular matrix) accumulation. We have shown that AT-1R [Ang II (angiotensin II) type I receptor] signalling induces ECM proteins via transactivation of PI3K (phosphoinositide 3-kinase) in mesangial cells. In the present study, we examined the mechanisms underlying the effect of high ambient glucose on cell proliferation and ECM expansion in a mesangial context. High glucose induced increases in PI3K activity, proliferation and ECM accumulation in mesangial cells. These effects were abrogated by losartan, an AT-1R antagonist, but not by [Sar1,Thr8]-Ang II (Sar is sarcosine), an inactive analogue of Ang II, or by a neutralizing antibody against Ang I/II. Overexpression of a constitutively active PI3Kalpha or AT-1R alone was sufficient to induce similar changes by high glucose. In contrast, overexpression of an inactive AT-1R lowered the basal levels and rendered the cells non-responsive to high glucose. Moreover, cells overexpressing wild-type AT-1R had enhanced sensitivity to acute Ang II stimulation. These cells, however, did not respond to conditioned medium obtained from mesangial cells cultured in high glucose. We further demonstrated that iAng (intracellular Ang II) can be induced by high glucose but only under certain conditions. Efficient suppression of iAng by short hairpin RNA against angiotensinogen, however, did not affect high glucose-induced effects on MES-13 cells. These results suggest that high ambient glucose induces activation of AT-1R in an Ang II-independent manner to transactivate PI3K, resulting in proliferation and ECM accumulation in mesangial cells.


Asunto(s)
Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Glucosa/farmacología , Células Mesangiales/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II/antagonistas & inhibidores , Bloqueadores del Receptor Tipo 1 de Angiotensina II , Animales , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Matriz Extracelular/metabolismo , Humanos , Isoenzimas/metabolismo , Isoenzimas/fisiología , Células Mesangiales/metabolismo , Células Mesangiales/fisiología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología
9.
Endocrinology ; 149(12): 6449-61, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18719028

RESUMEN

Recent studies have demonstrated that the beta2-adrenergic receptor (beta2AR)-Galphai signaling pathway exerts a cardiac antiapoptotic effect. The goals of this study were to determine the intracellular signaling factors involved in beta2AR-mediated protection against doxorubicin-induced apoptosis in H9c2 cardiomyocyte and explore the impact of high ambient glucose on the antiapoptotic effect. Under physiological glucose environment (100 mg/dl), beta2AR stimulation prevented doxorubicin-induced apoptosis, which was attenuated by cotreatment with wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, or transfection of a dominant-negative Akt. Inhibition of Src kinase with 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine or cSrc small interfering RNA 32 also attenuated the antiapoptotic effect. Inhibition of platelet-derived growth factor receptor (PDGFR) with AG1296 reversed the beta2AR-induced antiapoptotic effect. Transfection of an active Src cDNA (Y529F) alone was sufficient to render the cells resistant to apoptosis, and the resistance was blocked by wortmannin. Transfection of an active PI3K minigene (iSH2-p110) alone also induced resistance to apoptosis, and the resistance was reversed by an Akt-inhibitor but not by AG1296. High ambient glucose (450 mg/dl) caused two major effects: 1) it significantly reduced betaAR-induced PDGFR phosphorylation, Src kinase activity, and activation of PI3K signaling pathway; and 2) it partially attenuated beta2AR-induced antiapoptotic effect. These data provide in vitro evidence supporting a signaling cascade by which beta2AR exerts a protective effect against doxorubicin-induced apoptosis via sequential involvement of Galphai, Gbetagamma, Src, PDGFR, PI3K, and Akt. High ambient glucose significantly attenuates beta2AR-mediated cardioprotection by suppressing factors involved in this cascade including PDGFR, Src, and PI3K/Akt.


Asunto(s)
Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Glucosa/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores Adrenérgicos beta/fisiología , Animales , Apoptosis/fisiología , Línea Celular , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Immunoblotting , Inmunoprecipitación , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ratas , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
PLoS One ; 9(8): e104888, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25127116

RESUMEN

Doxorubicin (Dox) is one of the most widely used antitumor drugs, but its cumulative cardiotoxicity have been major concerns in cancer therapeutic practice for decades. Recent studies established that metformin (Met), an oral anti-diabetic drug, provides protective effects in Dox-induced cardiotoxicity. Met has been shown to increase fatty acid oxidation, an effect mediated by AMP activated protein kinase (AMPK). Here we delineate the intracellular signaling factors involved in Met mediated protection against Dox-induced cardiotoxicity in the H9c2 cardiomyoblast cell line. Treatment with low dose Met (0.1 mM) increased cell viabilities and Ki-67 expressions while decreasing LDH leakages, ROS generations and [Ca2+]i. The protective effect was reversed by a co-treatment with compound-C, an AMPK specific inhibitor, or by an over expression of a dominant-negative AMPKα cDNA. Inhibition of PKA with H89 or a suppression of Src kinase by a small hairpin siRNA also abrogated the protective effect of the low dose Met. Whereas, with a higher dose of Met (1.0 mM), the protective effects were abolished regardless of the enhanced AMPK, PKA/CREB1 and Src kinase activity. In high dose Met treated cells, expression of platelet-derived growth factor receptor (PDGFR) was significantly suppressed. Furthermore, the protective effect of low dose Met was totally reversed by co-treatment with AG1296, a PDGFR specific antagonist. These data provide in vitro evidence supporting a signaling cascade by which low dose Met exerts protective effects against Dox via sequential involvement of AMPK, PKA/CREB1, Src and PDGFR. Whereas high dose Met reverses the effect by suppressing PDGFR expression.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Metformina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Cardiotoxicidad , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Humanos , Mioblastos Cardíacos/efectos de los fármacos , Mioblastos Cardíacos/metabolismo , Ratas , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Familia-src Quinasas/metabolismo
12.
PLoS One ; 6(10): e26581, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22028912

RESUMEN

Sustained ß-adrenergic receptors (ßAR) activation leads to cardiac hypertrophy and prevents left ventricular (LV) atrophy during LV unloading. The immediate signaling pathways downstream from ßAR stimulation, however, have not been well investigated. The current study was to examine the early cardiac signaling mechanism(s) following ßAR stimulation. In adult C57BL/6 mice, acute ßAR stimulation induced significant increases in PI3K activity and activation of Akt and ERK1/2 in the heart, but not in lungs or livers. In contrast, the same treatment did not elicit these changes in ß(1)/ß(2)AR double knockout mice. We further showed the specificity of ß(2)AR in this crosstalk as treatment with formoterol, a ß(2)AR-selective agonist, but not dobutamine, a predominantly ß(1)AR agonist, activated cardiac Akt and ERK1/2. Acute ßAR stimulation also significantly increased the phosphorylation of mTOR (the mammalian target of rapamycin), P70S6K, ribosomal protein S6, GSK-3α/ß (glycogen synthase kinase-3α/ß), and FOXO1/3a (the forkhead box family of transcription factors 1 and 3a). Moreover, acute ßAR stimulation time-dependently decreased the mRNA levels of the muscle-specific E3 ligases atrogin-1 and muscle ring finger protein-1 (MuRF1) in mouse heart. Our results indicate that acute ßAR stimulation in vivo affects multiple cardiac signaling cascades, including the PI3K signaling pathway, ERK1/2, atrogin-1 and MuRF1. These data 1) provide convincing evidence for the crosstalk between ßAR and PI3K signaling pathways; 2) confirm the ß(2)AR specificity in this crosstalk in vivo; and 3) identify novel signaling factors involved in cardiac hypertrophy and LV unloading. Understanding of the intricate interplay between ß(2)AR activation and these signaling cascades should provide critical clues to the pathogenesis of cardiac hypertrophy and enable identification of targets for early clinical interaction of cardiac lesions.


Asunto(s)
Miocardio/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Agonistas Adrenérgicos beta/farmacología , Animales , Factores de Transcripción Forkhead/metabolismo , Técnicas de Inactivación de Genes , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Musculares/genética , Miocardio/metabolismo , Especificidad de Órganos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Adrenérgicos beta 1/deficiencia , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/deficiencia , Receptores Adrenérgicos beta 2/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/genética
13.
Cardiovasc Res ; 87(4): 704-12, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20410290

RESUMEN

AIMS: We have demonstrated an important role of bone marrow-derived stem cells in preservation of myocardial function. We investigated whether Akt-1 of lin(-)c-kit(+) stem cells preserves ventricular function following myocardial infarction (MI). METHODS AND RESULTS: Isolated lin(-)c-kit(+) cells were conjugated with anti-c-kit heteroconjugated to anti-vascular cell adhesion molecule to facilitate the attachment of stem cells into damaged tissues. Female severe combined immunodeficient mice were used as recipients. MI was created by ligation of the left descending artery. After 48 h, animals were divided into four groups: (i) sham (n = 5): animals underwent thoracotomy without MI; (ii) MI (n = 5): animals underwent MI and received medium; (iii) MI + wild-type (Wt) stem cells (n = 6): MI animals received 5 x 10(5) Wt lin(-)c-kit(+) stem cells; (iv) MI + Akt-1(-/-) stem cells (n = 6): MI animals received 5 x 10(5) Akt-1(-/-) lin(-)c-kit(+) stem cells. Two weeks later, left ventricular function was measured in the Langendorff mode. The peripheral administration of Wt armed stem cells into MI animals restored ventricular function, which was absent in animals receiving Akt-1(-/-) cells. Real-time PCR indicates a decrease in SRY3, a Y chromosome marker in hearts receiving Akt-1(-/-) cells. An increase in angiogenic response was demonstrated in hearts receiving Wt stem cells but not Akt-1(-/-) stem cells. CONCLUSION: Our results demonstrate that the peripheral administration of Wt lin(-)c-kit(+) stem cells restores ventricular function and promotes angiogenic response following MI. These benefits were abrogated in MI mice receiving Akt-1(-/-) stem cells, suggesting the pivotal role of Akt-1 in mediating stem cells to protect MI hearts.


Asunto(s)
Linaje de la Célula , Infarto del Miocardio/cirugía , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Trasplante de Células Madre , Células Madre/enzimología , Función Ventricular Izquierda , Animales , Anticuerpos Biespecíficos , Cardiomegalia/enzimología , Cardiomegalia/etiología , Cardiomegalia/prevención & control , Adhesión Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-kit/inmunología , Recuperación de la Función , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/inmunología
14.
Am J Physiol Heart Circ Physiol ; 295(4): H1690-4, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18723766

RESUMEN

The phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates multiple cellular processes including cell survival/apoptosis and growth. In the cardiac context, PI3Kalpha plays important roles in cardiac growth. We have shown that cardiac PI3K activity is highly regulated during development, with the highest levels found during the fetal-neonatal transition period and the lowest levels in the adult. There is a close relationship between cardiomyocyte proliferation and cardiac PI3K activity. In adult transgenic mice, however, the prolonged constitutive activation of PI3Kalpha in the heart results in hypertrophy. To develop a strategy to allow temporally controlled overexpression of cardiac PI3Kalpha, we engineered a tetracycline (tet) transactivator tet-off controlled transgenic mouse line with a conditional overexpression of a cardiac-specific fusion protein of the SH2 domain of p85 and p110alpha. Cardiac PI3K activity and Akt phosphorylation were significantly increased in adult mice after transgene induction following the removal of doxycycline for 2 wk. The heart weight-to-body weight ratio was not changed, and there were no signs of cardiomyopathy. The overexpression of PI3Kalpha resulted in increased left ventricular (LV) developed pressure and the maximal and minimal positive values of the first derivative of LV pressure, but not heart rate, as assessed in Langendorff hearts. Mice overexpressing PI3Kalpha also had increases in the levels of Ca(2+)-regulating proteins, including the L-type Ca(2+) channels, ryanodine receptors, and sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a. Thus the temporally controlled overexpression of cardiac PI3Kalpha does not induce hypertrophy or cardiomyopathy but results in increased contractility, probably via the increased expression of multiple Ca(2+)-regulating proteins. These distinct phenotypes suggest a fundamental difference between transgenic mice with temporal or prolonged activation of cardiac PI3Kalpha.


Asunto(s)
Contracción Miocárdica , Miocardio/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Función Ventricular Izquierda , Animales , Señalización del Calcio , Isoenzimas , Ratones , Ratones Transgénicos , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Tiempo , Regulación hacia Arriba , Presión Ventricular
15.
J Biol Chem ; 282(26): 18819-30, 2007 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-17493931

RESUMEN

Chronic activation of the angiotensin II (ANG II) type 1 receptor (AT-1R) is critical in the development of chronic kidney disease. ANG II activates mesangial cells (MCs) and stimulates the synthesis of extracellular matrix components. To determine the molecular mechanisms underlying the induction of MC collagen, a mouse mesangial cell line MES-13 was employed. ANG II treatment induced an increase in collagen synthesis, which was abrogated by co-treatment with losartan (an AT-1R antagonist), wortmannin (a phosphoinositide 3-kinase (PI3K) inhibitor), an Akt inhibitor, and stable transfection of dominant negative-Akt1. ANG II induced a significant increase in PI3K activity, which was abolished by co-treatment with losartan or 2',5'-dideoxyadenosine (2',5'-DOA, an adenylyl cyclase inhibitor) but not by PD123319 (an AT-2R antagonist) or H89 (a protein kinase A (PKA) inhibitor). The Epac (exchange protein directly activated by cAMP)-specific cAMP analog, 8-pHPT-2'-O-Me-cAMP, significantly increased PI3K activity, whereas a PKA-specific analog, 6-benzoyladenosine-cAMP, showed no effect. The ANG II-induced increase in PI3K activity was also blocked by co-treatment with PP2, an Src inhibitor, or AG1478, an epidermal growth factor receptor (EGFR) antagonist. ANG II induced phosphorylation of Akt and p70S6K and EGFR, which was abrogated by knockdown of c-Src by small interference RNA. Knockdown of Src also effectively abolished ANG II-induced collagen synthesis. Conversely, stable transfection of a constitutively active Src mutant enhanced basal PI3K activity and collagen production, which was abrogated by AG1478 but not by 2',5'-DOA. Moreover, acute treatment with ANG II significantly increased Src activity, which was abrogated with co-treatment of 2',5'-DOA. Taken together, these results suggest that ANG II induces collagen synthesis in MCs by activating the ANG II/AT-1R-EGFR-PI3K pathway. This transactivation is dependent on cAMP/Epac but not on PKA. Src kinase plays a pivotal role in this signaling pathway between cAMP and EGFR. This is the first demonstration that an AT1R-PI3K/Akt crosstalk, along with transactivation of EGFR, mediates ANG II-induced collagen synthesis in MCs.


Asunto(s)
Angiotensina II/metabolismo , Colágeno/biosíntesis , Células Mesangiales/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Vasoconstrictores/metabolismo , Adenilil Ciclasas/metabolismo , Angiotensina II/farmacología , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Receptores ErbB/metabolismo , Células Mesangiales/citología , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vasoconstrictores/farmacología , Familia-src Quinasas/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 293(1): H385-93, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17369456

RESUMEN

Stimulation of cardiac beta-adrenergic receptors (beta-AR) activates both the G(s)- and G(i)-coupled signaling cascades, including the phosphoinositide 3 kinase (PI3K) pathway, that have important physiological implications. Multiple isoforms of PI3K exist in the heart. The goals of this study were to examine the intracellular signaling pathways linking beta-AR to PI3K and to identify the PI3K isoform mediating this transactivation in a cardiac context. Acute beta-AR stimulation with isoproterenol resulted in increased tyrosine kinase-associated PI3K activity and phosphorylation of Akt and p70S6K in H9c2 cardiomyocytes. Cotreatment with ICI-118,551, but not CGP-20712, abolished the increase in PI3K activity, suggesting a beta(2)-AR-mediated event. PI3K activation was also abrogated by cotreatment with pertussis toxin, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolol[3,4-d]pyrimidine (PP2, a selective Src-family tyrosine kinases inhibitor), or AG-1296 [selective platelet-derived growth factor receptor (PDGFR) inhibitor] but not with an inhibitor for protein kinase A, protein kinase C, Ras, adenylyl cyclase, epidermal growth factor receptor, or insulin-like growth factor-1 receptor. beta-AR stimulation induced an increase in tyrosine phosphorylation of PDGFR, which was abolished by inhibition of Src either by PP2 or small interfering RNA. Moreover, H9c2 cardiomyocytes stably transfected with a vector expressing a Gbetagamma sequestrant peptide derived from the COOH-terminus of beta-AR kinase-1 failed to activate PI3K after beta-AR stimulation, suggesting Gbetagamma is required for the transactivation. Furthermore, acute beta-AR stimulation in vivo resulted in increases in PDGFR-associated PI3K and PI3Kalpha isoform activities but not the activities of other isoforms (PI3Kbeta, -delta, -gamma) in adult mouse heart. Taken together, these data provide in vitro and in vivo evidence for a novel mechanism of beta-AR-mediated transactivation of cardiac PI3Kalpha via sequential involvement of Galpha(i)/Gbetagamma, Src, and PDGFR.


Asunto(s)
Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Ratas
17.
Am J Physiol Heart Circ Physiol ; 289(5): H1834-42, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16006545

RESUMEN

Signaling pathways underlying transition of cardiomyocyte growth from hyperplasia in fetal/newborn to hypertrophy in postnatal/adult hearts are not well understood. We have shown that beta-adrenergic receptor (beta-AR)-mediated regulation of neonatal cardiomyocyte proliferation involves p70 ribosomal protein S6 kinase (p70S6K). Here we examined the ontogeny of phosphoinositide 3-kinase (PI3K)/p70S6K signaling pathway in rat hearts and investigated the influence of beta-AR on this pathway during development. Cardiac PI3K and p70S6K1 activities were high in the embryonic day 20 fetus, decreased gradually postnatally, and were low in the adult. In contrast, p70S6K2 was barely detectable. Phosphorylation of p70S6K1, Akt, and phosphoinositide-dependent protein kinase 1 were markedly increased in late gestation and early postnatal life but not in adult hearts. Phosphatase and tensin homolog on chromosome 10 (PTEN), a negative regulator of PI3K, was highly expressed in adult hearts but only at low levels and mostly in the phosphorylated (inactivated) form in the fetus. Beta-AR stimulation resulted in increased cardiac p70S6K1 activity only in animals > or = 2 wk old, whereas Akt level was increased in all developmental stages tested. These increases were accompanied by increased Bcl-2 associated death promoter (Ser136) phosphorylation without changes in PTEN level. Thus there is globally high input of cardiac PI3K signaling during the fetal-neonatal transition period. Inactivation of PTEN may in part contribute to the high activity of PI3K signaling, which coincides with the period of high cardiomyocyte proliferation. Beta-AR stimulation activates cardiac p70S6K1 and Akt in postnatal animals and may activate cardiac survival signals. These data provide further evidence for the importance of beta-AR and PI3K signaling in the regulation of cardiac growth during development.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Corazón/crecimiento & desarrollo , Corazón/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/efectos de los fármacos , Envejecimiento/fisiología , Animales , Western Blotting , Proliferación Celular , Femenino , Corazón/efectos de los fármacos , Miocardio/citología , Miocardio/enzimología , Miocardio/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Embarazo , Ratas , Ratas Sprague-Dawley
18.
J Allergy Clin Immunol ; 110(6 Suppl): S247-54, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12464932

RESUMEN

To study the mechanism underlying glucocorticoid regulation of the beta(1)-adrenergic receptor (beta(1)AR), we identified a 43-bp region (-1274 to -1232 from the translation start site) that contains a novel glucocorticoid regulatory unit (GRU) that confers glucocorticoid responsiveness. The sequence encompassing the GRU is (5')TAATTA(3'), which is a core-binding motif for the homeodomain proteins; an E-box ((5')CACGTG(3')) binding site for the Myc/Max family proteins, and an overlapping glucocorticoid response element half-site ((5')TGTTCT(3')). We showed that the half-site is critical for GRU-protein interactions, which also require binding of proteins to the E-box and the homeodomain region. Expression of proteins binding to the GRU was shown to be developmentally regulated, being high in embryonic hearts, reduced in newborn hearts, and undetectable in adult hearts. Overexpression of c-myc antisense significantly reduced glucocorticoid responsiveness of the beta(1)AR gene. We further demonstrated that transcriptional regulation of the beta(1)AR gene is closely related to that of the c-myc gene and that the beta(1)AR may be a potential target of c-myc. We conclude that the ovine beta(1)AR gene contains a novel, functional GRU and that the nuclear factors that transactivate through this element may have important developmental implications.


Asunto(s)
Catecolaminas/metabolismo , Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Humanos , Ratas , Receptores Adrenérgicos beta 1/genética , Ovinos
19.
Am J Physiol Heart Circ Physiol ; 284(6): H2146-52, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12742828

RESUMEN

The beta(1)-adrenergic receptor (beta(1)AR) gene contains binding sites for myc/max proteins within a glucocorticoid response element. Transcriptional activation of the beta(1)AR is the result of cooperative binding between c-myc and the glucocorticoid receptor on the beta(1)AR promoter. The transcriptional regulation of both beta(1)AR and c-myc are developmentally regulated. We used transcription rate assays of nuclei isolated from fetal hearts to demonstrate a fivefold increase in the transcription rate of beta(1)AR vs. postnatal hearts (P < 0.01). This was associated with a fourfold increase in c-myc transcription. Transcription rate assays performed in a rat fibroblast cell line that overexpresses c-myc (myc(+/+)) showed similarly increased beta(1)AR expression compared with the wild-type cell line. Transient transfection experiments in the myc(+/+) cells demonstrated robust expression of beta(1)AR promoter constructs, which was abrogated by mutation of the myc/max binding site or by cotransfection with a c-myc antisense expression vector. These results suggest that the regulation of cardiac beta(1)AR transcription and the expression of c-myc are tightly integrated.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Corazón/embriología , Miocardio/metabolismo , Receptores Adrenérgicos beta 1/biosíntesis , Receptores Adrenérgicos beta 1/genética , Animales , Animales Recién Nacidos/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Femenino , Cinética , Células Musculares/metabolismo , Oligodesoxirribonucleótidos Antisentido , Embarazo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Ratas , Ratas Sprague-Dawley , Ovinos , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Transfección
20.
J Biol Chem ; 279(30): 31948-55, 2004 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-15131122

RESUMEN

The immunosuppressant rapamycin has been shown to inhibit G(1)/S transition of the cell cycle. This inhibition is thought to be mediated by maintenance of the threshold levels of cyclin-dependent kinase (CDK) inhibitor p27(Kip1) (p27) and inhibition of p70 s6 kinase (p70(s6k)). However, recent evidence suggests that cells still remain sensitive to rapamycin in the absence of functional p27 or p70(s6k). Here, we show that rapamycin represses cyclin D3 levels in activated human T lymphocytes with no inhibitory effects on cyclin D2. Furthermore, rapamycin elicits similar cyclin D3 modulatory effects in B lymphocytes. The overall effect of rapamycin on cyclin D3 leads to impaired formation of active complexes with Cdk4 or Cdk6 and subsequent inhibition of cyclin D3/CDK kinase activity. Decrease in cyclin D3 protein levels is due to translational repression and not due to attenuated transcription of the cyclin D3 gene. Importantly, stable overexpression of cyclin D3 (2-2.5 fold) in Jurkat T cell transfectants renders them resistant to lower doses (1-10 ng/ml) of rapamycin. These results point to a critical role of cyclin D3 in rapamycin-mediated immunosuppressive effects in T cells and cell cycle regulation in lymphocytes in general.


Asunto(s)
Ciclinas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas , Sirolimus/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Ciclina D3 , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/química , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/química , Ciclinas/metabolismo , Fase G1/efectos de los fármacos , Humanos , Técnicas In Vitro , Ionomicina/farmacología , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Sustancias Macromoleculares , Linfocitos T/citología , Acetato de Tetradecanoilforbol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA