Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nutrients ; 12(8)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32751276

RESUMEN

Skeletal muscle is the metabolic powerhouse of the body, however, dysregulation of the mechanisms involved in skeletal muscle mass maintenance can have devastating effects leading to many metabolic and physiological diseases. The lack of effective solutions makes finding a validated nutritional intervention an urgent unmet medical need. In vitro testing in murine skeletal muscle cells and human macrophages was carried out to determine the effect of a hydrolysate derived from vicia faba (PeptiStrong: NPN_1) against phosphorylated S6, atrophy gene expression, and tumour necrosis factor alpha (TNF-α) secretion, respectively. Finally, the efficacy of NPN_1 on attenuating muscle waste in vivo was assessed in an atrophy murine model. Treatment of NPN_1 significantly increased the phosphorylation of S6, downregulated muscle atrophy related genes, and reduced lipopolysaccharide-induced TNF-α release in vitro. In a disuse atrophy murine model, following 18 days of NPN_1 treatment, mice exhibited a significant attenuation of muscle loss in the soleus muscle and increased the integrated expression of Type I and Type IIa fibres. At the RNA level, a significant upregulation of protein synthesis-related genes was observed in the soleus muscle following NPN_1 treatment. In vitro and preclinical results suggest that NPN_1 is an effective bioactive ingredient with great potential to prolong muscle health.


Asunto(s)
Alimentos Funcionales/análisis , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/tratamiento farmacológico , Hidrolisados de Proteína/farmacología , Vicia faba/química , Animales , Modelos Animales de Enfermedad , Ingredientes Alimentarios , Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
3.
Behav Brain Res ; 363: 135-144, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30684511

RESUMEN

Tuberous sclerosis complex (TSC) is a rare disease caused by mutations in the TSC1 or TSC2 genes and is characterized by widespread tumour growth, intractable epilepsy, cognitive deficits and autistic behaviour. CBD has been reported to decrease seizures and inhibit tumour cell progression, therefore we sought to determine the influence of CBD on TSC pathology in zebrafish carrying a nonsense mutation in the tsc2 gene. CBD treatment from 6 to 7 days post-fertilization (dpf) induced significant anxiolytic actions without causing sedation. Furthermore, CBD treatment from 3 dpf had no impact on tsc2-/- larvae motility nor their survival. CBD treatment did, however, reduce the number of phosphorylated rpS6 positive cells, and their cross-sectional cell size. This suggests a CBD mediated suppression of mechanistic target of rapamycin (mTOR) activity in the tsc2-/- larval brain. Taken together, these data suggest that CBD selectively modulates levels of phosphorylated rpS6 in the brain and additionally provides an anxiolytic effect. This is pertinent given the alterations in mTOR signalling in experimental models of TSC. Additional work is necessary to identify upstream signal modulation and to further justify the use of CBD as a possible therapeutic strategy to manage TSC.


Asunto(s)
Cannabidiol/farmacología , Proteína S6 Ribosómica/efectos de los fármacos , Esclerosis Tuberosa/tratamiento farmacológico , Animales , Encéfalo/metabolismo , Cannabidiol/metabolismo , Cannabinoides/metabolismo , Cannabinoides/farmacología , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Locomoción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Convulsiones/patología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Esclerosis Tuberosa/fisiopatología , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/efectos de los fármacos , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Transl Psychiatry ; 7(1): e1013, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28117842

RESUMEN

Tic disorders affect ~5% of the population and are frequently comorbid with obsessive-compulsive disorder, autism, and attention deficit disorder. Histamine dysregulation has been identified as a rare genetic cause of tic disorders; mice with a knockout of the histidine decarboxylase (Hdc) gene represent a promising pathophysiologically grounded model. How alterations in the histamine system lead to tics and other neuropsychiatric pathology, however, remains unclear. We found elevated expression of the histamine H3 receptor in the striatum of Hdc knockout mice. The H3 receptor has significant basal activity even in the absence of ligand and thus may modulate striatal function in this knockout model. We probed H3R function using specific agonists. The H3 agonists R-aminomethylhistamine (RAMH) and immepip produced behavioral stereotypies in KO mice, but not in controls. H3 agonist treatment elevated intra-striatal dopamine in KO mice, but not in controls. This was associated with elevations in phosphorylation of rpS6, a sensitive marker of neural activity, in the dorsal striatum. We used a novel chemogenetic strategy to demonstrate that this dorsal striatal activity is necessary and sufficient for the development of stereotypy: when RAMH-activated cells in the dorsal striatum were chemogenetically activated (in the absence of RAMH), stereotypy was recapitulated in KO animals, and when they were silenced the ability of RAMH to produce stereotypy was blocked. These results identify the H3 receptor in the dorsal striatum as a contributor to repetitive behavioral pathology.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Agonistas de los Receptores Histamínicos/farmacología , Receptores Histamínicos H3/metabolismo , Conducta Estereotipada/efectos de los fármacos , Trastornos de Tic/genética , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Histidina Descarboxilasa/genética , Imidazoles/farmacología , Ratones , Ratones Noqueados , Fosforilación , Piperidinas/farmacología , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Trastornos de Tic/metabolismo
5.
Arthritis Rheum ; 65(7): 1843-52, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23606170

RESUMEN

OBJECTIVE: Aging-associated changes in articular cartilage represent a main risk factor for osteoarthritis (OA). Autophagy is an essential cellular homeostasis mechanism. Aging-associated or experimentally induced defects in autophagy contribute to organismal- and tissue-specific aging, while enhancement of autophagy may protect against certain aging-related pathologies such as OA. The objective of this study was to determine whether glucosamine can activate autophagy. METHODS: Chondrocytes from normal human articular cartilage were treated with glucosamine (0.1- 10 mM). Autophagy activation and phosphorylation levels of Akt, FoxO3, and ribosomal protein S6 were determined by Western blotting. Autophagosome formation was analyzed by confocal microscopy. Reporter mice systemically expressing green fluorescent protein (GFP) fused to light chain 3 (LC3) (GFP-LC3-transgenic mice) were used to assess changes in autophagy in response to starvation and glucosamine treatment. RESULTS: Glucosamine treatment of chondrocytes activated autophagy, as indicated by increased LC3-II levels, formation of LC3 puncta, and increased LC3 turnover. This was associated with glucosamine-mediated inhibition of the Akt/FoxO3/mammalian target of rapamycin pathway. Administration of glucosamine to GFP-LC3-transgenic mice markedly activated autophagy in articular cartilage. CONCLUSION: Glucosamine modulates molecular targets of the autophagy pathway in vitro and in vivo, and the enhancement of autophagy is mainly dependent on the Akt/FoxO/mTOR pathway. These findings suggest that glucosamine is an effective autophagy activator and should motivate future studies on the efficacy of glucosamine in modifying aging-related cellular changes and supporting joint health.


Asunto(s)
Autofagia/efectos de los fármacos , Cartílago Articular/citología , Condrocitos/efectos de los fármacos , Glucosamina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Condrocitos/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Neuroreport ; 24(7): 388-93, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23568219

RESUMEN

Recent clinical studies show that a low dose of dissociative anesthetic ketamine (KET) induced a rapid antidepressant response that lasted for up to 7 days. This effect could be related to the capacity of KET to acutely induce molecular mechanisms of neuroplasticity engaged after chronic treatments. KET produces its actions by binding to the glutamate N-methyl-D-aspartic acid receptor, leading to increased activation of the mammalian target of rapamycin. Ribosomal protein S6 phosphorylation (rpS6P) is downstream to mammalian target of rapamycin and p70S6K activation, a molecular mechanism correlating synaptic protein synthesis and neuroplasticity. As neuroplasticity is also a key mechanism of addiction development, and considering the increasing abuse of KET, our aim was to examine the effect of acute KET administration on the expression of rpS6 in drug addiction-related cerebral areas. We tested in rats the effect of different KET doses (5 or 10 mg/kg, intraperitoneally) on rpS6P expression by immunolocalization in prelimbic (PRL) and infralimbic (IL) cortices, nucleus accumbens core (NAcC) and nucleus accumbens shell (NAcS), hippocampus (CA1 and CA3), and basolateral amygdala (BLA). Expression levels of rpS6 were quantified. A significant dose-related increase in rpS6P expression in PRL, IL, BLA, NAcC but not in the NAcS and hippocampus was found after acute KET. These data confirm acute KET-induced neuroplasticity effects, and extend these findings to drug addiction-related brain areas.


Asunto(s)
Anestésicos Disociativos/farmacología , Encéfalo/efectos de los fármacos , Ketamina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Masculino , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Proteína S6 Ribosómica/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/fisiopatología , Serina-Treonina Quinasas TOR/efectos de los fármacos
7.
Cancer Chemother Pharmacol ; 69(5): 1369-77, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22231376

RESUMEN

PURPOSE: As part of a phase 1 dose-escalation trial, the pharmacodynamic activity of the mammalian target of rapamycin (mTOR) inhibitor ridaforolimus was assessed in multiple tissues by measuring levels of phosphorylated 4E binding protein-1 (p-4E-BP1) or S6, two downstream markers of mTOR activity. METHODS: 32 patients (pts) were dosed intravenously with ridaforolimus once daily for 5 consecutive days (QD × 5) every 2 weeks. The pharmacodynamic activity of ridaforolimus was assessed in peripheral blood mononuclear cells (PBMCs; 32 pts), skin (28 pts), and tumor specimens (3 pts) collected before and after dosing by measuring levels of p-4E-BP1 by immunoblot analysis or pS6 by immunohistochemistry. Levels of these markers were assessed in up to 19, 5, and 2 pre- and post-dose time points in PBMC, skin, and tumor specimens, respectively. RESULTS: In preclinical models, ridaforolimus induced a dose-dependent inhibition of p-4E-BP1 in PBMCs that was associated with antitumor activity. Rapid and potent inhibition of mTOR was observed in PBMCs from all 32 pts dosed, with a median level of inhibition of 96% observed within 1 h after the first dose. Inhibition of mTOR (>90%) was sustained during the entire QD × 5 dosing period, and substantial inhibition was still observed after the 9-day holiday between dosing courses. Evidence of mTOR inhibition was also obtained in skin in pts from all dose cohorts, although it did not persist through the break between courses. After two to three doses of ridaforolimus, inhibition of mTOR was detected in the tumor from one of three pts analyzed. CONCLUSIONS: Ridaforolimus was shown to inhibit its intended target, mTOR, in PBMCs, skin, and tumors. In PBMCs and skin, inhibition was observed at all dose levels tested, thus supporting but not driving the selection of a recommended phase 2 dose.


Asunto(s)
Antineoplásicos/farmacología , Leucocitos Mononucleares/metabolismo , Neoplasias/tratamiento farmacológico , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antineoplásicos/administración & dosificación , Proteínas de Ciclo Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias/patología , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Sirolimus/administración & dosificación , Sirolimus/farmacología , Piel/metabolismo , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Jpn J Infect Dis ; 61(4): 279-85, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18653969

RESUMEN

A procyanidin-rich extract from French maritime pine, Pycnogenol(R) (PYC), is known as an antioxidant that exerts a variety of physiological activities and is widely used in human beings. We report here that PYC inhibits not only human immunodeficiency virus type-1 (HIV-1) binding to host cells, but also its replication after entry in susceptible cells in vitro. Prominent biochemical alterations induced by PYC were the elevated expression of an intracellular antioxidant protein, manganese superoxide dismutase (Mn-SOD), and the inhibition of phosphorylation of the ribosomal S6 protein. Interestingly, ectopic expression of Mn-SOD inhibited HIV-1 replication as well. Inhibition of HIV-1 replication associated with induced expression of Mn-SOD in cells treated with PYC suggests the potential of this natural antioxidant inducer as a new anti-HIV-1 agent.


Asunto(s)
Flavonoides/farmacología , VIH-1/efectos de los fármacos , Pinus/química , Extractos Vegetales/farmacología , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Biflavonoides/análisis , Catequina/análisis , Línea Celular , Flavonoides/química , VIH-1/patogenicidad , VIH-1/fisiología , Humanos , Fosforilación/efectos de los fármacos , Fitoterapia , Extractos Vegetales/química , Proantocianidinas/análisis , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo
9.
J Nutr Sci Vitaminol (Tokyo) ; 54(2): 142-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18490844

RESUMEN

Our recent study demonstrates that polypyrimidine tract-binding protein (PTB), which is a sequence specific RNA-binding protein, attenuates albumin synthesis in a cell-free translation system. In this study, the effects of food intake on regulation of albumin synthesis through binding of PTB to albumin messenger RNA (mRNA) were investigated. Rats were divided into 1 of 3 groups: fed; fasted for 36 h; or fasted for 36 h and then refed for 24 h. No significant differences in albumin mRNA levels were found among fed, fasted and refed rats. However, a decrease in the proportion of albumin mRNA associated with polysomes was identified in fasted rats. Furthermore, UV-cross linking analysis demonstrated that levels of albumin mRNA-PTB complex were increased in liver extracts from fasted rats. No significant differences in PTB levels in liver homogenate were found among the experimental groups. However, PTB level in the cytoplasmic fraction was higher in fasted rats than in fed rats. In refed rats, PTB level in the cytoplasmic fraction returned to a level comparable to that in fed rats, but was inhibited by treatment with rapamycin, a mammalian target of rapamycin (mTOR) inhibitor. These results suggest that localization of PTB is regulated by food intake through mTOR signaling, and alterations in level of albumin mRNA-PTB complex play a role in mediating the effects of food intake on albumin synthesis in the rat liver.


Asunto(s)
Ingestión de Alimentos , Regulación de la Expresión Génica/fisiología , Hígado/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Albúmina Sérica/genética , Animales , Northern Blotting/métodos , Western Blotting/métodos , Citoplasma/metabolismo , Dieta , Ayuno , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/sangre , Hígado/efectos de los fármacos , Masculino , Fosforilación/efectos de los fármacos , Polirribosomas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Albúmina Sérica/biosíntesis , Sirolimus/farmacología
10.
Exp Hematol ; 34(11): 1522-31, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17046572

RESUMEN

OBJECTIVE: Bone marrow stromal cell function is a critical influence on hematopoietic reconstitution following progenitor or stem cell transplantation. Stromal cells support hematopoietic cell migration, survival, and proliferation. We have previously reported that stromal cell matrix metalloproteinase-2 (MMP-2) is necessary for optimal support of pro-B-cell chemotaxis through its regulation of stromal cell-derived factor-1 (CXCL12) release. Following exposure to the topoisomerase II inhibitor, etoposide (VP-16), stromal cell MMP-2 protein expression is reduced. The current study investigated the mechanism by which VP-16 may alter translation of MMP-2 in bone marrow stromal cells. MATERIALS AND METHODS: Bone marrow stromal cells were exposed to chemotherapeutic agents etoposide, melphalan, and 4-hydroperoxycyclophosphamide (4HC) and evaluated for MMP-2 expression by enzyme-linked immunosorbent assay and support of pro-B-cell chemotaxis by chemotaxis assay. Western blot analyses were completed to evaluate phosphorylation of stromal cell translational regulatory proteins 4E binding protein-1 (4EBP-1), P70(S6K), and S6 or MMP-2 in the presence of chemotherapy, or the chemical inhibitors rapamycin or LY294002. RESULTS: Rapid dephosphorylation of 4EBP-1, P70(S6K), and S6 following VP-16 exposure was observed, consistent with blunted translational efficiency. We also observed that inhibition of stromal cell mammalian target of rapamycin with rapamycin, or phosphatidylinositol 3 kinase with LY294002, resulted in inhibition of stromal cell MMP-2 protein. In addition we found that the chemotherapeutic agents melphalan and 4HC disrupt bone marrow stromal cell MMP-2 protein expression and support of chemotaxis. CONCLUSIONS: These data suggest that one mechanism by which chemotherapy may alter stromal cells of the bone marrow microenvironment is through disrupted translation of proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Antimetabolitos Antineoplásicos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Fosfoproteínas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Proteínas de Ciclo Celular , Línea Celular , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Etopósido/farmacología , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/metabolismo , Melfalán/farmacología , Ratones , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína S6 Ribosómica/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/farmacología , Células del Estroma/metabolismo , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR
11.
J Hypertens ; 24(8): 1663-70, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16877971

RESUMEN

BACKGROUND AND OBJECTIVE: Inhibition of established left ventricular hypertrophy (LVH) and fibrosis may bring clinical benefits by reducing cardiac morbidity and mortality. The mammalian target of rapamycin, mTOR, is known to play a critical role in determining cell and organ size. We investigated whether mTOR inhibition can inhibit the chronic pressure-overload-induced LVH and fibrosis. METHODS: Male FVB/N mice underwent transverse aortic constriction (TAC) for 5 weeks to allow for establishment of LVH, followed by treatment with the mTOR inhibitor, Rapamune (2 mg/kg per day, gavage), for 4 weeks. Echocardiography was used to monitor changes in LVH and function. Haemodynamic, morphometric, histological and molecular analyses were conducted. RESULTS: Inhibition of mTOR by Rapamune was confirmed by a suppression of activated phosphorylation of ribosomal S6 protein and eukaryotic translation initiation factor-4E due to pressure overload. Despite a comparable degree of pressure overload between the vehicle- or Rapamune-treated TAC groups, Rapamune treatment for 4 weeks attenuated TAC-induced LVH by 46%, estimated by LV weight or myocyte size, and LV fractional shortening was also preserved versus vehicle-treated control (39 +/- 1 versus 32 +/- 2%, P < 0.05). Inhibition of established LVH by Rapamune was associated with a 38% reduction in collagen content. Moreover, altered gene expression due to pressure overload was largely restored. CONCLUSION: Despite sustained pressure overload, inhibition of mTOR by a 4-week period of Rapamune treatment attenuates chronically established LVH and cardiac fibrosis with preserved contractile function.


Asunto(s)
Presión Sanguínea , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Proteínas Quinasas/metabolismo , Análisis de Varianza , Animales , Factor Natriurético Atrial/efectos de los fármacos , Factor Natriurético Atrial/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/metabolismo , Fibrosis , Frecuencia Cardíaca , Inmunosupresores/farmacología , Masculino , Ratones , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Cadenas Pesadas de Miosina/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Volumen Sistólico , Serina-Treonina Quinasas TOR
12.
Biochemistry ; 45(7): 2189-99, 2006 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-16475807

RESUMEN

The effect of osmolyte sucrose on the stability and compaction of the folded and unfolded states of ribosomal protein S6 from Thermus thermophilus was analyzed. Confirming previous results obtained with sodium sulfate and trehalose, refolding stopped-flow measurements of S6 show that sucrose favors the conversion of the unfolded state ensemble to a highly compact structure (75% as compact as the folded state). This conversion occurs when the unfolded state is suddenly placed under native conditions and the compact state accumulates in a transient off-folding pathway. This effect of sucrose on the compaction of the unfolded state ensemble is counteracted by guanidinium hydrochloride. The compact state does not accumulate at higher guanidinium concentrations and the unfolded state ensemble does not display increased compaction in the presence of 6 M guanidinium as evaluated by collisional quenching of tryptophan fluorescence. In contrast, accessibility of the tryptophan residue of folded S6 above 1 M sucrose concentration decreased as a result of an increased compaction of the folded state. Unfolding stopped-flow measurements of S6 reflect this increased compaction of the folded state, but the unfolding pathway is not affected by sucrose. Compaction of folded and unfolded S6 induced by sucrose occurs under native conditions indicating that decreased protein conformational entropy significantly contributes to the mechanism of protein stabilization by osmolytes.


Asunto(s)
Pliegue de Proteína , Proteína S6 Ribosómica/química , Sacarosa/farmacología , Guanidina/farmacología , Cinética , Desnaturalización Proteica , Proteína S6 Ribosómica/efectos de los fármacos , Espectrometría de Fluorescencia , Termodinámica , Thermus thermophilus/química
13.
J Nutr ; 136(1): 27-33, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16365054

RESUMEN

We examined the role of the mammalian target of rapamycin (mTOR) in hepatic cell growth. To dissociate cell growth from cell proliferation, we employed an in vivo model of nonproliferative liver growth in rats, refeeding after 48 h of food deprivation. Starvation resulted in a decrease in liver mass, liver protein, and cell size, all of which were largely restored after 24 h of refeeding. Administration of the mTOR inhibitor, rapamycin, before the refeeding period partially inhibited the restoration of liver protein content. Refeeding was also associated with an increase in ribosomal protein S6 phosphorylation and phosphorylation of the eukaryotic initiation factor (eIF) 4E binding protein 1 (4E-BP1). 4E-BP1 phosphorylation was accompanied by a decrease in the abundance of the complex containing 4E-BP1 with eIF4E. These changes were prevented by rapamycin administration. However, association of eIF4E and eIF4G and eIF2alpha phosphorylation, both of which are stimulated by refeeding, were insensitive to rapamycin. The functional importance of these observations was confirmed by polysome fractionation, which showed that translation initiation of 5' oligopyrimidine tract-containing mRNAs, which encode ribosomal proteins, was inhibited by rapamycin, whereas translation of signal transducer and activator of transcription 1 (STAT1), a cap-dependent mRNA, was unaffected. The abundance of ribosomal proteins paralleled total protein content during refeeding in both control and rapamycin-injected rats. We conclude that accretion of liver protein during refeeding is dependent on mTOR-mediated activation of the translation of ribosomal proteins but not dependent on mTOR-mediated activation of cap-dependent translation initiation.


Asunto(s)
Inmunosupresores/farmacología , Hígado/efectos de los fármacos , Proteínas Quinasas/efectos de los fármacos , Proteína S6 Ribosómica/efectos de los fármacos , Sirolimus/farmacología , Animales , Ingestión de Alimentos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Privación de Alimentos , Hígado/crecimiento & desarrollo , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteína S6 Ribosómica/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR
14.
Am J Physiol Cell Physiol ; 287(5): C1482-92, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15229102

RESUMEN

Anti-retroviral therapy promotes clinical, immunologic, and virologic improvement in human immunodeficiency virus-infected patients. Whereas this therapy adversely affects carbohydrate and lipid metabolism, the effects of anti-retroviral drugs on muscle protein synthesis and degradation have not been reported. To examine these processes, we treated C2C12 myocytes with increasing concentrations of the protease inhibitor indinavir for 1 or 2 days. Treatment of myocytes with a therapeutic concentration of indinavir (20 microM) for 24 h decreased basal protein synthesis by 18%, whereas a 42% decline was observed after 48 h. A similar decrement, albeit quantitatively smaller, was detected with other protease inhibitors. Indinavir did not alter the rate of proteolysis. Likewise, indinavir did not impair the anabolic effect of insulin-like growth factor-I on protein synthesis. Mechanistically, indinavir decreased the phosphorylation of the S6 ribosomal protein (rpS6), and this reduction was associated with a decreased phosphorylation of p70S6 kinase and p90rsk as well as the upstream regulators ERK1/2 and MEK1/2. Indinavir also decreased the phosphorylation of Mnk1 and its upstream effectors, p38 MAPK and ERK1/2. Indinavir did not affect the phosphorylation of mTOR or 4E-BP1, but it did decrease the amount of the active eukaryotic initiation factor eIF4G-eIF4E complex. In conclusion, indinavir decreased protein synthesis in myocytes. This decrease was associated with the disruption of the ERK1/2 and p38 MAPK pathways and a reduction in both the level of functional eIF4F complex and rpS6 phosphorylation.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , Indinavir/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Células Musculares/efectos de los fármacos , Animales , División Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Factor 4F Eucariótico de Iniciación/efectos de los fármacos , Factor 4F Eucariótico de Iniciación/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
15.
Mutagenesis ; 18(5): 405-10, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12960407

RESUMEN

It has been shown that the ethylating agent diethylsulphate (DES) induces centromere-containing micronuclei with kinetics suggesting that molecules other than DNA could be targets. In quiescent Chinese hamster fibroblasts CHEF/18, O6-alkylated bases inhibit ribosomal protein S6 kinase (S6K1), the terminal member of a kinase cascade responsible for an increased rate of protein synthesis, but not extracellular signal-activated kinases (ERK1/2) or terminal kinases of a second cascade which activates transcription. The inhibition correlates with the appearance of abnormal metaphases at the following mitosis, suggesting that alkylation of the nucleotide pool and inhibition of S6K1 could be one of the mechanisms leading to chromosome loss by alkylating agents. To clarify the role of protein kinases in chromosome loss induced by alkylating agents, we have studied the effects of DES and methylnitrosourea (MNU) on S6K1 and ERK1/2 activation by growth factors. The alkylating agents were studied in a battery of Chinese hamster fibroblasts (CHEF/18, CHO and ClB) with normal and mutated p53 to control for DNA damage-induced activation of p53, which could indirectly inhibit protein kinases. The role of repair in induction of micronuclei was studied in mismatch repair-proficient CHO and repair-deficient ClB cells. Our results indicate that DES induced micronuclei in a mismatch repair-independent manner, within 8 h of treatment, in agreement with a role for S6K1 inhibition in micronucleus formation. MNU induced centromere-containing micronuclei only in CHO cells, one cell cycle after treatment, without any detectable influences on either kinase cascade, suggesting a role for mismatch repair in chromosome loss.


Asunto(s)
Alquilantes/toxicidad , Aneugénicos/toxicidad , Metilnitrosourea/toxicidad , Ésteres del Ácido Sulfúrico/toxicidad , Aneuploidia , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Reparación del ADN/genética , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Pruebas de Micronúcleos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Mutación , Fosforilación , Proteína S6 Ribosómica/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Proteínas Quinasas S6 Ribosómicas/efectos de los fármacos , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...