Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 488(7411): 394-8, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22810587

RESUMEN

Cardiac hypertrophy is initiated as an adaptive response to sustained overload but progresses pathologically as heart failure ensues. Here we report that genetic loss of APJ, a G-protein-coupled receptor, confers resistance to chronic pressure overload by markedly reducing myocardial hypertrophy and heart failure. In contrast, mice lacking apelin (the endogenous APJ ligand) remain sensitive, suggesting an apelin-independent function of APJ. Freshly isolated APJ-null cardiomyocytes exhibit an attenuated response to stretch, indicating that APJ is a mechanosensor. Activation of APJ by stretch increases cardiomyocyte cell size and induces molecular markers of hypertrophy. Whereas apelin stimulates APJ to activate Gαi and elicits a protective response, stretch signals in an APJ-dependent, G-protein-independent fashion to induce hypertrophy. Stretch-mediated hypertrophy is prevented by knockdown of ß-arrestins or by pharmacological doses of apelin acting through Gαi. Taken together, our data indicate that APJ is a bifunctional receptor for both mechanical stretch and the endogenous peptide apelin. By sensing the balance between these stimuli, APJ occupies a pivotal point linking sustained overload to cardiomyocyte hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipoquinas , Animales , Aorta/patología , Apelina , Receptores de Apelina , Arrestinas/deficiencia , Arrestinas/genética , Arrestinas/metabolismo , Presión Sanguínea , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Cardiomegalia/prevención & control , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Mecanorreceptores/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , beta-Arrestinas
2.
Proc Natl Acad Sci U S A ; 110(9): 3333-8, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23341587

RESUMEN

Circadian rhythms govern a wide variety of physiological and metabolic functions in many organisms, from prokaryotes to humans. We previously reported that silent information regulator 1 (SIRT1), a NAD(+)-dependent deacetylase, contributes to circadian control. In addition, SIRT1 activity is regulated in a cyclic manner in virtue of the circadian oscillation of the coenzyme NAD(+). Here we used specific SIRT1 activator compounds both in vitro and in vivo. We tested a variety of compounds to show that the activation of SIRT1 alters CLOCK:BMAL1-driven transcription in different systems. Activation of SIRT1 induces repression of circadian gene expression and decreases H3 K9/K14 acetylation at corresponding promoters in a time-specific manner. Specific activation of SIRT1 was demonstrated in vivo using liver-specific SIRT1-deficient mice, where the effect of SIRT1 activator compounds was shown to be dependent on SIRT1. Our findings demonstrate that SIRT1 can fine-tune circadian rhythm and pave the way to the development of pharmacological strategies to address a broad range of therapeutic indications.


Asunto(s)
Ritmo Circadiano/genética , Activadores de Enzimas/farmacología , Sirtuina 1/metabolismo , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Línea Celular , Cromatina/metabolismo , Ritmo Circadiano/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Noqueados , NAD/metabolismo , Transcripción Genética/efectos de los fármacos
3.
Crit Care Med ; 43(6): e199-202, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25978169

RESUMEN

OBJECTIVES: Sirtuin 1 influences gene expression and other cellular functions through deacetylation of histone and nonhistone proteins. We here sought to determine the effects of a small molecule sirtuin 1 activator, SRT2104, on inflammation and coagulation induced by lipopolysaccharide in humans. DESIGN: A randomized, double-blind, placebo-controlled study. SETTING: An academic hospital. SUBJECTS: Twenty-four healthy humans. INTERVENTIONS: All subjects received an intravenous injection with lipopolysaccharide. Subjects were randomized to one of three groups (n=8 per group): 1) pretreatment with oral SRT2104 for 7 days (2 g/d), 2) pretreatment with a single SRT2104 dose (2 g), or 3) placebo. MEASUREMENTS AND MAIN RESULTS: SRT2104 attenuated lipopolysaccharide-induced release of the cytokines interleukin-6 (mean peak levels of 58.8% [p<0.05] and 80.9% [p=0.078] after single and repeated SRT2104 administration, respectively, relative to those measured after placebo treatment) and interleukin-8 (mean peak levels of 57.0% [p<0.05 vs placebo] and 77.1% [p<0.05 vs placebo] after single and repeated SRT2104 ingestion, respectively, while not affecting tumor necrosis factor-α and interleukin-10 release). SRT2104 also reduced the lipopolysaccharide-induced acute phase protein response (C-reactive protein). SRT2104 inhibited activation of coagulation, as reflected by lower plasma levels of the prothrombin fragment F1+2 (mean peak levels 57.9% [p<0.05] and 64.2% [p<0.05] after single and repeated SRT2104 administration, respectively, relative to those measured after placebo treatment). Activation of the vascular endothelium (plasma von Willebrand levels) and the fibrinolytic system (plasma tissue-type plasminogen activator and plasminogen activator inhibitor type I) was not influenced by SRT2104. CONCLUSIONS: This is the first human study to demonstrate biological anti-inflammatory and anticoagulant responses consistent with the activation of sirtuin 1 by a small molecule.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Endotoxinas/metabolismo , Compuestos Heterocíclicos con 2 Anillos/farmacología , Lipopolisacáridos/farmacología , Sirtuina 1/biosíntesis , Método Doble Ciego , Humanos , Masculino , Adulto Joven
4.
Br J Clin Pharmacol ; 78(1): 69-77, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24446723

RESUMEN

AIM: SRT2104 is a selective activator of SIRT1. In animal models, SRT2104 improves glucose homeostasis and increases insulin sensitivity. We evaluated the tolerability and pharmacokinetics of SRT2104, and its effects on glycaemic control, in adults with type 2 diabetes mellitus. METHOD: Type 2 diabetics with glycosylated haemoglobin (HbA1c) ≥ 7.5% and ≤10.5%, fasting glucose ≥160 and ≤240 mg dl(-1) , and on stable doses of metformin were evenly randomized to placebo or SRT2104 0.25 g, 0.5 g, 1.0 g or 2.0 g, administered orally once daily for 28 days. Changes in fasting and post-prandial glucose and insulin were analyzed. RESULTS: Safety evaluation found no major differences between groups in the frequency of adverse events. SRT2104 concentrations did not increase in a dose-proportional fashion. Significant variability in exposure was observed. Treatment with SRT2104 did not lead to any consistent, dose-related changes in glucose or insulin. Day 28 change from baseline (mean (SD)): fasting glucose (mmol l(-1) ) = -1.17 (2.42), -1.11 (3.45), -0.52 (2.60), -0.97 (2.83) and -0.15 (2.38) for placebo, 0.25 g, 0.5 g, 1.0 g and 2.0 g, respectively. Day 28 change from baseline (mean (SD)): fasting insulin (mmol l(-1) ) = 1.0 (51.66), 8.9 (95.04), -6.9 (41.45), 4.1 (57.16) and 15.2 (138.79) for placebo, 0.25 g, 0.5 g, 1.0 g and 2.0 g, respectively) Treatment with SRT2104 was associated with improvement in lipid profiles. CONCLUSION: Treatment with SRT2104 for 28 days did not result in improved glucose or insulin control which is likely due to the observed pharmacokinetics which were not dose proportional and had large between subject variability.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Activadores de Enzimas/uso terapéutico , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Sirtuina 1/metabolismo , Adulto , Anciano , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Quimioterapia Combinada , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/efectos adversos , Activadores de Enzimas/farmacocinética , Activadores de Enzimas/farmacología , Femenino , Compuestos Heterocíclicos con 2 Anillos/efectos adversos , Compuestos Heterocíclicos con 2 Anillos/farmacocinética , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Insulina/sangre , Masculino , Metformina/uso terapéutico , Persona de Mediana Edad
5.
Proc Natl Acad Sci U S A ; 106(1): 262-7, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19116277

RESUMEN

In a search for more effective anti-diabetic treatment, we used a process coupling low-affinity biochemical screening with high-throughput co-crystallography in the design of a series of compounds that selectively modulate the activities of all three peroxisome proliferator-activated receptors (PPARs), PPARalpha, PPARgamma, and PPARdelta. Transcriptional transactivation assays were used to select compounds from this chemical series with a bias toward partial agonism toward PPARgamma, to circumvent the clinically observed side effects of full PPARgamma agonists. Co-crystallographic characterization of the lead molecule, indeglitazar, in complex with each of the 3 PPARs revealed the structural basis for its PPAR pan-activity and its partial agonistic response toward PPARgamma. Compared with full PPARgamma-agonists, indeglitazar is less potent in promoting adipocyte differentiation and only partially effective in stimulating adiponectin gene expression. Evaluation of the compound in vivo confirmed the reduced adiponectin response in animal models of obesity and diabetes while revealing strong beneficial effects on glucose, triglycerides, cholesterol, body weight, and other metabolic parameters. Indeglitazar has now progressed to Phase II clinical evaluations for Type 2 diabetes mellitus (T2DM).


Asunto(s)
Descubrimiento de Drogas/métodos , Hipoglucemiantes/uso terapéutico , PPAR gamma/agonistas , Receptores Activados del Proliferador del Peroxisoma/agonistas , Adipocitos/citología , Adiponectina/genética , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Humanos , Hipoglucemiantes/farmacología , Ratones , Obesidad/tratamiento farmacológico , PPAR gamma/genética , Receptores Activados del Proliferador del Peroxisoma/genética , Ratas , Activación Transcripcional/efectos de los fármacos
6.
J Biol Chem ; 285(43): 32695-32703, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20702418

RESUMEN

SIRT1 is a protein deacetylase that has emerged as a therapeutic target for the development of activators to treat diseases of aging. SIRT1-activating compounds (STACs) have been developed that produce biological effects consistent with direct SIRT1 activation. At the molecular level, the mechanism by which STACs activate SIRT1 remains elusive. In the studies reported herein, the mechanism of SIRT1 activation is examined using representative compounds chosen from a collection of STACs. These studies reveal that activation of SIRT1 by STACs is strongly dependent on structural features of the peptide substrate. Significantly, and in contrast to studies reporting that peptides must bear a fluorophore for their deacetylation to be accelerated, we find that some STACs can accelerate the SIRT1-catalyzed deacetylation of specific unlabeled peptides composed only of natural amino acids. These results, together with others of this study, are at odds with a recent claim that complex formation between STACs and fluorophore-labeled peptides plays a role in the activation of SIRT1 (Pacholec, M., Chrunyk, B., Cunningham, D., Flynn, D., Griffith, D., Griffor, M., Loulakis, P., Pabst, B., Qiu, X., Stockman, B., Thanabal, V., Varghese, A., Ward, J., Withka, J., and Ahn, K. (2010) J. Biol. Chem. 285, 8340-8351). Rather, the data suggest that STACs interact directly with SIRT1 and activate SIRT1-catalyzed deacetylation through an allosteric mechanism.


Asunto(s)
Activadores de Enzimas/química , Péptidos/química , Sirtuina 1/química , Activación Enzimática , Humanos , Especificidad por Sustrato
7.
Proc Natl Acad Sci U S A ; 105(25): 8754-9, 2008 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-18559859

RESUMEN

The amyloid hypothesis states that a variety of neurotoxic beta-amyloid (Abeta) species contribute to the pathogenesis of Alzheimer's disease. Accordingly, a key determinant of disease onset and progression is the appropriate balance between Abeta production and clearance. Enzymes responsible for the degradation of Abeta are not well understood, and, thus far, it has not been possible to enhance Abeta catabolism by pharmacological manipulation. We provide evidence that Abeta catabolism is increased after inhibition of plasminogen activator inhibitor-1 (PAI-1) and may constitute a viable therapeutic approach for lowering brain Abeta levels. PAI-1 inhibits the activity of tissue plasminogen activator (tPA), an enzyme that cleaves plasminogen to generate plasmin, a protease that degrades Abeta oligomers and monomers. Because tPA, plasminogen and PAI-1 are expressed in the brain, we tested the hypothesis that inhibitors of PAI-1 will enhance the proteolytic clearance of brain Abeta. Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Abeta levels, restore long-term potentiation deficits in hippocampal slices from transgenic Abeta-producing mice, and reverse cognitive deficits in these mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fibrinolisina/metabolismo , Fibrinolíticos/metabolismo , Animales , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Inactivadores Plasminogénicos/metabolismo , Activador de Tejido Plasminógeno/antagonistas & inhibidores , Activador de Tejido Plasminógeno/metabolismo
8.
J Infect Dis ; 201(8): 1250-7, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20205571

RESUMEN

BACKGROUND: Nonsteroidal agonists have been developed that selectively bind to and activate estrogen receptor beta (ERbeta) rather than estrogen receptor alpha (ERalpha). ERbeta is expressed equally in both male and female mammals in multiple extragonadal tissues. Work reported elsewhere has demonstrated that ERbeta agonists have beneficial effects in multiple (but not all) models of inflammatory diseases and also increase survival in experimentally induced sepsis. METHODS: In these experiments, ERbeta agonists (ERB-041 or WAY-202196) were compared with vehicle control in the murine cecal ligation and puncture (CLP) model and in the pneumococcal pneumonia model of sepsis. The effect of WAY-202196 on the gene expression profile in the CLP model was further studied by transcriptome analysis of lung and small intestine tissue samples. RESULTS: ERbeta agonists provided a significant survival benefit in both experimental models of bacterial sepsis. This survival advantage was accompanied by reduced histologic evidence of tissue damage, reduced transcription of multiple proinflammatory proteins by transcriptome analysis and was not associated with increased bacterial outgrowth. CONCLUSIONS: ERbeta agonist administration provided a survival advantage in septic animals and appears to be a promising therapeutic modality in sepsis.


Asunto(s)
Receptor beta de Estrógeno/agonistas , Naftoles/uso terapéutico , Oxazoles/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Receptor beta de Estrógeno/biosíntesis , Receptor beta de Estrógeno/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos BALB C , Neumonía Neumocócica/tratamiento farmacológico , Sepsis/fisiopatología , Transcripción Genética/efectos de los fármacos
9.
J Lipid Res ; 50(12): 2358-70, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19318684

RESUMEN

Liver X receptors (LXRs) are ligand-activated transcription factors that coordinate regulation of gene expression involved in several cellular functions but most notably cholesterol homeostasis encompassing cholesterol transport, catabolism, and absorption. WAY-252623 (LXR-623) is a highly selective and orally bioavailable synthetic modulator of LXR, which demonstrated efficacy for reducing lesion progression in the murine LDLR(-/-) atherosclerosis model with no associated increase in hepatic lipogenesis either in this model or Syrian hamsters. In nonhuman primates with normal lipid levels, WAY-252623 significantly reduced total (50-55%) and LDL-cholesterol (LDLc) (70-77%) in a time- and dose-dependent manner as well as increased expression of the target genes ABCA1/G1 in peripheral blood cells. Statistically significant decreases in LDLc were noted as early as day 7, reached a maximum by day 28, and exceeded reductions observed for simvastatin alone (20 mg/kg). Transient increases in circulating triglycerides and liver enzymes reverted to baseline levels over the course of the study. Complementary microarray analysis of duodenum and liver gene expression revealed differential activation of LXR target genes and suggested no direct activation of hepatic lipogenesis. WAY-252623 displays a unique and favorable pharmacological profile suggesting synthetic LXR ligands with these characteristics may be suitable for evaluation in patients with atherosclerotic dyslipidemia.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , LDL-Colesterol/efectos de los fármacos , LDL-Colesterol/metabolismo , Indazoles/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Macaca fascicularis/metabolismo , Receptores Nucleares Huérfanos/agonistas , Animales , Aterosclerosis/metabolismo , Células CACO-2 , Cricetinae , Modelos Animales de Enfermedad , Humanos , Indazoles/sangre , Indazoles/química , Ligandos , Hígado/enzimología , Hígado/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/metabolismo
10.
J Cardiovasc Pharmacol Ther ; 14(3): 207-14, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19721133

RESUMEN

The antiarrhythmic dipeptide, GAP-134, ([2S,4R]-1[2-aminoacetyl]-4-benzamido-pyrrolidine-2-carboxylic acid) was evaluated in canine ischemia/reperfusion model. In dogs subjected to 60-minute ischemia and 4-hour reperfusion, GAP-134 was administered 10 minutes before reperfusion as a bolus + intravenous (IV) infusion. The doses administered were 0.25 microg/kg bolus + 0.19 microg/kg per hour infusion; 2.5 microg/kg + 1.9 microg/kg per hour; 25 mg/kg + 19 mg/kg per hour; 75 mg/kg + 57 mg/kg per hour. Ventricular ectopy was quantified during reperfusion, including premature ventricular contractions (PVC) and ventricular tachycardia (VT). Total incidence of VT was reduced significantly with the 2 highest doses of GAP-134 (1.7 + 0.8; 2.2 + 1.4 events; P < .05) compared to controls (23.0 + 6.1). Total PVCs were reduced significantly from 11.1 + 1.6% in control animals to 2.0% + 0.7% and 1.8% + 0.8% after the 2 highest doses of GAP-134. Infarct size, expressed as percentage of left ventricle, was reduced significantly from 19.0% + 3.5% in controls to 7.9% + 1.5% and 7.1% + 0.8% (P < .05) at the 2 highest doses of GAP-134. GAP-134 is an effective antiarrhythmic agent with potential to reduce ischemia/reperfusion injury.


Asunto(s)
Antiarrítmicos/farmacología , Benzamidas/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio/patología , Prolina/análogos & derivados , Taquicardia Ventricular/prevención & control , Complejos Prematuros Ventriculares/prevención & control , Animales , Antiarrítmicos/administración & dosificación , Benzamidas/administración & dosificación , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Infusiones Intravenosas , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Prolina/administración & dosificación , Prolina/farmacología , Taquicardia Ventricular/etiología , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/patología , Complejos Prematuros Ventriculares/fisiopatología
11.
Sci Rep ; 9(1): 4107, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30858438

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) has been linked to several important chronic medical conditions many of which are associated with advancing age. A variety of inputs including the amino acid leucine are required for full mTORC1 activation. The cytoplasmic proteins Sestrin1 and Sestrin2 specifically bind to the multiprotein complex GATOR2 and communicate leucine sufficiency to the mTORC1 pathway activation complex. Herein, we report NV-5138, a novel orally bioavailable compound that binds to Sestrin2 and activates mTORC1 both in vitro and in vivo. NV-5138 like leucine transiently activates mTORC1 in several peripheral tissues, but in contrast to leucine uniquely activates this complex in the brain due lack of metabolism and utilization in protein synthesis. As such, NV-5138 will permit the exploration in areas of unmet medical need including neuropsychiatric conditions and cognition which have been linked to the activation status of mTORC1.


Asunto(s)
Encéfalo/metabolismo , Descubrimiento de Drogas , Leucina/análogos & derivados , Leucina/farmacocinética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Administración Oral , Animales , Diseño de Fármacos , Células HEK293 , Humanos , Leucina/administración & dosificación , Masculino , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transaminasas/metabolismo
12.
J Clin Invest ; 129(6): 2542-2554, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30990795

RESUMEN

Preclinical studies demonstrate that rapid acting antidepressants, including ketamine require stimulation of mTORC1 signaling. This pathway is regulated by neuronal activity, endocrine and metabolic signals, notably the amino acid leucine, which activates mTORC1 signaling via binding to the upstream regulator sestrin. Here, we examined the antidepressant actions of NV-5138, a novel highly selective small molecule modulator of sestrin that penetrates the blood brain barrier. The results demonstrate that a single dose of NV-5138 produced rapid and long-lasting antidepressant effects, and rapidly reversed anhedonia caused by chronic stress exposure. The antidepressant actions of NV-5138 required BDNF release as the behavioral responses are blocked by infusion of a BDNF neutralizing antibody into the medial prefrontal cortex (mPFC) or in mice with a knock-in of a BDNF polymorphism that blocks activity dependent BDNF release. NV-5138 administration also rapidly increased synapse number and function in the mPFC, and reversed the synaptic deficits caused by chronic stress. Together, the results demonstrate that NV-5138 produced rapid synaptic and antidepressant behavioral responses via activation of the mTORC1 pathway and BDNF signaling, indicating that pharmacological modulation of sestrin is a novel approach for development of rapid acting antidepressants.


Asunto(s)
Antidepresivos , Conducta Animal/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transmisión Sináptica/efectos de los fármacos , Animales , Antidepresivos/química , Antidepresivos/farmacocinética , Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de Choque Térmico/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/genética
13.
Cell Chem Biol ; 26(9): 1203-1213.e13, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31231029

RESUMEN

The mechanistic target of rapamycin (mTOR) is a central regulator of cellular metabolic processes. Dysregulation of this kinase complex can result in a variety of human diseases. Rapamycin and its analogs target mTORC1 directly; however, chronic treatment in certain cell types and in vivo results in the inhibition of both mTORC1 and mTORC2. We have developed a high-throughput cell-based screen for the detection of phosphorylated forms of the mTORC1 (4E-BP1, S6K1) and mTORC2 (Akt) substrates and have identified and characterized a chemical scaffold that demonstrates a profile consistent with the selective inhibition of mTORC1. Stable isotope labeling of amino acids in cell culture-based proteomic target identification revealed that class I glucose transporters were the primary target for these compounds yielding potent inhibition of glucose uptake and, as a result, selective inhibition of mTORC1. The link between the glucose uptake and selective mTORC1 inhibition are discussed in the context of a yet-to-be discovered glucose sensor.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Sirolimus/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Glucosa/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina/efectos de los fármacos , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/metabolismo , Fosforilación , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Sirolimus/metabolismo , Factores de Transcripción/metabolismo
14.
Thromb Haemost ; 99(2): 343-51, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18278184

RESUMEN

P-selectin inhibition has been evaluated as a therapeutic for prevention and treatment of venous thrombosis. In this study, a novel oral small-molecule inhibitor of P-selectin, PSI-421, was evaluated in a baboon model of stasis induced deep vein thrombosis (DVT). Experimental groups included i) primates receiving a single oral dose of 1 mg/kg PSI-421 two days prior and continued six days after thrombosis (n = 3); ii) primates receiving a single daily subcutaneous dose of 0.57 mg/kg enoxaparin sodium two days prior and continued six days post thrombosis (n = 3); and iii) primates receiving no treatment (n = 3). PSI-421 treated primates had greater percent vein reopening and less vein wall inflammation than the enoxaparin and controls at day 6. Microparticle tissue factor activity (MPTFA) was significantly lower in the animals receiving PSI-421 immediately after thrombosis (T+6 hours day 0) suggesting lower potential for thrombogenesis in these animals. PSI-421 also reduced soluble P-selectin levels versus controls at T+6 hours day 0, day 2 and 6. Experimental animals in any group showed no adverse effects on coagulation. This study is the first to demonstrate a reduction in MPTFA associated with vein reopening and reduced vein inflammation due to oral P-selectin inhibition in a baboon model of DVT.


Asunto(s)
Anticoagulantes/farmacología , Enoxaparina/farmacología , Fibrinolíticos/farmacología , Hidroxiquinolinas/farmacología , Vena Ilíaca/efectos de los fármacos , Selectina-P/efectos de los fármacos , Trombosis de la Vena/prevención & control , Administración Oral , Animales , Anticoagulantes/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Modelos Animales de Enfermedad , Enoxaparina/administración & dosificación , Inhibidores del Factor Xa , Fibrinolíticos/administración & dosificación , Fibrinolíticos/sangre , Hidroxiquinolinas/administración & dosificación , Hidroxiquinolinas/sangre , Vena Ilíaca/metabolismo , Vena Ilíaca/patología , Vena Ilíaca/fisiopatología , Inflamación/metabolismo , Inflamación/prevención & control , Inyecciones Subcutáneas , Masculino , Selectina-P/metabolismo , Papio anubis , Flebografía , Tromboplastina/metabolismo , Factores de Tiempo , Ultrasonografía Doppler en Color , Grado de Desobstrucción Vascular , Trombosis de la Vena/sangre , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología , Trombosis de la Vena/fisiopatología
15.
Arterioscler Thromb Vasc Biol ; 27(9): 1895-900, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17600222

RESUMEN

The formation of the proteolytic complex composed of the serine protease Factor VIIa and the cell-associated glycoprotein tissue factor (FVIIa/TF) initiates a cascade of amplified zymogen activation reactions leading to thrombus formation. The critical role of the coagulation cascade in pathological thrombosis has been the basis for significant efforts to design selective inhibitors of the protease components as new anticoagulant alternatives for the treatment of thrombotic diseases. However, for the new generation of anticoagulant drugs in development that primarily target protease complexes distal from FVIIa/TF, the differential between efficacy and safety as defined by bleeding is unresolved. Targeting the FVIIa/TF complex has several theoretical advantages that exploit the amplified nature of the coagulation cascade. However, progress on the development of clinical-stage FVIIa/TF-based anticoagulants has not been as successful to date. This review summarizes recent efforts in the discovery of synthetic inhibitors of FVIIa/TF.


Asunto(s)
Diseño de Fármacos , Factor VIIa/antagonistas & inhibidores , Tromboplastina/antagonistas & inhibidores , Anticoagulantes/química , Anticoagulantes/farmacología , Humanos , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología
16.
Nat Commun ; 9(1): 548, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416044

RESUMEN

The small G-protein Rheb activates the mechanistic target of rapamycin complex 1 (mTORC1) in response to growth factor signals. mTORC1 is a master regulator of cellular growth and metabolism; aberrant mTORC1 signaling is associated with fibrotic, metabolic, and neurodegenerative diseases, cancers, and rare disorders. Point mutations in the Rheb switch II domain impair its ability to activate mTORC1. Here, we report the discovery of a small molecule (NR1) that binds Rheb in the switch II domain and selectively blocks mTORC1 signaling. NR1 potently inhibits mTORC1 driven phosphorylation of ribosomal protein S6 kinase beta-1 (S6K1) but does not inhibit phosphorylation of AKT or ERK. In contrast to rapamycin, NR1 does not cause inhibition of mTORC2 upon prolonged treatment. Furthermore, NR1 potently and selectively inhibits mTORC1 in mouse kidney and muscle in vivo. The data presented herein suggest that pharmacological inhibition of Rheb is an effective approach for selective inhibition of mTORC1 with therapeutic potential.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular Tumoral , Cristalografía por Rayos X , Células HEK293 , Humanos , Células Jurkat , Células MCF-7 , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Fosforilación/efectos de los fármacos , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Bibliotecas de Moléculas Pequeñas/química
17.
Thromb Haemost ; 95(3): 469-75, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16525575

RESUMEN

Under high shear arterial blood flow von Willebrand Factor (vWF) binds the platelet receptor glycoprotein (GP) Ibalpha, leading to platelet adhesion, activation and thrombosis. Blockade of vWF-GPIb alpha interactions by GPG-290 was investigated in a canine model of coronary artery thrombosis alone and in combination with clopidogrel. GPG-290 (100 microg/kg, n=6; 500 microg/kg, n=6) prolonged time to thrombotic occlusion (TTO) to 105+/-34 and 156+/-23 (p<0.05) min, respectively compared to the saline treated control group (32+/-6 min, n=6). Patency of the injured vessel was sustained in 1/6 (100 microg/kg) and 3/6 vessels (500 microg/kg) 4 hours after injury, in contrast to 0/6 in the control group. There was an increase in bleeding after the 500 microg/kg dose, but only at the 1 hr time point. Clopidogrel was studied in two dosing regimens representing either a clinical pretreatment regimen (PTR) of 4.3 mg/kg on day -2 followed by 1.1 mg/kg daily for 2 days prior to the procedure or pre-procedural loading dose regimen (LDR) of 4.3 mg/kg 3 hr pre-procedure. The PTR and LDR clopidogrel treatments prolonged TTO to 98.2+/-30.0 min and 136.1+/-39.5 min (p<0.05), and sustained patency in 1/6 and 4/8 vessels, respectively. However, template bleeding time in the LDR clopidogrel group was sustained higher than the control group. The combination of PTR clopidogrel and GPG-290 (100 microg/kg) prolonged TTO equivalent to LDR clopidogrel alone (141.4 +/- 35.1 min) and sustained patency in 3/7 dogs, without increased bleeding while LDR clopidogrel combined with 100 microg/kg GPG-290 prevented occlusion in 5/8 dogs and further prolonged TTO (173.5+/-32.6 min) but was associated with increased bleeding compared to control. GPG-290 is an antithrombotic agent that may be combined with lower doses of clopidogrel to yield similar antithrombotic efficacy as higher loading doses.


Asunto(s)
Trombosis Coronaria/prevención & control , Fibrinolíticos/farmacología , Complejo GPIb-IX de Glicoproteína Plaquetaria/farmacología , Animales , Tiempo de Sangría , Clopidogrel , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Eptifibatida , Fibrinolíticos/uso terapéutico , Péptidos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/uso terapéutico , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Ticlopidina/uso terapéutico , Factores de Tiempo , Grado de Desobstrucción Vascular/efectos de los fármacos , Factor de von Willebrand/antagonistas & inhibidores
18.
Cancer Res ; 63(11): 2997-3000, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12782609

RESUMEN

An association between cancer and thrombosis has been recognized for more than a century. However, the manner by which tumor growth is regulated by coagulation in vivo remains unclear. To assess the role of coagulation on tumor growth, in vivo, we tested coagulation inhibitors specific for either tissue factor (TF)/factor VIIa (fVIIa) complexes or factor Xa (fXa) for antitumor activity. Here, we show that two inhibitors of TF/fVIIa, TF pathway inhibitor (TFPI) and the nematode anticoagulant protein rNAPc2, inhibit both primary and metastatic tumor growth in mice. In addition, we show that rNAPc2 is also a potent inhibitor of angiogenesis. In contrast, rNAP5, a second nematode anticoagulant protein that specifically inhibits fXa, does not exhibit antitumor activity. Because the hemostatic activity of TF/fVIIa is mediated through activation of fXa, these data suggest that proteolytic activity of TF/fVIIa promotes tumor growth and angiogenesis through a novel proangiogenic mechanism and independently of hemostasis.


Asunto(s)
Factor VIIa/antagonistas & inhibidores , Proteínas del Helminto/farmacología , Lipoproteínas/farmacología , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Tromboplastina/antagonistas & inhibidores , Animales , Anticoagulantes/farmacología , División Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Melanoma Experimental/patología , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/prevención & control
19.
Shock ; 45(4): 411-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26974318

RESUMEN

The sirtuin family consists of seven NAD+-dependent enzymes affecting a broad array of regulatory protein networks by primarily catalyzing the deacetylation of key lysine residues in regulatory proteins. The enzymatic activity of SIRT1 can be enhanced by small molecule activators known as SIRT1 activator compounds (STACs). We tested the therapeutic potential of the STAC SRT3025 in two preclinical models of severe infection, the murine cecal ligation and puncture (CLP) model to induce peritonitis and intratracheal installation of Streptococcus pneumoniae to induce severe bacterial pneumonia. SRT3025 provided significant survival benefits over vehicle control in both the peritonitis and pneumococcal pneumonia models when administered with appropriate antimicrobial agents. The survival benefit of SRT3025 in the CLP model was absent in SIRT1 knockout showing the SIRT1 dependency of SRT3025's effects. SRT3025 administration promoted bacterial clearance and significantly reduced inflammatory cytokines from the lungs of animals challenged with S. pneumoniae. SRT3025 treatment was also accompanied by striking changes in the transcription profiles in multiple inflammatory and metabolic pathways in liver, spleen, small bowel, and lung tissue. Remarkably, these organ-specific changes in the transcriptome analyses were similar following CLP or pneumococcal challenge despite different sets of pathogens at disparate sites of infection. Pharmacologic activation of SIRT1 modulates the innate host response and could represent a novel treatment strategy for severe infection.


Asunto(s)
Anilidas/farmacología , Sepsis , Sirtuina 1/inmunología , Infecciones Estreptocócicas , Streptococcus pneumoniae , Tiazoles/farmacología , Transcripción Genética , Animales , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Femenino , Inmunidad Innata/efectos de los fármacos , Ratones , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Sepsis/patología , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/patología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología
20.
Lancet ; 362(9400): 1953-8, 2003 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-14683653

RESUMEN

BACKGROUND: Infection with the Ebola virus induces overexpression of the procoagulant tissue factor in primate monocytes and macrophages, suggesting that inhibition of the tissue-factor pathway could ameliorate the effects of Ebola haemorrhagic fever. Here, we tested the notion that blockade of fVIIa/tissue factor is beneficial after infection with Ebola virus. METHODS: We used a rhesus macaque model of Ebola haemorrhagic fever, which produces near 100% mortality. We administered recombinant nematode anticoagulant protein c2 (rNAPc2), a potent inhibitor of tissue factor-initiated blood coagulation, to the macaques either 10 min (n=6) or 24 h (n=3) after a high-dose lethal injection of Ebola virus. Three animals served as untreated Ebola virus-positive controls. Historical controls were also used in some analyses. FINDINGS: Both treatment regimens prolonged survival time, with a 33% survival rate in each treatment group. Survivors are still alive and healthy after 9 months. All but one of the 17 controls died. The mean survival for the six rNAPc2-treated macaques that died was 11.7 days compared with 8.3 days for untreated controls (p=0.0184). rNAPc2 attenuated the coagulation response as evidenced by modulation of various important coagulation factors, including plasma D dimers, which were reduced in nearly all treated animals; less prominent fibrin deposits and intravascular thromboemboli were observed in tissues of some animals that succumbed to Ebola virus. Furthermore, rNAPc2 attenuated the proinflammatory response with lower plasma concentrations of interleukin 6 and monocyte chemoattractant protein-1 (MCP-1) noted in the treated than in the untreated macaques. INTERPRETATION: Post-exposure protection with rNAPc2 against Ebola virus in primates provides a new foundation for therapeutic regimens that target the disease process rather than viral replication.


Asunto(s)
Factor VIIa/antagonistas & inhibidores , Proteínas del Helminto/farmacología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Tromboplastina/antagonistas & inhibidores , Animales , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/inmunología , Modelos Animales de Enfermedad , Ebolavirus/efectos de los fármacos , Ebolavirus/inmunología , Proteínas del Helminto/inmunología , Proteínas del Helminto/uso terapéutico , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/mortalidad , Macaca mulatta , Proteínas Recombinantes/uso terapéutico , Tasa de Supervivencia , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA