Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 161(6): 463-477, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35536742

RESUMEN

In the central nervous system, most neurons co-express TrkB and TrkC, the tyrosine kinase receptors for brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3). As NT3 can also activate TrkB, it has been difficult to understand how NT3 and TrkC can exert unique roles in the assembly of neuronal circuits. Using neurons differentiated from human embryonic stem cells expressing both TrkB and TrkC, we compared Trk activation by BDNF and NT3. To avoid the complications resulting from TrkB activation by NT3, we also generated neurons from stem cells engineered to lack TrkB. We found that NT3 activates TrkC at concentrations lower than those of BDNF needed to activate TrkB. Downstream of Trk activation, the changes in gene expression caused by TrkC activation were found to be similar to those resulting from TrkB activation by BDNF, including a number of genes involved in synaptic plasticity. At high NT3 concentrations, receptor selectivity was lost as a result of TrkB activation. In addition, TrkC was down-regulated, as was also the case with TrkB at high BDNF concentrations. By contrast, receptor selectivity as well as reactivation were preserved when neurons were exposed to low neurotrophin concentrations. These results indicate that the selectivity of NT3/TrkC signalling can be explained by the ability of NT3 to activate TrkC at concentrations lower than those needed to activate TrkB. They also suggest that in a therapeutic perspective, the dosage of Trk receptor agonists will need to be taken into account if prolonged receptor activation is to be achieved.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Glicoproteínas de Membrana/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Regulación hacia Abajo , Humanos , Neuronas/metabolismo , Neurotrofina 3/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor trkB/genética , Receptor trkC/genética , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(30): E7023-E7032, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987039

RESUMEN

The diverse physiological roles of the neurotrophin family have long prompted exploration of their potential as therapeutic agents for nerve injury and neurodegenerative diseases. To date, clinical trials of one family member, brain-derived neurotrophic factor (BDNF), have disappointingly failed to meet desired endpoints. Contributing to these failures is the fact that BDNF is pharmaceutically a nonideal biologic drug candidate. It is a highly charged, yet is a net hydrophobic molecule with a low molecular weight that confers a short t1/2 in man. To circumvent these shortcomings of BDNF as a drug candidate, we have employed a function-based cellular screening assay to select activating antibodies of the BDNF receptor TrkB from a combinatorial human short-chain variable fragment antibody library. We report here the successful selection of several potent TrkB agonist antibodies and detailed biochemical and physiological characterization of one such antibody, ZEB85. By using a human TrkB reporter cell line and BDNF-responsive GABAergic neurons derived from human ES cells, we demonstrate that ZEB85 is a full agonist of TrkB, comparable in potency to BDNF toward human neurons in activation of TrkB phosphorylation, canonical signal transduction, and mRNA transcriptional regulation.


Asunto(s)
Comunicación Autocrina , Neuronas GABAérgicas/metabolismo , Biblioteca de Genes , Glicoproteínas de Membrana/agonistas , Receptor trkB/agonistas , Transducción de Señal/efectos de los fármacos , Anticuerpos de Cadena Única , Transcripción Genética/efectos de los fármacos , Línea Celular , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Fosforilación/efectos de los fármacos , Receptor trkB/genética , Receptor trkB/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología
3.
Biochem Pharmacol ; : 116401, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38945278

RESUMEN

The peptide hormone relaxin plays a critical role in tissue remodeling in a variety of tissues through activation of its cognate receptor, RXFP1. Relaxin's ability to modify extracellular matrices has provided a strong rationale for treating fibrosis in a variety of tissues. Treatment with recombinant relaxin peptides in clinical studies of heart failure has not yet proven useful, likely due to the short half-life of infused peptide. To circumvent this particular pharmacokinetic pitfall we have used a Protein-in-Protein (PiP) antibody technology described previously, to insert a single-chain human relaxin construct into the complementarity-determining region (CDR) of an immunoglobulin G (IgG) backbone, creating a relaxin molecule with a half-life of ∼4-5 days in mice. Relaxin-PiP biologics displaced Europium-labeled human relaxin in RXFP1-expressing cells and demonstrated full agonist activity on both human and mouse RXFP1 receptors. Relaxin-PiPs did not show signal transduction bias, as they activated cAMP in THP-1 cells, and cGMP and pERK signaling in primary human cardiac fibroblasts. In an induced carbon tetrachloride mouse model of liver fibrosis one relaxin-PiP, R2-PiP, caused reduction of liver lesions, ameliorated collagen accumulation in the liver with the corresponding reduction of Collagen1a1 gene expression, and increased cell proliferation in hepatic parenchyma. These relaxin biologics represent a novel approach to the design of a long-acting RXFP1 agonist to probe the clinical utility of relaxin/RXFP1 signaling to treat a variety of human fibrotic diseases.

4.
Front Mol Neurosci ; 15: 945348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35845610

RESUMEN

Signaling of BDNF via its TrkB receptor is crucial in regulating several critical aspects of the architecture and function of neurons both during development and in the adult central nervous system. Indeed, several neurological conditions, such as neurodevelopmental and neurodegenerative disorders are associated with alterations both in the expression levels of BDNF and TrkB, and in their intracellular signaling. Thus, the possibility of promoting BDNF/TrkB signaling has become relevant as a potential therapeutic intervention for neurological disorders. However, the clinical potential of BDNF itself has been limited due to its restricted diffusion rate in biological tissue, poor bioavailability and pharmacological properties, as well as the potential for unwanted side effects due to its ability to also signal via the p75NTR pathway. Several small molecule and biologic drug candidate TrkB agonists have been developed and are reported to have effects in rescuing both the pathological alterations and disease related symptoms in mouse models of several neurological diseases. However, recent side-by-side comparative studies failed to show their specificity for activating TrkB signaling cascades, suggesting the need for the generation and validation of improved candidates. In the present study, we examine the ability of the novel, fully human TrkB agonist antibody ZEB85 to modulate the architecture, activity and synaptic plasticity of hippocampal murine neurons under physiological conditions. Moreover, we show here that ZEB85 prevents ß-amyloid toxicity in cultured hippocampal neurons, in a manner which is comparable to BDNF.

5.
J Pharmacol Exp Ther ; 339(1): 115-24, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21775475

RESUMEN

Ghrelin influences a variety of metabolic functions through a direct action at its receptor, the GhrR (GhrR-1a). Ghrelin knockout (KO) and GhrR KO mice are resistant to the negative effects of high-fat diet (HFD) feeding. We have generated several classes of small-molecule GhrR antagonists and evaluated whether pharmacologic blockade of ghrelin signaling can recapitulate the phenotype of ghrelin/GhrR KO mice. Antagonist treatment blocked ghrelin-induced and spontaneous food intake; however, the effects on spontaneous feeding were absent in GhrR KO mice, suggesting target-specific effects of the antagonists. Oral administration of antagonists to HFD-fed mice improved insulin sensitivity in both glucose tolerance and glycemic clamp tests. The insulin sensitivity observed was characterized by improved glucose disposal with dramatically decreased insulin secretion. It is noteworthy that these results mimic those obtained in similar tests of HFD-fed GhrR KO mice. HFD-fed mice treated for 56 days with antagonist experienced a transient decrease in food intake but a sustained body weight decrease resulting from decreased white adipose, but not lean tissue. They also had improved glucose disposal and a striking reduction in the amount of insulin needed to achieve this. These mice had reduced hepatic steatosis, improved liver function, and no evidence of systemic toxicity relative to controls. Furthermore, GhrR KO mice placed on low- or high-fat diets had lifespans similar to the wild type, emphasizing the long-term safety of ghrelin receptor blockade. We have therefore demonstrated that chronic pharmacologic blockade of the GhrR is an effective and safe strategy for treating metabolic syndrome.


Asunto(s)
Resistencia a la Insulina/fisiología , Insulina/metabolismo , Receptores de Ghrelina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Fármacos Antiobesidad/farmacología , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/antagonistas & inhibidores , Ghrelina/farmacología , Técnica de Clampeo de la Glucosa , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/tratamiento farmacológico , Receptores de Ghrelina/fisiología , Estrés Fisiológico/fisiología
6.
BMC Physiol ; 11: 1, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21211044

RESUMEN

BACKGROUND: We and others have demonstrated previously that ghrelin receptor (GhrR) knock out (KO) mice fed a high fat diet (HFD) have increased insulin sensitivity and metabolic flexibility relative to WT littermates. A striking feature of the HFD-fed GhrR KO mouse is the dramatic decrease in hepatic steatosis. To characterize further the underlying mechanisms of glucose homeostasis in GhrR KO mice, we conducted both hyperglycemic (HG) and hyperinsulinemic-euglycemic (HI-E) clamps. Additionally, we investigated tissue glucose uptake and specifically examined liver insulin sensitivity. RESULTS: Consistent with glucose tolerance-test data, in HG clamp experiments, GhrR KO mice showed a reduction in glucose-stimulated insulin release relative to WT littermates. Nevertheless, a robust 1st phase insulin secretion was still achieved, indicating that a healthy ß-cell response is maintained. Additionally, GhrR KO mice demonstrated both a significantly increased glucose infusion rate and significantly reduced insulin requirement for maintenance of the HG clamp, consistent with their relative insulin sensitivity. In HI-E clamps, both LFD-fed and HFD-fed GhrR KO mice showed higher peripheral insulin sensitivity relative to WT littermates as indicated by a significant increase in insulin-stimulated glucose disposal (Rd), and decreased hepatic glucose production (HGP). HFD-fed GhrR KO mice showed a marked increase in peripheral tissue glucose uptake in a variety of tissues, including skeletal muscle, brown adipose tissue and white adipose tissue. GhrR KO mice fed a HFD also showed a modest, but significant decrease in conversion of pyruvate to glucose, as would be anticipated if these mice displayed increased liver insulin sensitivity. Additionally, the levels of UCP2 and UCP1 were reduced in the liver and BAT, respectively, in GhrR KO mice relative to WT mice. CONCLUSIONS: These results indicate that improved glucose homeostasis of GhrR KO mice is characterized by robust improvements of glucose disposal in both normal and metabolically challenged states, relative to WT controls. GhrR KO mice have an intact 1st phase insulin response but require significantly less insulin for glucose disposal. Our experiments reveal that the insulin sensitivity of GhrR KO mice is due to both BW independent and dependent factors. We also provide several lines of evidence that a key feature of the GhrR KO mouse is maintenance of hepatic insulin sensitivity during metabolic challenge.


Asunto(s)
Técnica de Clampeo de la Glucosa/métodos , Resistencia a la Insulina/genética , Insulina/sangre , Receptores de Ghrelina/deficiencia , Animales , Grasas de la Dieta/administración & dosificación , Prueba de Tolerancia a la Glucosa/métodos , Índice Glucémico/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R747-54, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20018821

RESUMEN

To define the relationship between the respiratory quotient (RQ) and energy intake (EI) and to determine the impact of spontaneous locomotor activity (LMA) in the development of diet-induced obesity (DIO), we fed C57BL/6 mice a high-fat diet (HFD) for either 4 days or 17 wk and analyzed them using indirect calorimetry. Importantly, changes in body mass during calorimetry (DeltaM(b)) significantly covaried with RQ and EI; adjusting the data for DeltaM(b) permitted an analysis of the energy-balanced state. The 24-h RQ strongly predicted 24-h EI, and the slope of this relationship was diet dependent (HFD or chow) but independent of the HFD feeding period. Early-stage DIO was characterized by dark-period hyperphagia and fat storage, offset by greater light-period lipid oxidation; later stage DIO mice had a milder hyperphagia and lower substrate flexibility. Consequently, whereas 24-h RQ equaled the food quotient of the HFD in both early- and late-stage DIO, the range of RQ values was negatively correlated with, and mostly explained by, 24-h EI only in late-stage DIO. Lean and early-stage DIO mice had similar LMA values that were reduced in late-stage DIO. However, LMA significantly explained variance in total energy expenditure (EE) in only early-stage DIO mice. This indicated that the link between LMA and EE was a transient adaptive response to early DIO, whereas the later loss of LMA did not explain body weight gain in C57BL/6 DIO mice.


Asunto(s)
Peso Corporal/fisiología , Metabolismo Energético/fisiología , Hiperfagia/metabolismo , Obesidad/metabolismo , Consumo de Oxígeno/fisiología , Animales , Calorimetría Indirecta , Grasas de la Dieta/farmacología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Valor Predictivo de las Pruebas
8.
J Pharmacol Exp Ther ; 329(3): 1178-86, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19252061

RESUMEN

The orexigenic peptide ghrelin has been shown to have prokinetic activity in the gastrointestinal (GI) system of several species, including humans. In this series of experiments, we have evaluated the prokinetic activity of novel, small-molecule ghrelin receptor (GhrR) agonists after parenteral and peroral dosing in mice and rats. Gastric emptying, small intestinal transport, and fecal output were determined after intraperitoneal and intracerebroventricular dosing of GhrR agonists, using ghrelin as a positive control. These same parameters were evaluated after oral gavage dosing of the synthetic agonists. Regardless of dose route, GhrR agonist treatment increased gastric emptying, small intestinal transit, and fecal output. However, fecal output was only increased by GhrR agonist treatment if mice were able to feed during the stimulatory period. Thus, GhrR agonists can stimulate upper GI motility, and the orexigenic action of the compounds can indirectly contribute to prokinetic activity along the entire GI tract. The orexigenic and prokinetic effects of either ghrelin or small-molecule GhrR agonists were selective for the GhrR because they were absent when evaluated in GhrR knockout mice. We next evaluated the efficacy of the synthetic GhrR agonists dosed in a model of opiate-induced bowel dysfunction induced by a single injection of morphine. Oral dosing of a GhrR agonist normalized GI motility in opiate-induced dysmotility. These data demonstrate the potential utility of GhrR agonists for treating gastrointestinal hypomotility disorders.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Ghrelina/administración & dosificación , Ghrelina/farmacología , Hormonas Peptídicas/administración & dosificación , Hormonas Peptídicas/farmacología , Receptores de Ghrelina/agonistas , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Enfermedad de Bowen/inducido químicamente , Enfermedad de Bowen/tratamiento farmacológico , Enfermedad de Bowen/fisiopatología , Sistema Nervioso Central/efectos de los fármacos , Defecación/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Vaciamiento Gástrico/efectos de los fármacos , Tránsito Gastrointestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Hormonas Peptídicas/sangre , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo
9.
Mol Cell Biol ; 26(1): 28-38, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16354677

RESUMEN

Human SIRT1 is an enzyme that deacetylates the p53 tumor suppressor protein and has been suggested to modulate p53-dependent functions including DNA damage-induced cell death. In this report, we used EX-527, a novel, potent, and specific small-molecule inhibitor of SIRT1 catalytic activity to examine the role of SIRT1 in p53 acetylation and cell survival after DNA damage. Treatment with EX-527 dramatically increased acetylation at lysine 382 of p53 after different types of DNA damage in primary human mammary epithelial cells and several cell lines. Significantly, inhibition of SIRT1 catalytic activity by EX-527 had no effect on cell growth, viability, or p53-controlled gene expression in cells treated with etoposide. Acetyl-p53 was also increased by the histone deacetylase (HDAC) class I/II inhibitor trichostatin A (TSA). EX-527 and TSA acted synergistically to increase acetyl-p53 levels, confirming that p53 acetylation is regulated by both SIRT1 and HDACs. While TSA alone reduced cell survival after DNA damage, the combination of EX-527 and TSA had no further effect on cell viability and growth. These results show that, although SIRT1 deacetylates p53, this does not play a role in cell survival following DNA damage in certain cell lines and primary human mammary epithelial cells.


Asunto(s)
Daño del ADN , Inhibidores de Histona Desacetilasas , Sirtuinas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Catálisis/efectos de los fármacos , Línea Celular , Supervivencia Celular , ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Humanos , Ácidos Hidroxámicos/farmacología , Sirtuina 1
10.
Regul Pept ; 150(1-3): 55-61, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18453014

RESUMEN

Stimulation of the ghrelin receptor (GhrR) by ghrelin results in a variety of metabolic changes including increased food intake, fat storage and insulin resistance. Loss of ghrelin signaling is protective against diet-induced obesity, suggesting that ghrelin plays a significant homeostatic role in conditions of metabolic stress. We examined glycemic control in GhrR -/- mice fed a high-fat diet, and used indirect calorimetry to assess fuel substrate usage and energy expenditure. GhrR -/- mice fed a high-fat diet had several measures of greater insulin sensitivity, including: lower fasted blood glucose and plasma insulin, lower %Hb(A1c), lower insulin levels during glucose tolerance tests, and improved performance in hyperinsulinemic-euglycemic and hyperglycemic clamp studies. GhrR -/- mice fed a high-fat diet did not develop hepatic steatosis and had lower total cholesterol, relative to controls. Furthermore, GhrR -/- mice demonstrated a lower intestinal triglyceride secretion rate of dietary lipid. GhrR -/- mice have higher respiratory quotients (RQ), indicating a preference for carbohydrate as fuel. The range of RQ values was wider in GhrR -/- mice, indicating greater metabolic flexibility and insulin sensitivity in these animals. We therefore propose that loss of ghrelin signaling promotes insulin sensitivity and metabolic flexibility, and protects against several fatty diet-induced features of metabolic syndrome due to convergent changes in the intake, absorption and utilization of energy.


Asunto(s)
Grasas de la Dieta/metabolismo , Metabolismo Energético/fisiología , Resistencia a la Insulina/fisiología , Receptores de Ghrelina/genética , Animales , Glucemia/análisis , Calorimetría Indirecta/métodos , Colesterol/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Ayuno , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Hemoglobina Glucada/metabolismo , Insulina/sangre , Ratones , Ratones Noqueados , Triglicéridos/metabolismo
11.
Nat Rev Drug Discov ; 4(7): 569-80, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15976816

RESUMEN

There has recently been significant progress in our understanding of the mechanisms that regulate ageing, and it has been shown that changes in single genes can dramatically extend lifespan and increase resistance to many diseases. Furthermore, many of these genes belong to evolutionarily conserved pathways that also control energy metabolism. In this review, we describe the shared molecular machinery that regulates ageing and energy metabolism. Although drugs to slow ageing face severe regulatory hurdles, it is likely that an understanding of ageing pathways will help to identify novel drug targets to treat metabolic disorders and other age-related diseases.


Asunto(s)
Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Diseño de Fármacos , Metabolismo/efectos de los fármacos , Metabolismo/fisiología , Proteínas Quinasas Activadas por AMP , Adipocitos/efectos de los fármacos , Envejecimiento/genética , Animales , Ingestión de Alimentos/fisiología , Humanos , Insulina/fisiología , Esperanza de Vida , Metabolismo/genética , Complejos Multienzimáticos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología
12.
Aging Cell ; 5(2): 119-26, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16626391

RESUMEN

Molecular genetics in lower organisms has allowed the elucidation of pathways that modulate the aging process. In certain instances, evolutionarily conserved genes and pathways have been shown to regulate lifespan in mammals as well. Many gene products known to affect lifespan are intimately involved in the control of energy metabolism, including the fuel sensor AMP-activated protein kinase (AMPK). We have shown previously that over-expression of an AMPK alpha subunit in Caenorhabditis elegans, designated aak-2, increases lifespan. Here we show the interaction of aak-2 with other pathways known to control aging in worms. Lifespan extension caused by daf-2/insulin-like signaling mutations was highly dependent on aak-2, as was the lifespan extension caused by over-expression of the deacetylase, sir-2.1. Similarly, there was partial requirement for aak-2 in lifespan extension by mitochondrial mutations (isp-1 and clk-1). Conversely, aak-2 was not required for lifespan extension in mutants lacking germline stem cells (glp-1) or mutants of the eating response (eat-2). These results show that aging is controlled by overlapping but distinct pathways and that AMPK/aak-2 represents a node in a network of evolutionarily conserved biochemical pathways that control aging.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/fisiología , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Nucleótidos de Adenina/metabolismo , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Conducta Alimentaria , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Mutación de Línea Germinal/genética , Larva/crecimiento & desarrollo , Longevidad , Mitocondrias/metabolismo , Modelos Biológicos , Unión Proteica , Sirtuinas/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo
13.
Curr Alzheimer Res ; 4(2): 153-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17430240

RESUMEN

Identification of genes and pathways that alter lifespan has allowed for new insights into factors that control the aging process as well as disease. While strong molecular links exist between aging and metabolism, we hypothesize that targeting the mechanisms involved in aging will also give rise to therapeutics that treat other devastating age-related diseases, such as neurodegeneration, cancer, inflammation and cardiovascular disease. Insulin sensitivity, glycemic control and adiposity are not only hallmarks of the major metabolic diseases, type 2 diabetes and obesity, but they also represent significant risk factors for the development of Alzheimer's Disease and cognitive impairment. Insulin/IGF-1 signaling is an important pathway regulating aging and disease in a variety of species, including mammals. Here we describe an important role for the gut-derived peptide ghrelin in upstream signaling through the insulin/IGF-1 pathway and exemplify modulation of ghrelin signaling as an approach to mechanistic treatment of multiple age-related diseases by virtue of its ability to regulate key metabolic functions.


Asunto(s)
Adiposidad , Envejecimiento/fisiología , Glucemia , Resistencia a la Insulina , Longevidad/fisiología , Animales , Humanos
14.
J Neurosci ; 23(20): 7577-85, 2003 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12930796

RESUMEN

The principal alpha subunit of voltage-gated sodium channels is associated with auxiliary beta subunits that modify channel function and mediate protein-protein interactions. We have identified a new beta subunit termed beta4. Like the beta1-beta3 subunits, beta4 contains a cleaved signal sequence, an extracellular Ig-like fold, a transmembrane segment, and a short intracellular C-terminal tail. Using TaqMan reverse transcription-PCR analysis, in situ hybridization, and immunocytochemistry, we show that beta4 is widely distributed in neurons in the brain, spinal cord, and some sensory neurons.beta4 is most similar to the beta2 subunit (35% identity), and, like the beta2 subunit, the Ig-like fold of beta4 contains an unpaired cysteine that may interact with the alpha subunit. Under nonreducing conditions, beta4 has a molecular mass exceeding 250 kDa because of its covalent linkage to Nav1.2a, whereas on reduction, it migrates with a molecular mass of 38 kDa, similar to the mature glycosylated forms of the other beta subunits. Coexpression of beta4 with brain Nav1.2a and skeletal muscle Nav1.4 alpha subunits in tsA-201 cells resulted in a negative shift in the voltage dependence of channel activation, which overrode the opposite effects of beta1 and beta3 subunits when they were present. This novel, disulfide-linked beta subunit is likely to affect both protein-protein interactions and physiological function of multiple sodium channel alpha subunits.


Asunto(s)
Subunidades de Proteína/química , Subunidades de Proteína/fisiología , Canales de Sodio/química , Canales de Sodio/fisiología , Secuencia de Aminoácidos , Animales , Adhesión Celular , Línea Celular , Clonación Molecular , Disulfuros/química , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido , Canales de Sodio/genética , Canales de Sodio/metabolismo , Distribución Tisular , Subunidad beta-2 de Canal de Sodio Activado por Voltaje , Subunidad beta-4 de Canal de Sodio Activado por Voltaje
15.
J Med Chem ; 48(25): 8045-54, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16335928

RESUMEN

High-throughput screening against the human sirtuin SIRT1 led to the discovery of a series of indoles as potent inhibitors that are selective for SIRT1 over other deacetylases and NAD-processing enzymes. The most potent compounds described herein inhibit SIRT1 with IC50 values of 60-100 nM, representing a 500-fold improvement over previously reported SIRT inhibitors. Preparation of enantiomerically pure indole derivatives allowed for their characterization in vitro and in vivo. Kinetic analyses suggest that these inhibitors bind after the release of nicotinamide from the enzyme and prevent the release of deacetylated peptide and O-acetyl-ADP-ribose, the products of enzyme-catalyzed deacetylation. These SIRT1 inhibitors are low molecular weight, cell-permeable, orally bioavailable, and metabolically stable. These compounds provide chemical tools to study the biology of SIRT1 and to explore therapeutic uses for SIRT1 inhibitors.


Asunto(s)
Carbazoles/síntesis química , Inhibidores de Histona Desacetilasas , Indoles/síntesis química , Sirtuinas/antagonistas & inhibidores , Animales , Disponibilidad Biológica , Células CHO , Carbazoles/química , Carbazoles/farmacología , Permeabilidad de la Membrana Celular , Cricetinae , Cricetulus , Estabilidad de Medicamentos , Fluorometría , Histona Desacetilasas/química , Humanos , Técnicas In Vitro , Indoles/química , Indoles/farmacología , Cinética , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , NAD/química , NAD+ Nucleosidasa/química , Niacinamida/química , Ratas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Sirtuina 1 , Sirtuinas/química , Estereoisomerismo , Relación Estructura-Actividad
16.
FEBS Lett ; 530(1-3): 73-8, 2002 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-12387869

RESUMEN

PYRIN-containing Apaf-1-like proteins (PYPAFs) are a recently identified family of proteins thought to function in apoptotic and inflammatory signaling pathways. PYPAF1 and PYPAF7 proteins have been found to assemble with the PYRIN-CARD protein ASC and coordinate the activation of NF-kappaB and pro-caspase-1. To determine if other PYPAF family members function in pro-inflammatory signaling pathways, we screened five other PYPAF proteins (PYPAF2, PYPAF3, PYPAF4, PYPAF5 and PYPAF6) for their ability to activate NF-kappaB and pro-caspase-1. Co-expression of PYPAF5 with ASC results in a synergistic activation of NF-kappaB and the recruitment of PYPAF5 to punctate structures in the cytoplasm. The expression of PYPAF5 is highly restricted to granulocytes and T-cells, indicating a role for this protein in inflammatory signaling. In contrast, PYPAF2, PYPAF3, PYPAF4 and PYPAF6 failed to colocalize with ASC and activate NF-kappaB. PYPAF5 also synergistically activated caspase-1-dependent cytokine processing when co-expressed with ASC. These findings suggest that PYPAF5 functions in immune cells to coordinate the transduction of pro-inflammatory signals to the activation of NF-kappaB and pro-caspase-1.


Asunto(s)
Proteínas Portadoras/fisiología , Caspasa 1/fisiología , Péptidos y Proteínas de Señalización Intracelular , FN-kappa B/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Cartilla de ADN , Activación Enzimática , Humanos , Leucocitos/metabolismo , Datos de Secuencia Molecular , Transducción de Señal
17.
J Biomol Screen ; 7(1): 79-85, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11897058

RESUMEN

The study of ion channel-mediated changes in membrane potential using the conventional bisoxonol fluorescent dye DiBAC(4)(3) has several limitations, including a slow onset of response and multistep preparation, that limit both the fidelity of the results and the throughput of membrane potential assays. Here, we report the characterization of the FLIPR Membrane Potential Assay Kit (FMP) in cells expressing voltage- and ligand-gated ion channels. The steady-state and kinetics fluorescence properties of FMP were compared with those of DiBAC(4)(3), using both FLIPR and whole-cell patch-clamp recording. Our experiments with the voltage-gated K(+) channel, hElk-1, revealed that FMP was 14-fold faster than DiBAC(4)(3) in response to depolarization. On addition of 60 mM KCl, the kinetics of fluorescence changes of FMP using FLIPR were identical to those observed in the electrophysiological studies using whole-cell current clamp. In addition, KCl concentration-dependent increases in FMP fluorescence correlated with the changes of membrane potential recorded in whole-cell patch clamp. In studies examining vanilloid receptor-1, a ligand-gated nonselective cation channel, FMP was superior to DiBAC(4)(3) with respect to both kinetics and amplitude of capsaicin-induced fluorescence changes. FMP has also been used to measure the activation of K(ATP) and hERG. Thus this novel membrane potential dye represents a powerful tool for developing high-throughput screening assays for ion channels.


Asunto(s)
Biotecnología/métodos , Membrana Celular/metabolismo , Colorantes Fluorescentes/farmacología , Canales Iónicos , Adenosina Trifosfato/metabolismo , Animales , Automatización , Células CHO , Cationes , Línea Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Electrofisiología , Humanos , Cinética , Técnicas de Placa-Clamp , Potasio/metabolismo , Cloruro de Potasio/química , Espectrometría de Fluorescencia , Factores de Tiempo
18.
Future Med Chem ; 2(12): 1751-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21428798

RESUMEN

BACKGROUND: SRT1720 and SRT2183 were described recently as activators of the NAD+-dependent deacetylase, SIRT1. These molecules enhanced metabolic function when administered to rodents at doses of 100-500 mg/kg/day, purportedly by activating SIRT1 enzymatic activity in various tissues; however, considerable controversy surrounds these claims. RESULTS: We find that these molecules do not activate SIRT1 deacetylase activity when tested in a variety of enzymatic assay formats and conditions. The compounds effectively decrease acetylated p53 in cells treated with DNA damaging agents but do so in cells that lack SIRT1, calling into question their designation as direct activators of SIRT1. In contrast, we find that the compounds inhibit p300 histone acetyltransferase activity in vitro, suggesting a possible mechanism for their effects in vivo. CONCLUSION: Structural features of these molecules may account for false-positive activation using fluorescence-based assays.


Asunto(s)
Activadores de Enzimas/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Sirtuinas/metabolismo , Acetilación , Animales , Línea Celular , Activadores de Enzimas/química , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Ratones , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
19.
Growth Horm IGF Res ; 20(1): 73-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19747867

RESUMEN

The goal of this study was to examine factors that contribute to energy balance in female GHR -/- mice. We measured energy intake, energy expenditure (EE), fuel utilization, body mass (M(b)) changes and physical activity in 17month-old female GHR -/- mice and their age-matched wild type littermates. The GHR -/- mice were smaller, consumed more food per unit M(b), had greater EE per unit M(b) and had an increase in 24-h EE/M(b) that was similar to the increase in their surface-area-to-volume ratio. Locomotor activity (LMA) was reduced in the GHR -/- mice, but the energetic cost associated with their LMA was greater than in wild type controls. Furthermore, M(b) and LMA were independent explanatory covariates of most of the variance in EE, and when adjusted for M(b) and LMA, the GHR -/- mice had higher EE during both the light and dark phases of the daily cycle. Respiratory quotient was lower in GHR -/- mice during the light phase, which indicated a greater utilization of lipid relative to carbohydrate in these mice. Additionally, GHR -/- mice had higher ratios of caloric intake to EE at several intervals during the dark phase, and this effect was greater and more sustained in the final 3h of the dark phase. Therefore, we conclude that GHR -/- mice are able to overcome the substantial energetic challenges of dwarfism through several mechanisms that promote stable M(b). Relative to wild type mice, the GHR -/- mice consumed more calories per unit M(b), which offset the disproportionate increase in their daily energy expenditure. While GHR -/- mice oxidized a greater proportion of lipid during the light phase in order to meet their energy requirements, they achieved greater energy efficiency and storage during the dark phase through a combination of higher energy consumption and lower LMA.


Asunto(s)
Metabolismo Energético/fisiología , Receptores de Somatotropina/metabolismo , Ciclos de Actividad/fisiología , Animales , Enanismo/metabolismo , Ingestión de Energía/fisiología , Femenino , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Mutantes , Actividad Motora/fisiología , Receptores de Somatotropina/genética , Receptores de Somatotropina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA