Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Biol Chem ; 299(4): 103057, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36822331

RESUMEN

CLEC16A is an E3 ubiquitin ligase that regulates mitochondrial quality control through mitophagy and is associated with over 20 human diseases. CLEC16A forms a complex with another E3 ligase, RNF41, and a ubiquitin-specific peptidase, USP8; however, regions that regulate CLEC16A activity or the assembly of the tripartite mitophagy regulatory complex are unknown. Here, we report that CLEC16A contains an internal intrinsically disordered protein region (IDPR) that is crucial for CLEC16A function and turnover. IDPRs lack a fixed secondary structure and possess emerging yet still equivocal roles in protein stability, interactions, and enzymatic activity. We find that the internal IDPR of CLEC16A is crucial for its degradation. CLEC16A turnover was promoted by RNF41, which binds and acts upon the internal IDPR to destabilize CLEC16A. Loss of this internal IDPR also destabilized the ubiquitin-dependent tripartite CLEC16A-RNF41-USP8 complex. Finally, the presence of an internal IDPR within CLEC16A was confirmed using NMR and CD spectroscopy. Together, our studies reveal that an IDPR is essential to control the reciprocal regulatory balance between CLEC16A and RNF41, which could be targeted to improve mitochondrial health in disease.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Mitofagia , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Lectinas Tipo C/metabolismo
2.
Am J Physiol Renal Physiol ; 326(1): F30-F38, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37916286

RESUMEN

Plasma nucleosides-pseudouridine (PU) and N2N2-dimethyl guanosine (DMG) predict the progression of type 2 diabetic kidney disease (DKD) to end-stage renal disease, but the mechanisms underlying this relationship are not well understood. We used a well-characterized model of type 2 diabetes (db/db mice) and control nondiabetic mice (db/m mice) to characterize the production and excretion of PU and DMG levels using liquid chromatography-mass spectrometry. The fractional excretion of PU and DMG was decreased in db/db mice compared with control mice at 24 wk before any changes to renal function. We then examined the dynamic changes in nucleoside metabolism using in vivo metabolic flux analysis with the injection of labeled nucleoside precursors. Metabolic flux analysis revealed significant decreases in the ratio of urine-to-plasma labeling of PU and DMG in db/db mice compared with db/m mice, indicating significant tubular dysfunction in diabetic kidney disease. We observed that the gene and protein expression of the renal tubular transporters involved with nucleoside transport in diabetic kidneys in mice and humans was reduced. In conclusion, this study strongly suggests that tubular handling of nucleosides is altered in early DKD, in part explaining the association of PU and DMG with human DKD progression observed in previous studies.NEW & NOTEWORTHY Tubular dysfunction explains the association between the nucleosides pseudouridine and N2N2-dimethyl guanosine and diabetic kidney disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Humanos , Ratones , Animales , Nefropatías Diabéticas/metabolismo , Seudouridina/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Nucleósidos/metabolismo , Eliminación Renal , Riñón/metabolismo , Guanosina/metabolismo
3.
Cardiovasc Drugs Ther ; 29(2): 121-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25820670

RESUMEN

INTRODUCTION: Irisin is a newly identified 112 amino acid hormone, derived as a product of fibronectin type III domain containing 5 (FNDC5), which is highly related to metabolic activity in skeletal muscle and brown fat. The effects of irisin on cardiovascular functions are unknown. PURPOSE: To explore the effects of central and peripheral irisin on cardiovascular functions. METHODS: Irisin was either administrated into 3rd ventricle of rats or intravenously, and its effects on blood pressure and cardiac contractibility measured. RESULTS: Administration of recombinant human irisin into the 3rd brain ventricle of rats activated neurons in the paraventricular nuclei of the hypothalamus. Central administration of irisin increased blood pressure and cardiac contractibility. Exogenous irisin reversed atenolol-induced inhibition of cardiac contractibility. In contrast, peripheral administration of irisin reduced blood pressure in both control and spontaneously hypertensive rats. Irisin dilated mesenteric artery rings through ATP-sensitive potassium channels. CONCLUSION: Our studies indicate that central and peripheral irisin may differentially regulate cardiovascular activities.


Asunto(s)
Presión Sanguínea/fisiología , Fibronectinas/fisiología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Administración Intravenosa , Animales , Presión Sanguínea/efectos de los fármacos , Fibronectinas/administración & dosificación , Fibronectinas/farmacología , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Infusiones Intraventriculares , Canales KATP/efectos de los fármacos , Canales KATP/fisiología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Ratas , Ratas Endogámicas SHR , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
4.
Exp Physiol ; 98(12): 1696-704, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23955309

RESUMEN

Previous studies have demonstrated that mammalian target of rapamycin (mTOR) signalling in the hypothalamus is involved in the control of energy homeostasis. The aim of this study was to characterize the effect of mTOR signalling in the dorsal motor nucleus of the vagus (DMNV) on energy intake. Phospho-mTOR was detected in the DMNV neurons, and its levels were increased by energy deprivation. Rapamycin significantly inhibited mTOR activity and reduced food intake when administrated into the fourth ventricle. Exposure of DMNV neurons to ghrelin increased the phosphorylation of mTOR. Injection of ghrelin into the fourth ventricle significantly increased food intake relative to the control vehicle. Pretreatment with rapamycin for 15 min attenuated the orexigenic effect of ghrelin. A reduction in the phosphorylation of mTOR was observed following injection of nesfatin-1 into the fourth ventricle. When administrated by injection into the fourth ventricle, nesfatin-1 suppressed food intake in comparison with the control vehicle. The anorexigenic effect of nesfatin-1 was significantly attenuated by pretreatment with leucine for 15 min. All these findings suggest that mTOR signalling in the DMNV neurons regulates both the nutrient and the hormonal signals for the modulation of food intake.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Ingestión de Alimentos/fisiología , Ghrelina/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Nervio Vago/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Ingestión de Energía , Ayuno/fisiología , Masculino , Nucleobindinas , Ratas , Ratas Sprague-Dawley
5.
Autophagy ; 19(2): 525-543, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35604110

RESUMEN

CLEC16A regulates mitochondrial health through mitophagy and is associated with over 20 human diseases. However, the key structural and functional regions of CLEC16A, and their relevance for human disease, remain unknown. Here, we report that a disease-associated CLEC16A variant lacks a C-terminal intrinsically disordered protein region (IDPR) that is critical for mitochondrial quality control. IDPRs comprise nearly half of the human proteome, yet their mechanistic roles in human disease are poorly understood. Using carbon detect NMR, we find that the CLEC16A C terminus lacks secondary structure, validating the presence of an IDPR. Loss of the CLEC16A C-terminal IDPR in vivo impairs mitophagy, mitochondrial function, and glucose-stimulated insulin secretion, ultimately causing glucose intolerance. Deletion of the CLEC16A C-terminal IDPR increases CLEC16A ubiquitination and degradation, thus impairing assembly of the mitophagy regulatory machinery. Importantly, CLEC16A stability is dependent on proline bias within the C-terminal IDPR, but not amino acid sequence order or charge. Together, we elucidate how an IDPR in CLEC16A regulates mitophagy and implicate pathogenic human gene variants that disrupt IDPRs as novel contributors to diabetes and other CLEC16A-associated diseases.Abbreviations : CAS: carbon-detect amino-acid specific; IDPR: intrinsically disordered protein region; MEFs: mouse embryonic fibroblasts; NMR: nuclear magnetic resonance.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Mitofagia , Humanos , Animales , Ratones , Mitofagia/genética , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Autofagia , Fibroblastos/metabolismo , Ubiquitinación , Proteínas de Transporte de Monosacáridos/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 303(5): G570-7, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22723266

RESUMEN

Nesfatin-1, a novel hypothalamic peptide, inhibits nocturnal feeding behavior and gastrointestinal motility in rodents. The effects of nesfatin-1 on gastrointestinal secretory function, including gastric acid production, have not been evaluated. Nesfatin-1 was injected into the fourth intracerebral ventricle (4V) of chronically cannulated rats to identify a nesfatin dose sufficient to inhibit food intake. Nesfatin-1 (2 µg) inhibited dark-phase food intake, in a dose-dependent fashion, for >3 h. Gastric acid production was evaluated in urethane-anesthetized rats. Nesfatin-1 (2 µg) was introduced via the 4V following endocrine stimulation of gastric acid secretion by pentagastrin (2 µg·kg(-1)·h(-1) iv), vagal stimulation with 2-deoxy-D-glucose (200 mg/kg sc), or no stimulus. Gastric secretions were collected via gastric cannula and neutralized by titration to determine acid content. Nesfatin-1 did not affect basal and pentagastrin-stimulated gastric acid secretion, whereas 2-deoxy-D-glucose-stimulated gastric acid production was inhibited by nesfatin-1 in a dose-dependent manner. c-Fos immunofluorescence in brain sections was used to evaluate in vivo neuronal activation by nesfatin-1 administered via the 4V. Nesfatin-1 caused activation of efferent vagal neurons, as evidenced by a 16-fold increase in the mean number of c-Fos-positive neurons in the dorsal motor nucleus of the vagus (DMNV) in nesfatin-1-treated animals vs. controls (P < 0.01). Finally, nesfatin-induced Ca(2+) signaling was evaluated in primary cultured DMNV neurons from neonatal rats. Nesfatin-1 caused dose-dependent Ca(2+) increments in 95% of cultured DMNV neurons. These studies demonstrate that central administration of nesfatin-1, at doses sufficient to inhibit food intake, results in inhibition of vagally stimulated secretion of gastric acid. Nesfatin-1 activates DMNV efferent vagal neurons in vivo and triggers Ca(2+) signaling in cultured DMNV neurons.


Asunto(s)
Proteínas de Unión al Calcio/farmacología , Proteínas de Unión al ADN/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Proteínas del Tejido Nervioso/farmacología , Nervio Vago/efectos de los fármacos , Animales , Calcio/metabolismo , Masculino , Nucleobindinas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Vago/fisiología
7.
BMC Neurosci ; 12: 95, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21955513

RESUMEN

BACKGROUND: The arcuate nucleus of the hypothalamus regulates food intake. Ankyrin repeat and SOCS box containing protein 4 (Asb-4) is expressed in neuropeptide Y and proopiomelanocortin (POMC) neurons in the arcuate nucleus, target neurons in the regulation of food intake and metabolism by insulin and leptin. However, the target protein(s) of Asb-4 in these neurons remains unknown. Insulin receptor substrate 4 (IRS4) is an adaptor molecule involved in the signal transduction by both insulin and leptin. In the present study we examined the colocalization and interaction of Asb-4 with IRS4 and the involvement of Asb-4 in insulin signaling. RESULTS: In situ hybridization showed that the expression pattern of Asb-4 was consistent with that of IRS4 in the rat brain. Double in situ hybridization showed that IRS4 colocalized with Asb-4, and both Asb-4 and IRS4 mRNA were expressed in proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons within the arcuate nucleus of the hypothalamus. In HEK293 cells co-transfected with Myc-tagged Asb-4 and Flag-tagged IRS4, Asb-4 co-immunoprecipitated with IRS4; In these cells endogenous IRS4 also co-immunoprecipitated with transfected Myc-Asb-4; Furthermore, Asb-4 co-immunoprecipitated with IRS4 in rat hypothalamic extracts. In HEK293 cells over expression of Asb-4 decreased IRS4 protein levels and deletion of the SOCS box abolished this effect. Asb-4 increased the ubiquitination of IRS4; Deletion of SOCS box abolished this effect. Expression of Asb-4 decreased both basal and insulin-stimulated phosphorylation of AKT at Thr308. CONCLUSIONS: These data demonstrated that Asb-4 co-localizes and interacts with IRS4 in hypothalamic neurons. The interaction of Asb-4 with IRS4 in cell lines mediates the degradation of IRS4 and decreases insulin signaling.


Asunto(s)
Hipotálamo/citología , Hipotálamo/metabolismo , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neuronas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Células HEK293 , Humanos , Insulina/metabolismo , Insulina/fisiología , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología
8.
JCI Insight ; 5(24)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33232298

RESUMEN

Inflammatory damage contributes to ß cell failure in type 1 and 2 diabetes (T1D and T2D, respectively). Mitochondria are damaged by inflammatory signaling in ß cells, resulting in impaired bioenergetics and initiation of proapoptotic machinery. Hence, the identification of protective responses to inflammation could lead to new therapeutic targets. Here, we report that mitophagy serves as a protective response to inflammatory stress in both human and rodent ß cells. Utilizing in vivo mitophagy reporters, we observed that diabetogenic proinflammatory cytokines induced mitophagy in response to nitrosative/oxidative mitochondrial damage. Mitophagy-deficient ß cells were sensitized to inflammatory stress, leading to the accumulation of fragmented dysfunctional mitochondria, increased ß cell death, and hyperglycemia. Overexpression of CLEC16A, a T1D gene and mitophagy regulator whose expression in islets is protective against T1D, ameliorated cytokine-induced human ß cell apoptosis. Thus, mitophagy promotes ß cell survival and prevents diabetes by countering inflammatory injury. Targeting this pathway has the potential to prevent ß cell failure in diabetes and may be beneficial in other inflammatory conditions.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/metabolismo , Mitofagia/fisiología , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Apoptosis , Supervivencia Celular , Complicaciones de la Diabetes , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/metabolismo , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Estrés Oxidativo , Cultivo Primario de Células , Sustancias Protectoras/metabolismo , Transducción de Señal
9.
Endocrinology ; 149(9): 4710-6, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18535105

RESUMEN

Ghrelin, a gastric peptide hormone, has been reported to regulate GH secretion and energy homeostasis. Here, we examined the effect of des-acyl ghrelin driven from the fatty acid-binding protein-4 (FABP4) promoter on adiposity and glucose metabolism. A high level of expression of des-acyl ghrelin (692 +/- 293 fmol/g fat) in adipose tissue was detected in FABP4-ghrelin transgenic mice, but not in wild-type littermates. Circulating des-acyl ghrelin was significantly higher in FABP4-ghrelin transgenic mice (8409 +/- 3390 pm) compared with wild-type mice (513 +/- 58 pm). No significant change was observed for plasma acylated ghrelin and obestatin. Epididymal and perirenal fat masses decreased 35 +/- 9 and 52 +/- 9%, respectively, in FABP4-ghrelin transgenic mice. FABP4-ghrelin transgenic mice are resistant to obesity induced by high-fat diet. Brown fat mass was not affected by overexpression of ghrelin in adipose tissue. Glucose tolerance tests showed glucose levels to be significantly lower in FABP4-ghrelin transgenic mice than in controls after glucose administration. Insulin sensitivity testing showed that FABP4-ghrelin transgenic mice had a 28 +/- 5% greater hypoglycemic response to insulin. Our study demonstrates that overexpression of ghrelin from the FABP4 promoter impairs the development of white adipose tissues, and alters glucose tolerance and insulin sensitivity in mice.


Asunto(s)
Adiposidad/etnología , Metabolismo Energético/genética , Ghrelina/genética , Glucosa/metabolismo , Tejido Adiposo/anatomía & histología , Tejido Adiposo/metabolismo , Animales , Peso Corporal/genética , Ingestión de Alimentos/genética , Proteínas de Unión a Ácidos Grasos/genética , Ghrelina/metabolismo , Intolerancia a la Glucosa/genética , Insulina/sangre , Resistencia a la Insulina/genética , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas
10.
Cell Signal ; 19(6): 1185-92, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17276034

RESUMEN

Asb-4 is a gene that is specifically expressed in the hypothalamic energy homeostasis-associated areas and is down-regulated in the arcuate nucleus of fasted Sprague Dawley and obese Zucker rats. It has two functional domains, the ankyrin repeat and the SOCS box. The function of Asb-4 is unclear. We used yeast two hybridization to search for protein(s) that interact with Asb-4. With Asb-4 minus its SOCS box (Asb-4/Deltasb) as a bait, we screened mouse testis and arcuate nucleus cDNA libraries and identified G-protein pathway suppressor 1 (GPS1, also known as CSN1) as an Asb-4 interacting protein. GPS1 co-immunoprecipitated with Asb-4 both in vitro and in human HEK293 cells. When Asb-4 and GPS1 were co-transfected into HEK293 cells, expression of Asb-4 reduced the protein level of GPS1. Deletion of the SOCS box (Asb4/Deltasb) did not abolish the inhibitory effect of Asb-4 on GPS1, indicating that the SOCS box was not needed for its inhibitory effect. In NIH 3T3 L1 cells, expression of GPS1 enhanced c-Jun NH2-terminal kinase (JNK) activity. Co-expression of Asb-4 with GPS1 inhibited JNK activity. Treatment of the cells with insulin (20 nM) stimulated JNK activity. Expression of GPS1 potentiated the stimulatory effect of insulin, whereas co-expression of Asb-4 along with GPS1 inhibited JNK activity. In HEK293 cells expression of GPS1 elevated phosphorylation of insulin receptor substrate 1 (IRS-1) at serine307, co-expression of Asb-4 with GPS1 reduced the IRS-1ser307 phosphorylation. The present study demonstrates that Asb-4 interacts with GPS1 and inhibits JNK activity.


Asunto(s)
Repetición de Anquirina , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Línea Celular , Humanos , Inmunoprecipitación , Proteínas Sustrato del Receptor de Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Neuronas/citología , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo
11.
Diabetes ; 67(2): 265-277, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180353

RESUMEN

Mitophagy is a cellular quality-control pathway, which is essential for elimination of unhealthy mitochondria. While mitophagy is critical to pancreatic ß-cell function, the posttranslational signals governing ß-cell mitochondrial turnover are unknown. Here, we report that ubiquitination is essential for the assembly of a mitophagy regulatory complex, comprised of the E3 ligase Nrdp1, the deubiquitinase enzyme USP8, and Clec16a, a mediator of ß-cell mitophagy with unclear function. We discover that the diabetes gene Clec16a encodes an E3 ligase, which promotes nondegradative ubiquitin conjugates to direct its mitophagy effectors and stabilize the Clec16a-Nrdp1-USP8 complex. Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new-onset diabetes, impairs ß-cell mitophagy, oxygen consumption, and insulin secretion. Indeed, patients treated with lenalidomide develop compromised ß-cell function. Moreover, the ß-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Thus, the Clec16a-Nrdp1-USP8 complex relies on ubiquitin signals to promote mitophagy and maintain mitochondrial quality control necessary for optimal ß-cell function.


Asunto(s)
Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/metabolismo , Mitofagia , Proteínas de Transporte de Monosacáridos/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular , Células Cultivadas , Cruzamientos Genéticos , Endopeptidasas/química , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/química , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitofagia/efectos de los fármacos , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/genética , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Bancos de Tejidos , Técnicas de Cultivo de Tejidos , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/genética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos
12.
Regul Pept ; 139(1-3): 115-21, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17188372

RESUMEN

HEK 293 cells stably expressing human melanocortin-3 receptor (MC3R) were exposed to melanocortin receptor agonist, NDP-MSH (10(-)(10)-10(-)(6) M). ERK1/2 was phosphorylated in a dose-dependent manner with an EC(50) of 3.3+/-1.5 x 10(-)(9) M, similar to the IC(50) of NDP-MSH binding to the MC3R. ERK1/2 phosphorylation was blocked by the melanocortin receptor antagonists SHU9119. NDP-MSH-induced ERK1/2 phosphorylation was sensitive to pertussis toxin and the PI3K inhibitor, wortmannin. Rp-cAMPS, BAPTA-AM and Myr-PKC did not inhibit the NDP-MSH-induced ERK1/2 phosphorylation. NDP-MSH stimulated cellular proliferation in a dose-dependent manner with a similar EC(50) to ERK1/2 phosphorylation, 2.1+/-0.6 x 10(-)(9) M. Cellular proliferation was blocked by AGRP (86-132) and by the MEK inhibitor, PD98059. The NDP-MSH did not inhibit serum deprivation-induced apoptosis. MC3R activation induces ERK1/2 phosphorylation via PI3K and this pathway is involved in cellular proliferation in HEK cells expressing MC3R.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Melanocortina Tipo 3/fisiología , Proteína Relacionada con Agouti , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación/efectos de los fármacos , Receptor de Melanocortina Tipo 3/antagonistas & inhibidores , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
13.
Chem Biol ; 13(12): 1297-305, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17185225

RESUMEN

Agouti (ASIP) and Agouti-related protein (AgRP) are endogenous antagonists of melanocortin receptors that play critical roles in the regulation of pigmentation and energy balance, respectively, and which arose from a common ancestral gene early in vertebrate evolution. The N-terminal domain of ASIP facilitates antagonism by binding to an accessory receptor, but here we show that the N-terminal domain of AgRP has the opposite effect and acts as a prodomain that negatively regulates antagonist function. Computational analysis reveals similar patterns of evolutionary constraint in the ASIP and AgRP C-terminal domains, but fundamental differences between the N-terminal domains. These studies shed light on the relationships between regulation of pigmentation and body weight, and they illustrate how evolutionary structure function analysis can reveal both unique and common mechanisms of action for paralogous gene products.


Asunto(s)
Evolución Molecular , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína de Señalización Agouti , Proteína Relacionada con Agouti , Secuencia de Aminoácidos , Animales , Biología Computacional , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia
14.
Mol Biol Cell ; 15(5): 2484-91, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15034137

RESUMEN

Ghrelin, a novel gastric hormone, regulates food intake and energy metabolism via central mechanisms. The peripheral effect of ghrelin on adiposity is poorly understood. We established a stable 3T3-L1 cell line expressing ghrelin to study the direct effect of ghrelin on adipogenesis. Cells overexpressing ghrelin demonstrate significantly attenuated differentiation of preadipocytes into adipocytes. Expression of peroxisome proliferator-activator receptor-gamma is significantly inhibited as demonstrated by decrease of peroxisome proliferator-activator receptor-gamma mRNA and protein. Both ghrelin overexpression and exogenous ghrelin stimulate cell proliferation. Phosphorylation of mitogen-activated protein kinase is increased after treatment of cells with ghrelin. Ghrelin binding activity is demonstrated in both native and ghrelin-overexpressing 3T3-L1 cells by radiolabeled ghrelin, although reverse transcription-polymerase chain reaction with the primer sequence of the previously identified ghrelin receptor subtypes detected no signal. Our results demonstrate that ghrelin inhibits adipogenesis by stimulation of cell proliferation via the mediation of a ghrelin receptor, likely a novel unidentified subtype.


Asunto(s)
Adipocitos/citología , Hormonas Peptídicas/metabolismo , Adipocitos/metabolismo , Animales , Unión Competitiva , Bromodesoxiuridina/farmacocinética , Diferenciación Celular , Línea Celular , Citoplasma/ultraestructura , Regulación hacia Abajo , Activación Enzimática , Ghrelina , Lípidos/biosíntesis , Lípidos/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Hormonas Peptídicas/genética , Fosforilación , Unión Proteica , ARN Mensajero/análisis , Ensayo de Unión Radioligante , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ghrelina , Fase S/genética , Transfección
15.
J Mol Biol ; 346(4): 1059-70, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15701517

RESUMEN

Expression of the agouti signaling protein (ASIP) during hair growth produces the red/yellow pigment pheomelanin. ASIP, and its neuropeptide homolog the agouti-related protein (AgRP) involved in energy balance, are novel, paracrine signaling molecules that act as inverse agonists at distinct subsets of melanocortin receptors. Ubiquitous ASIP expression in mice gives rise to a pleiotropic phenotype characterized by a uniform yellow coat color, obesity, overgrowth, and metabolic derangements similar to type II diabetes in humans. Here we report the synthesis and NMR structure of ASIP's active, cysteine-rich, C-terminal domain. ASIP adopts the inhibitor cystine knot fold and, along with AgRP, are the only known mammalian proteins in this structure class. Moreover, ASIP populates two distinct conformers resulting from a cis peptide bond at Pro102-Pro103 and a coexistence of cis/trans isomers of Ala104-Pro105. Pharmacologic studies of Pro-->Ala mutants demonstrate that the minor conformation with two cis peptide bonds is responsible for activity at all MCRs. The loop containing the heterogeneous Ala-Pro peptide bond is conserved in mammals, and suggests that ASIP is either trapped by evolution in this unusual configuration or possesses function outside of strict MCR antagonism.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/química , Fragmentos de Péptidos/química , Proteína de Señalización Agouti , Proteína Relacionada con Agouti , Secuencia de Aminoácidos , Animales , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Pliegue de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
16.
Peptides ; 27(11): 2846-57, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16806584

RESUMEN

The melanocortin-4 receptor (MC4R) is a seven transmembrane member of the melanocortin receptor family. The GT1-1 cell line exhibits endogenous expression of MC4R. In this study, GT1-1 cells were used to study MC4R signaling pathways and to examine the effects of melanocortin receptor agonist NDP-MSH on apoptosis. MC4R mRNA expression was demonstrated by RT-PCR. Functional melanocortin receptor expression was implied by specific binding of NDP-MSH and cAMP production. NDP-MSH-stimulated GnRH release in a dose-dependent manner. Serum deprivation-induced apoptosis in GT1-1 cells, and the NDP-MSH inhibited this effect. The melanocortin receptor antagonist SHU9119 blocked the antiapoptotic actions of NDP-MSH, and the MAP kinase inhibitor PD98059 significantly attenuated the antiapoptotic effect. NDP-MSH-stimulated ERK1/2 phosphorylation in a dose-dependent manner. ERK1/2 phosphorylation could be abolished by SHU9119. In GT1-1 cells, melanocortin receptor activation causes ERK1/2 phosphorylation. In these cells, MC4R activation is also associated with antiapoptotic effects.


Asunto(s)
Apoptosis , Hipotálamo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Neuronas/metabolismo , Receptor de Melanocortina Tipo 4/fisiología , Animales , Línea Celular Transformada , Línea Celular Tumoral , Humanos , Hipotálamo/citología , Ratones , Neuronas/citología , Transducción de Señal
17.
Peptides ; 26(10): 1978-87, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16009463

RESUMEN

The specific melanocortin receptors, MC3R and MC4R, are directly linked to metabolism and body weight control. These receptors are activated by the peptide hormone alpha-MSH and antagonized by the agouti-related protein (AGRP). Whereas alpha-MSH acts broadly on most members of the MCR family (with the exception of MC2R), AGRP is highly specific for only MC3R and MC4R. AGRP is a complex ligand of approximately 100 amino acids. Within AGRP, MCR recognition and antagonism is localized to a 34 residue, cysteine-rich domain that adopts an inhibitor cystine knot (ICK) fold. An oxidatively folded peptide corresponding to this domain, referred to as mini-AGRP, exhibits full antagonist function and selectivity for MC3R and MC4R. Here we investigate a series of chimera proteins based on the mini-AGRP scaffold. Amino acid sequences derived from peptide agonists are grafted into the mini-AGRP active loop, implicated in receptor recognition, with the goal of producing ICK based agonists specific for MC3R and MC4R. Several constructs indeed exhibited potent agonist activity; however, with all chimeras, receptor selectivity is significantly altered. Pharmacologic data indicate that the chimeras do not interact with MC receptors through native AGRP like contacts. A model to explain the data suggest that there is only partial overlap of the agonist versus antagonist binding surfaces within MC receptors. Moreover, accessibility to the binding pocket is highly receptor specific with MC3R being the least tolerant of ligand alterations.


Asunto(s)
Proteína Relacionada con Agouti/genética , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/fisiología , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Arginina/genética , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Receptores de Melanocortina/metabolismo , Receptores de Melanocortina/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/fisiología , alfa-MSH/genética
18.
Neurosci Lett ; 595: 7-11, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25841790

RESUMEN

The mechanisms underlying alterations in brain functions in response to physical exercise are not fully understood. The present study examined the central effect of irisin, a 112 amino acid polypeptide hormone secreted from the skeletal muscle after exercise, on the locomotion in rats. Central administration of irisin significantly increased the locomotion. Relative to control animals treated with IgG Fc peptide, rats receiving irisin demonstrated a marked increase in total travel distance, ambulatory counts and time, and vertical counts and time. These changes were associated with a significant decrease in resting time. Central treatment of irisin also induced significant increases in oxygen consumption, carbon dioxide production and heat production, indicating an increase in metabolic activity. Our study suggests that physical activity may signal to the central nervous system to coordinate locomotion with metabolic activity via irisin.


Asunto(s)
Fibronectinas/farmacología , Actividad Motora/efectos de los fármacos , Animales , Dióxido de Carbono/metabolismo , Fibronectinas/metabolismo , Inyecciones Intraventriculares , Locomoción/efectos de los fármacos , Masculino , Consumo de Oxígeno/efectos de los fármacos , Ratas Sprague-Dawley
19.
Peptides ; 24(4): 603-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12860205

RESUMEN

Agouti and agouti-related protein (AgRP) are endogenous antagonists of the melanocortin receptors (MCxR). Previous data showed that recombinant full-length agouti and a synthetic fragment of AgRP, AgRP (83-132), are inverse agonists at the MC1R and MC4R, respectively. This study demonstrates the smaller analogs AgRP (87-120) and ASIP [90-132 (L89Y)], and short peptides Yc[CRFFNAFC]Y and Qc[CRFFRSAC]S are also MC4R inverse agonists. Furthermore, the relative affinity of the series of MC4R ligands for displacement of radiolabeled antagonist 125I-AgRP (86-132) versus radiolabeled agonist 125I-NDP-MSH did not correlate with ligand efficacy, which is more consistent with an induced-fit model than a simple two-state model of MC4R activation. These data shed new light on the determinants and mechanism of inverse agonism at the MC4R.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas/fisiología , beta-MSH/análogos & derivados , Proteína de Señalización Agouti , Proteína Relacionada con Agouti , Secuencias de Aminoácidos , Línea Celular , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Hipotálamo/patología , Cinética , Ligandos , Hormonas Estimuladoras de los Melanocitos/farmacología , Modelos Moleculares , Obesidad , Péptidos/química , Péptidos Cíclicos/farmacología , Pigmentación , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Melanocortina/antagonistas & inhibidores , Receptores de Melanocortina/química , Proteínas Recombinantes/química , alfa-MSH/metabolismo , beta-MSH/farmacología
20.
Zhongguo Zhong Yao Za Zhi ; 29(4): 366-70, 2004 Apr.
Artículo en Zh | MEDLINE | ID: mdl-15706880

RESUMEN

OBJECTIVE: To study the modulatory effect of Panax gingseng and coadministration with Veratrum nigrum on the activity and mRNA expression of cytochrome P450 isoenzymes in rat liver. METHOD: Rat liver microsomal cytochrome P450, b5, aminopyrine N-demethylase(APND), p-nitrophenol-hydroxylase(pNPH)activities were quantitated by UV chromatography. The mRNA expression level of five CYP isoenzymes CYP1A1, CYP2B1/2, CYP2C11, CYP2E1 and CYP3A1 were detected by semi-quantitative reverse transcriptase-polymerase chain reaction(RT-PCR). RESULT: P. gingseng coadministrated with V. nigrum obviously decreased the P450 contents of liver microsomes, and the b5 contents. Both single and combined used inhibited the activities of aminopyrine N-demethylase. At the mRNA level, the expression of CYP2C11 markedly induced exposure to V. nigrum, but combinative groups decreased the expression of CYP2C11. The combination of P. gingseng and V. nigrum induced the expression of CYP1A1. P. gingseng has inhibitory effect on CYP2B1/2 and inductive effect used with V. nigrum. The combination of P. gingseng with V. nigrum also induced the expression of CYP3A1. CONCLUSION: P. gingseng used singly has some different modulation effects compared with combinative used, which may occur because of drug-drug interaction based on cytochrome P450. To elucidate the drug-drug interaction, it needs further analysis and metabolism research.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Medicamentos Herbarios Chinos/farmacología , Microsomas Hepáticos/metabolismo , Panax , Veratrum , Aminopirina N-Demetilasa/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Citocromos b5/metabolismo , Incompatibilidad de Medicamentos , Medicamentos Herbarios Chinos/aislamiento & purificación , Femenino , Técnicas In Vitro , Isoenzimas/biosíntesis , Isoenzimas/genética , Masculino , Panax/química , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Veratrum/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA