Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Gerontol A Biol Sci Med Sci ; 78(Suppl 1): 38-43, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37325967

RESUMEN

The discovery of the growth hormone secretagogues (GHS) and the reverse pharmacology leading to the discovery of GHS receptor which enabled the identification of ghrelin as the natural ligand for the receptor have opened a new horizon in growth hormone (GH) physiology, pathophysiology, and therapeutics. Major progress has been made and we now have orally active GHS which are able to restore optimal pulsatile GH secretion which cannot be overstimulated as insulin-like growth factor feedback regulates the peaks to the optimum level. This enables GH to be restored in the older to levels normally seen in 20- to 30-year-old people; this leads to an increase in fat-free mass and redistribution of fat to the limbs. As these agents are ultimately approved and investigated further, it is likely that they will be shown to restore growth in children with moderate-to-mild GH deficiency; their benefits will be investigated in other indications such as nonalcoholic fatty liver disease, frailty, anemia, osteoporosis, and immune compromise in older subjects. The exquisite regulation of GH secretion reflects the importance of GH pulsatility in the regulation of somatotroph action of GH.


Asunto(s)
Ghrelina , Hormona de Crecimiento Humana , Anciano , Humanos , Hormona del Crecimiento , Hormona Liberadora de Hormona del Crecimiento/farmacología , Hormona Liberadora de Hormona del Crecimiento/fisiología , Hormona de Crecimiento Humana/metabolismo , Hormona de Crecimiento Humana/uso terapéutico , Secretagogos , Adulto Joven
2.
J Transl Int Med ; 10(2): 146-155, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35959447

RESUMEN

Background and Objectives: Activation of ghrelin receptor growth hormone secretagogue receptor (GHS-R) by endogenous or synthetic ligands amplifies pulsatile release of growth hormone (GH) and enhances food intake, very relevant to development and growth. GHS-R is a G-protein coupled receptor that has great druggable potential. Understanding the precise ligand and receptor interactions is crucial to advance the application of GHS-R. Materials and Methods: We used radiolabeled ligand-binding assay and growth hormone release assay to assess the binding and functional characteristics of GHS-R to synthetic agonists MK-0677 and GHS-25, as well as to endogenous peptide ligand ghrelin. We analyzed the ligand-dependent activity of GHS-R by measuring aequorin-based [Ca++]i responses. To define a ligand-binding pocket of GHS-R, we generated a series of human/puffer fish GHS-R chimeras by domain swapping, as well as a series of mutants by site-directed mutagenesis. Results: We found that the synthetic ligands have high binding affinity to GHS-R in the in vitro competitive binding assay. Remarkably, the in vivo GH secretagogue activity is higher with the synthetic agonists MK-0677 and GHS-25 than that of ghrelin. Importantly, the activity was completely abolished in GHS-R knockout mice. In GHS-R chimera analysis, we identified the C-terminal region, particularly the transmembrane domain 6 (TM6), to be critical for the ligand-dependent activity. Our site-directed mutagenesis study further revealed that amino acid residues D99 and W276 in GHS-R are essential for ligand binding. Interestingly, critical residues distinctively interact with different ligands, MK-0677 activation depends on E124, while ghrelin and GHS-25 preferentially interact with F279. Conclusion: The ligand-binding pocket of human GHS-R is mainly defined by interactive residues in TM6 and the adjacent region of the receptor. This novel finding in GHS-R binding domains advances the structural/ functional understanding of GHS-R, which will help to select/design better GHS-R agonists/ antagonists for future therapeutic applications.

3.
Int J Mol Sci ; 22(8)2021 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-33920473

RESUMEN

Growth hormone secretagogue receptor (GHS-R) is widely known to regulate food intake and adiposity, but its role in glucose homeostasis is unclear. In this study, we investigated the expression of GHS-R in mouse pancreatic islets and its role in glycemic regulation. We used Ghsr-IRES-tauGFP mice, with Green Fluorescent Protein (GFP) as a surrogate for GHS-R, to demonstrate the GFP co-localization with insulin and glucagon expression in pancreatic islets, confirming GHS-R expression in ß and α cells. We then generated ß-cell-specific GHSR-deleted mice with MIP-Cre/ERT and validated that GHS-R suppression was restricted to the pancreatic islets. MIP-Cre/ERT;Ghsrf/f mice showed normal energy homeostasis with similar body weight, body composition, and indirect calorimetry profile. Interestingly, MIP-Cre/ERT;Ghsrf/f mice exhibited an impressive phenotype in glucose homeostasis. Compared to controls, MIP-Cre/ERT;Ghsrf/f mice showed lower fasting blood glucose and insulin; reduced first-phase insulin secretion during a glucose tolerance test (GTT) and glucose-stimulated insulin secretion (GSIS) test in vivo. The isolated pancreatic islets of MIP-Cre/ERT;Ghsrf/f mice also showed reduced insulin secretion during GSIS ex vivo. Further, MIP-Cre/ERT;Ghsrf/f mice exhibited improved insulin sensitivity during insulin tolerance tests (ITT). Overall, our results confirmed GHS-R expression in pancreatic ß and α cells; GHS-R cell-autonomously regulated GSIS and modulated systemic insulin sensitivity. In conclusion, ß cell GHS-R was an important regulator of glucose homeostasis, and GHS-R antagonists may have therapeutic potential for Type 2 Diabetes.


Asunto(s)
Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Ratones , Ratones Noqueados , Receptores de Ghrelina/genética
4.
Cell Metab ; 29(6): 1320-1333.e8, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31105045

RESUMEN

Endocannabinoids acting on the cannabinoid-1 receptor (CB1R) or ghrelin acting on its receptor (GHS-R1A) both promote alcohol-seeking behavior, but an interaction between the two signaling systems has not been explored. Here, we report that the peripheral CB1R inverse agonist JD5037 reduces ethanol drinking in wild-type mice but not in mice lacking CB1R, ghrelin peptide or GHS-R1A. JD5037 treatment of alcohol-drinking mice inhibits the formation of biologically active octanoyl-ghrelin without affecting its inactive precursor desacyl-ghrelin. In ghrelin-producing stomach cells, JD5037 reduced the level of the substrate octanoyl-carnitine generated from palmitoyl-carnitine by increasing fatty acid ß-oxidation. Blocking gastric vagal afferents abrogated the ability of either CB1R or GHS-R1A blockade to reduce ethanol drinking. We conclude that blocking CB1R in ghrelin-producing cells reduces alcohol drinking by inhibiting the formation of active ghrelin and its signaling via gastric vagal afferents. Thus, peripheral CB1R blockade may have therapeutic potential in the treatment of alcoholism.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Encéfalo/fisiología , Intestinos/fisiología , Receptor Cannabinoide CB1/genética , Aciltransferasas/genética , Aciltransferasas/fisiología , Consumo de Bebidas Alcohólicas/fisiopatología , Alcoholismo/genética , Alcoholismo/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Células Cultivadas , Eliminación de Gen , Ghrelina/metabolismo , Ghrelina/fisiología , Intestinos/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptores de Ghrelina/genética , Receptores de Ghrelina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonamidas/farmacología
5.
Curr Biol ; 26(22): 2992-3003, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27773571

RESUMEN

It is well recognized that G-protein-coupled receptors (GPCRs) can activate Ras-regulated kinase pathways to produce lasting changes in neuronal function. Mechanisms by which GPCRs transduce these signals and their relevance to brain disorders are not well understood. Here, we identify a major Ras regulator, neurofibromin 1 (NF1), as a direct effector of GPCR signaling via Gßγ subunits in the striatum. We find that binding of Gßγ to NF1 inhibits its ability to inactivate Ras. Deletion of NF1 in striatal neurons prevents the opioid-receptor-induced activation of Ras and eliminates its coupling to Akt-mTOR-signaling pathway. By acting in the striatal medium spiny neurons of the direct pathway, NF1 regulates opioid-induced changes in Ras activity, thereby sensitizing mice to psychomotor and rewarding effects of morphine. These results delineate a novel mechanism of GPCR signaling to Ras pathways and establish a critical role of NF1 in opioid addiction.


Asunto(s)
Analgésicos Opioides/metabolismo , Neurofibromina 1/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Animales , Femenino , Masculino , Ratones , Neostriado/metabolismo , Neurofibromina 1/metabolismo , Neuronas/metabolismo , Unión Proteica
6.
Sci Signal ; 9(424): fs6, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27095592

RESUMEN

The gut-derived hormone ghrelin regulates growth hormone release, appetite, metabolism, and immune function through its receptor, the growth hormone secretagogue receptor 1a (GHSR1a). In this issue of Science Signaling, Chebani et al decode GHSR1a signaling by using transgenic rats with a mutation in GHSR1a that prevents its interaction with ß-arrestin. These mutant rats are more responsive to endogenous ghrelin, resulting in increased fat deposition and insulin resistance without affecting food intake.


Asunto(s)
Ghrelina , Receptores de Ghrelina , Animales , Proteínas de Unión al GTP/metabolismo , Ratas , Ratas Transgénicas , Transducción de Señal
7.
Endocrinology ; 157(4): 1430-42, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26812158

RESUMEN

Aging is associated with attenuated ghrelin signaling. During aging, chronic caloric restriction (CR) produces health benefits accompanied by enhanced ghrelin production. Ghrelin receptor (GH secretagogue receptor 1a) agonists administered to aging rodents and humans restore the young adult phenotype; therefore, we tested the hypothesis that the metabolic benefits of CR are mediated by endogenous ghrelin. Three month-old male mice lacking ghrelin (Ghrelin-/-) or ghrelin receptor (Ghsr-/-), and their wild-type (WT) littermates were randomly assigned to 2 groups: ad libitum (AL) fed and CR, where 40% food restriction was introduced gradually to allow Ghrelin-/- and Ghsr-/- mice to metabolically adapt and avoid severe hypoglycemia. Twelve months later, plasma ghrelin, metabolic parameters, ambulatory activity, hypothalamic and liver gene expression, as well as body composition were measured. CR increased plasma ghrelin and des-acyl ghrelin concentrations in WT and Ghsr-/- mice. CR of WT, Ghsr-/-, and Ghrelin-/- mice markedly improved metabolic flexibility, enhanced ambulatory activity, and reduced adiposity. Inactivation of Ghrelin or Ghsr had no effect on AL food intake or food anticipatory behavior. In contrast to the widely held belief that endogenous ghrelin regulates food intake, CR increased expression of hypothalamic Agrp and Npy, with reduced expression of Pomc across genotypes. In the AL context, ablation of ghrelin signaling markedly inhibited liver steatosis, which correlated with reduced Pparγ expression and enhanced Irs2 expression. Although CR and administration of GH secretagogue receptor 1a agonists both benefit the aging phenotype, we conclude the benefits of chronic CR are a consequence of enhanced metabolic flexibility independent of endogenous ghrelin or des-acyl ghrelin signaling.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Restricción Calórica , Ghrelina/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Factores de Edad , Proteína Relacionada con Agouti/genética , Animales , Composición Corporal/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Expresión Génica , Ghrelina/sangre , Ghrelina/genética , Humanos , Hipotálamo/citología , Hipotálamo/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptido Y/genética , Distribución Aleatoria , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
8.
Cell ; 163(5): 1176-1190, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590421

RESUMEN

The ghrelin receptor (GHSR1a) and dopamine receptor-1 (DRD1) are coexpressed in hippocampal neurons, yet ghrelin is undetectable in the hippocampus; therefore, we sought a function for apo-GHSR1a. Real-time single-molecule analysis on hippocampal neurons revealed dimerization between apo-GHSR1a and DRD1 that is enhanced by DRD1 agonism. In addition, proximity measurements support formation of preassembled apo-GHSR1a:DRD1:Gαq heteromeric complexes in hippocampal neurons. Activation by a DRD1 agonist produced non-canonical signal transduction via Gαq-PLC-IP3-Ca(2+) at the expense of canonical DRD1 Gαs cAMP signaling to result in CaMKII activation, glutamate receptor exocytosis, synaptic reorganization, and expression of early markers of hippocampal synaptic plasticity. Remarkably, this pathway is blocked by genetic or pharmacological inactivation of GHSR1a. In mice, GHSR1a inactivation inhibits DRD1-mediated hippocampal behavior and memory. Our findings identify a previously unrecognized mechanism essential for DRD1 initiation of hippocampal synaptic plasticity that is dependent on GHSR1a, and independent of cAMP signaling.


Asunto(s)
Dopamina/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Hipocampo/metabolismo , Memoria , Ratones , Plasticidad Neuronal , Receptores de Dopamina D1/agonistas
9.
Psychopharmacology (Berl) ; 232(23): 4285-92, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26319159

RESUMEN

RATIONALE: Schizophrenic-spectrum patients commonly display deficits in preattentive information processing as evidenced, for example, by disrupted prepulse inhibition (PPI), a measure of sensorimotor gating. Similar disruptions in PPI can be induced in rodents and primates by the psychotomimetic drug phencyclidine (PCP), a noncompetitive inhibitor of the NMDA receptor. Mounting evidence suggests that the hunger hormone ghrelin and its constitutively active receptor influences neuronal circuits involved in the regulation of mood and cognition. OBJECTIVES: In the present series of experiments, we investigated the effects of ghrelin and the growth hormone secretagogue receptor (GHS-R1A) neutral antagonist, JMV 2959, on acoustic startle responses (ASR), PPI, and PCP-induced alterations in PPI. RESULTS: Intraperitoneal (i.p.) administration of ghrelin (0.033, 0.1, and 0.33 mg/kg) did not alter the ASR or PPI in rats. Conversely, i.p. injection of JMV 2959 (1, 3, and 6 mg/kg), dose dependently decreased the ASR and increased PPI. Pretreatment with JMV 2959 at a dose with no effect on ASR or PPI per se, completely blocked PCP-induced (2 mg/kg) deficits in PPI while pretreatment with the highest dose of ghrelin did not potentiate or alter PPI responses of a sub-threshold dose of PCP (0.75 mg/kg). CONCLUSION: These findings indicate that the GHS-R1A is involved in specific behavioral effects of PCP and may have relevance for patients with schizophrenia.


Asunto(s)
Glicina/análogos & derivados , Fenciclidina/farmacología , Inhibición Prepulso/efectos de los fármacos , Receptores de Ghrelina/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Triazoles/farmacología , Animales , Relación Dosis-Respuesta a Droga , Glicina/farmacología , Masculino , Fenciclidina/antagonistas & inhibidores , Inhibición Prepulso/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Esquizofrenia/tratamiento farmacológico , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
10.
Biol Reprod ; 93(1): 24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26019260

RESUMEN

Cisplatin administration induces DNA damage resulting in germ cell apoptosis and subsequent testicular atrophy. Although 50 percent of male cancer patients receiving cisplatin-based chemotherapy develop long-term secondary infertility, medical treatment to prevent spermatogenic failure after chemotherapy is not available. Under normal conditions, testicular p53 promotes cell cycle arrest, which allows time for DNA repair and reshuffling during meiosis. However, its role in the setting of cisplatin-induced infertility has not been studied. Ghrelin administration ameliorates the spermatogenic failure that follows cisplatin administration in mice, but the mechanisms mediating these effects have not been well established. The aim of the current study was to characterize the mechanisms of ghrelin and p53 action in the testis after cisplatin-induced testicular damage. Here we show that cisplatin induces germ cell damage through inhibition of p53-dependent DNA repair mechanisms involving gamma-H2AX and ataxia telangiectasia mutated protein kinase. As a result, testicular weight and sperm count and motility were decreased with an associated increase in sperm DNA damage. Ghrelin administration prevented these sequelae by restoring the normal expression of gamma-H2AX, ataxia telangiectasia mutated, and p53, which in turn allows repair of DNA double stranded breaks. In conclusion, these findings indicate that ghrelin has the potential to prevent or diminish infertility caused by cisplatin and other chemotherapeutic agents by restoring p53-dependent DNA repair mechanisms.


Asunto(s)
Cisplatino/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Ghrelina/farmacología , Testículo/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Testículo/metabolismo , Proteína p53 Supresora de Tumor/genética
11.
Biol Reprod ; 92(3): 76, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25631345

RESUMEN

The chemotherapeutic drug cisplatin causes a number of dose-dependent side effects, including cachexia and testicular damage. Patients receiving a high cumulative dose of cisplatin may develop permanent azoospermia and subsequent infertility. Thus, the development of chemotherapeutic regimens with the optimal postsurvival quality of life (fertility) is of high importance. This study tested the hypothesis that ghrelin administration can prevent or minimize cisplatin-induced testicular damage and cachexia. Ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR-1a), are expressed and function in the testis. Targeted deletion of ghrelin, or its receptor, significantly increases the rate of cell death in the testis, suggesting a protective role. Intraperitoneal administration of vehicle, ghrelin, or cisplatin alone or in combination with ghrelin, in cycles of 9 or 18 days, to adult male C57Bl/6 mice was performed. Body weight was measured daily and testicular and epididymal weight, sperm density and motility, testicular histology, and testicular cell death were analyzed at the time of euthanization. Ghrelin coadministration decreased the severity of cisplatin-induced cachexia and gonadal toxicity. Body, testicular, and epididymal weights significantly increased as testicular cell death decreased with ghrelin coadministration. The widespread damage to the seminiferous epithelium induced by cisplatin administration was less severe in mice simultaneously treated with ghrelin. Furthermore, ghrelin diminished the deleterious effects of cisplatin on testis and body weight homeostasis in wild-type but not Ghsr(-/-) mice, showing that ghrelin's actions are mediated via GHSR. Ghrelin or more stable GHSR agonists potentially offer a novel therapeutic approach to minimize the testicular damage that occurs after gonadotoxin exposure.


Asunto(s)
Apoptosis/efectos de los fármacos , Caquexia/inducido químicamente , Caquexia/prevención & control , Cisplatino/efectos adversos , Cisplatino/farmacología , Ghrelina/fisiología , Receptores de Ghrelina/fisiología , Testículo/fisiopatología , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Peso Corporal/efectos de los fármacos , Caquexia/fisiopatología , Ghrelina/deficiencia , Ghrelina/farmacología , Homeostasis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Tamaño de los Órganos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/patología , Testículo/efectos de los fármacos
12.
Artículo en Inglés | MEDLINE | ID: mdl-25183960

RESUMEN

The orexigenic peptide hormone ghrelin is synthesized in the stomach and its receptor growth hormone secretagogue receptor (GHSR1a) is expressed mainly in the central nervous system (CNS). In this review, we confine our discussion to the physiological role of GHSR1a in the brain. Paradoxically, despite broad expression of GHSR1a in the CNS, other than trace amounts in the hypothalamus, ghrelin is undetectable in the brain. In our efforts to elucidate the function of the ligand-free ghrelin receptor (apo-GHSR1a), we identified subsets of neurons that co-express GHSR1a and dopamine receptors. In this review, we focus on interactions between apo-GHSR1a and dopamine-2 receptor (DRD2) and formation of GHSR1a:DRD2 heteromers in hypothalamic neurons that regulate appetite, and discuss implications for the treatment of Prader-Willi syndrome (PWS). GHSR1a antagonists of distinct chemical structures, a quinazolinone and a triazole, respectively, enhance and inhibit dopamine signaling through GHSR1a:DRD2 heteromers by an allosteric mechanism. This finding illustrates a potential strategy for designing the next generation of drugs for treating eating disorders as well as psychiatric disorders caused by abnormal dopamine signaling. Treatment with a GHSR1a antagonist that enhances dopamine/DRD2 activity in GHSR1a:DRD2 expressing hypothalamic neurons has the potential to inhibit the uncontrollable hyperphagia associated with PWS. DRD2 antagonists are prescribed for treating schizophrenia, but these block dopamine signaling in all DRD2 expressing neurons and are associated with adverse side effects, including enhanced appetite and excessive weight gain. A GHSR1a antagonist of structural class that allosterically blocks dopamine/DRD2 action in GHSR1a:DRD2 expressing neurons would have no effect on neurons expressing DRD2 alone; therefore, the side effects of DRD2 antagonists would potentially be reduced thereby enhancing patient compliance.

13.
Endocrinology ; 155(12): 4843-55, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25211592

RESUMEN

The stomach hormone ghrelin and hypothalamic melanocortin neurons belong to a gut-brain circuit controlling appetite and metabolic homeostasis. Mice lacking melanocortin-3 receptor (Mc3rKO) or growth hormone secretagogue receptor (GhsrKO) genes exhibit attenuated food anticipatory activity (FAA), a rise in locomotor activity anticipating mealtime, suggesting common circuitry regulating anticipatory responses to nutrient loading. To investigate the interaction between Ghsrs and Mc3rs, we compared food anticipatory responses in GhsrKO, Mc3rKO, and double Ghsr;Mc3r knockout (DKO) mice subjected to a hypocaloric restricted feeding (RF) protocol in constant dark or 12-hour light, 12-hour dark settings. DKO are viable, exhibiting no overt behavioral or metabolic phenotypes in ad libitum or fasting conditions. FAA was initially attenuated in all mutant strains in constant darkness. However, GhsrKO eventually exhibited a robust food anticipatory response, suggesting compensation. Mc3rKO and DKO did not compensate, indicating a continued requirement for Mc3rs in maintaining the expression of FAA in situations of RF. Abnormal regulation of hypothalamic agouti-related peptide/neuropeptide Y (AgRP/Npy) neurons previously observed during fasting may contribute to attenuated FAA in Mc3rKO. AgRP and Npy expression measured 1 hour before food presentation correlated positively with FAA. Absence of Mc3rs (but not Ghsrs) was associated with lower AgRP/Npy expression, suggesting attenuated responses to signals of negative energy balance. These observations support the importance of Mc3rs as modulators of anticipatory responses to feeding, with mice able to compensate for loss of Ghsrs. The behavioral deficits of Mc3rKO displayed during RF may be partially explained by reduced hunger sensations owing to abnormal regulation of orexigenic AgRP/Npy neurons.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Apetito/fisiología , Actividad Motora/fisiología , Receptor de Melanocortina Tipo 3/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Composición Corporal , Oscuridad , Metabolismo Energético , Privación de Alimentos , Genotipo , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Endocrinology ; 155(11): 4287-95, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25060361

RESUMEN

Ghrelin receptor-deficient (Ghsr-/-) mice that lack acylated ghrelin (AG) signaling retain a metabolic response to unacylated ghrelin (UAG). Recently, we showed that Ghsr-deficiency affects bone metabolism. The aim of this study was to further establish the impact of AG and UAG on bone metabolism. We compared bone metabolism in Ghsr-/- (lacking only AG signaling) and ghrelin-deficient (Ghrl-/-; both AG and UAG deficient) male mice. Ghrl-/- mice had lower cortical bone mass, whereas Ghsr-/- mice had lower trabecular bone mass. This demonstrates bone compartment-specific effects of AG and a role for UAG in bone metabolism. Also, Ghrl-/- but not Ghsr-/- mice had increased bone formation rate and increased osteogenic stem cell numbers in their bone marrow. In ex vivo bone marrow cultures both AG and UAG inhibited osteoblast differentiation. This indicated that bone resorption must be increased in these mice. Accordingly, osteoclastogenesis rate was faster in bone marrow cultures from Ghsr-/- and Ghrl-/- mice, and osteoclast formation was inhibited by AG signaling and partially suppressed by UAG. In osteoblast cultures, AG markedly induced osteoprotegerin gene expression and both peptides reduced RANKL/osteoprotegerin ratio. These data describe unique cell-type specific effects of AG and UAG within a single tissue, supporting a tight and complex control of bone formation and resorption as well as a link between nutrition and bone metabolism. The balance between AG and UAG actions in the bone marrow may lead to bone compartmental-specific effects.


Asunto(s)
Remodelación Ósea/genética , Huesos/metabolismo , Ghrelina/genética , Receptores de Ghrelina/genética , Acilación , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Remodelación Ósea/efectos de los fármacos , Huesos/anatomía & histología , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Células Cultivadas , Ghrelina/metabolismo , Ghrelina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Microtomografía por Rayos X
15.
Prog Brain Res ; 211: 183-200, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24968781

RESUMEN

Dopamine neurotransmission is traditionally accepted as occurring through the five dopamine receptors that transduce its signal. Recent evidence has demonstrated that the range of physiologically relevant dopamine signaling complexes is greatly expanded by the ability of dopamine receptors to interact with other dopamine receptors and with receptors of other endogenous signaling ligands. These novel heteromeric complexes have functional properties distinct from the component receptors or are able to modulate the canonical signaling and function of the cognate receptors. These dopamine receptor heteromers provide new insight into physiological mechanisms and pathophysiological processes involving dopamine.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Receptores Dopaminérgicos/metabolismo , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Animales , Humanos
16.
Arthritis Rheumatol ; 66(7): 1779-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24591481

RESUMEN

OBJECTIVE: Obesity is a major risk factor for the development of osteoarthritis (OA) that is associated with a state of low-grade inflammation and increased circulating levels of adipokines and free fatty acids (FFAs). The aim of this study was to analyze the effects of saturated (palmitate) and monounsaturated (oleate) FFAs on articular chondrocytes, synoviocytes, and cartilage. METHODS: Human articular chondrocytes and fibroblast-like synoviocytes obtained from young healthy donors and OA chondrocytes from patients undergoing total knee replacement surgery were treated with palmitate or oleate alone or in combination with interleukin-1ß (IL-1ß). Cell viability, caspase activation, and gene expression of proinflammatory factors, extracellular matrix (ECM) proteins, and proteases were studied. In addition, chondrocyte viability, IL-6 production, and matrix damage were assessed in bovine and human articular cartilage explants cultured with FFAs in the presence or absence of IL-1ß. RESULTS: Palmitate, but not oleate, induced caspase activation and cell death in IL-1ß-stimulated normal chondrocytes, and up-regulated the expression of IL-6 and cyclooxygenase 2 in chondrocytes and fibroblast-like synoviocytes through Toll-like receptor 4 (TLR-4) signaling. In cartilage explants, palmitate induced chondrocyte death, IL-6 release, and ECM degradation. Palmitate synergized with IL-1ß in stimulating proapoptotic and proinflammatory cellular responses. Pharmacologic inhibition of caspases or TLR-4 signaling reduced palmitate and IL-1ß induced cartilage damage. CONCLUSION: Palmitate acts as a proinflammatory and catabolic factor that, in synergy with IL-1ß, induces chondrocyte apoptosis and articular cartilage breakdown. Collectively, our data suggest that elevated levels of saturated FFAs that are often found in patients who are obese may contribute to the pathogenesis of OA.


Asunto(s)
Apoptosis/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Interleucina-1beta/farmacología , Osteoartritis de la Rodilla/tratamiento farmacológico , Palmitatos/farmacología , Adulto , Anciano , Animales , Apoptosis/inmunología , Cartílago Articular/inmunología , Bovinos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/inmunología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Sinergismo Farmacológico , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Ácidos Grasos no Esterificados/inmunología , Ácidos Grasos no Esterificados/farmacología , Humanos , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Persona de Mediana Edad , Ácido Oléico/inmunología , Ácido Oléico/farmacología , Osteoartritis de la Rodilla/inmunología , Palmitatos/inmunología , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Adulto Joven
17.
FASEB J ; 28(1): 364-72, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24081904

RESUMEN

A major limitation of exogenous vitamin D3 administration for the treatment of prostate cancer is the marginal, if any, clinical efficacy. We dissected the basis for the resistance to the vitamin D3 antitumor properties and specifically examined the effect of its major catabolic enzyme, CYP24A1, in prostate cancer. Local CYP24A1 expression levels and the effect of selective modulation were analyzed using tissue microarrays from needle core biopsy specimens and xenograft-bearing mouse models. CYP24A1 mRNA was elevated in malignant human prostate tissues compared to benign lesions. High CYP24A1 protein levels were seen in poorly differentiated and highly advanced stages of prostate cancer and correlated with parallel increase in the tumor proliferation rate. The use of CYP24A1 RNAi enhanced the cytostatic effects of vitamin D3 in human prostate cancer cells. Remarkably, subcutaneous and orthotopic xenografts of prostate cancer cells harboring CYP24A1 shRNA resulted in a drastic reduction in tumor volume when mice were subjected to vitamin D3 supplementation. CYP24A1 may be a predictive marker of vitamin D3 clinical efficacy in patients with advanced prostate cancer. For those with up-regulated CYP24A1, combination therapy with RNAi targeting CYP24A1 could be considered to improve clinical responsiveness to vitamin D3.


Asunto(s)
Colecalciferol/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Esteroide Hidroxilasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones SCID , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroide Hidroxilasas/genética , Vitamina D3 24-Hidroxilasa , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Endocrinology ; 154(9): 3118-29, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23832960

RESUMEN

Cachexia, defined as an involuntary weight loss ≥ 5%, is a serious and dose-limiting side effect of chemotherapy that decreases survival in cancer patients. Alterations in lipid metabolism are thought to cause the lipodystrophy commonly associated with cachexia. Ghrelin has been proposed to ameliorate the alterations in lipid metabolism due to its orexigenic and anabolic properties. However, the mechanisms of action through which ghrelin could potentially ameliorate chemotherapy-associated cachexia have not been elucidated. The objectives of this study were to identify mechanisms by which the chemotherapeutic agent cisplatin alters lipid metabolism and to establish the role of ghrelin in reversing cachexia. Cisplatin-induced weight and fat loss were prevented by ghrelin. Cisplatin increased markers of lipolysis in white adipose tissue (WAT) and of ß-oxidation in liver and WAT and suppressed lipogenesis in liver, WAT, and muscle. Ghrelin prevented the imbalance between lipolysis, ß-oxidation, and lipogenesis in WAT and muscle. Pair-feeding experiments demonstrated that the effects of cisplatin and ghrelin on lipogenesis, but not on lipolysis and ß-oxidation, were due to a reduction in food intake. Thus, ghrelin prevents cisplatin-induced weight and fat loss by restoring adipose tissue functionality. An increase in caloric intake further enhances the anabolic effects of ghrelin.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Antineoplásicos/efectos adversos , Estimulantes del Apetito/uso terapéutico , Caquexia/tratamiento farmacológico , Cisplatino/efectos adversos , Ghrelina/uso terapéutico , Lipólisis/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Adiposidad/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Caquexia/inducido químicamente , Caquexia/metabolismo , Caquexia/patología , Ingestión de Energía/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Distribución Aleatoria , Pérdida de Peso/efectos de los fármacos
19.
Endocr Dev ; 25: 5-15, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23652387

RESUMEN

The growth hormone (GH) secretagogue receptor (GHS-R1a) is a G protein-coupled receptor (GPCR) expressed in the brain as well as other areas of the body. In the early 1990s, this receptor was expression cloned in MERCK laboratories by using a group of synthesized small molecules known to increase GH release in humans and other animals. Since its discovery, hundreds of studies have shown the importance of this receptor and its endogenous ligand, ghrelin, in metabolism, neurotransmission, and behavior. Even more relevant are the prospective benefits that will result from pharmacologic manipulation of GHS-R1a. Multiple GHS-R1a agonists and antagonists are available for experimentation, and some have been used in patients with promising results. Studies in rodents have revealed intriguing potential roles for GHS-R1a modulation. Our goal in this chapter is to connect these studies with the inherent advantages of targeting this receptor pharmacologically.


Asunto(s)
Receptores de Ghrelina/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiología , Ghrelina/metabolismo , Ghrelina/farmacología , Glucosa/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Obesidad/genética , Obesidad/metabolismo , Multimerización de Proteína/genética , Multimerización de Proteína/fisiología , Receptores de Ghrelina/genética , Receptores de Ghrelina/historia , Receptores de Ghrelina/metabolismo , Transducción de Señal
20.
J Clin Invest ; 123(2): 611-22, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23281394

RESUMEN

Cachexia is a wasting syndrome associated with cancer, AIDS, multiple sclerosis, and several other disease states. It is characterized by weight loss, fatigue, loss of appetite, and skeletal muscle atrophy and is associated with poor patient prognosis, making it an important treatment target. Ghrelin is a peptide hormone that stimulates growth hormone (GH) release and positive energy balance through binding to the receptor GHSR-1a. Only acylated ghrelin (AG), but not the unacylated form (UnAG), can bind GHSR-1a; however, UnAG and AG share several GHSR-1a-independent biological activities. Here we investigated whether UnAG and AG could protect against skeletal muscle atrophy in a GHSR-1a-independent manner. We found that both AG and UnAG inhibited dexamethasone-induced skeletal muscle atrophy and atrogene expression through PI3Kß-, mTORC2-, and p38-mediated pathways in myotubes. Upregulation of circulating UnAG in mice impaired skeletal muscle atrophy induced by either fasting or denervation without stimulating muscle hypertrophy and GHSR-1a-mediated activation of the GH/IGF-1 axis. In Ghsr-deficient mice, both AG and UnAG induced phosphorylation of Akt in skeletal muscle and impaired fasting-induced atrophy. These results demonstrate that AG and UnAG act on a common, unidentified receptor to block skeletal muscle atrophy in a GH-independent manner.


Asunto(s)
Ghrelina/química , Ghrelina/farmacología , Atrofia Muscular/prevención & control , Acilación , Animales , Caquexia/metabolismo , Caquexia/prevención & control , Línea Celular , Ghrelina/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos/metabolismo , Desnervación Muscular , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA