Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 18(1): 304, 2021 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-34961526

RESUMEN

BACKGROUND: Neuroinflammation is a key pathological component of neurodegenerative disease and is characterized by microglial activation and the secretion of proinflammatory mediators. We previously reported that a surge in prostaglandin D2 (PGD2) production and PGD2-induced microglial activation could provoke neuroinflammation. We also reported that a lipid sensor GPR120 (free fatty acid receptor 4), which is expressed in intestine, could be activated by polyunsaturated fatty acids (PUFA), thereby mediating secretion of glucagon-like peptide-1 (GLP-1). Dysfunction of GPR120 results in obesity in both mice and humans. METHODS: To reveal the relationship between PGD2-microglia-provoked neuroinflammation and intestinal PUFA/GPR120 signaling, we investigated neuroinflammation and neuronal function with gene and protein expression, histological, and behavioral analysis in GPR120 knockout (KO) mice. RESULTS: In the current study, we discovered notable neuroinflammation (increased PGD2 production and microglial activation) and neurodegeneration (declines in neurogenesis, hippocampal volume, and cognitive function) in GPR120 KO mice. We also found that Hematopoietic-prostaglandin D synthase (H-PGDS) was expressed in microglia, microglia were activated by PGD2, H-PGDS expression was upregulated in GPR120 KO hippocampus, and inhibition of PGD2 production attenuated this neuroinflammation. GPR120 KO mice exhibited reduced intestinal, plasma, and intracerebral GLP-1 contents. Peripheral administration of a GLP-1 analogue, liraglutide, reduced PGD2-microglia-provoked neuroinflammation and further neurodegeneration in GPR120 KO mice. CONCLUSIONS: Our results suggest that neurological phenotypes in GPR120 KO mice are probably caused by dysfunction of intestinal GPR120. These observations raise the possibility that intestinal GLP-1 secretion, stimulated by intestinal GPR120, may remotely contributed to suppress PGD2-microglia-provoked neuroinflammation in the hippocampus.


Asunto(s)
Hipocampo/patología , Microglía/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neuroinflamatorias/genética , Prostaglandina D2/genética , Receptores Acoplados a Proteínas G/genética , Supresión Genética/genética , Animales , Conducta Animal , Ácidos Grasos Insaturados/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Liraglutida/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/psicología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/psicología , Prostaglandina D2/biosíntesis
2.
J Neurosci ; 39(24): 4668-4683, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-30988167

RESUMEN

Sex differences in behavior allow animals to effectively mate and reproduce. However, the mechanism by which biological sex regulates behavioral states, which underlie the regulation of sex-shared behaviors, such as locomotion, is largely unknown. In this study, we studied sex differences in the behavioral states of Caenorhabditis elegans and found that males spend less time in a low locomotor activity state than hermaphrodites and that dopamine generates this sex difference. In males, dopamine reduces the low activity state by acting in the same pathway as polycystic kidney disease-related genes that function in male-specific neurons. In hermaphrodites, dopamine increases the low activity state by suppression of octopamine signaling in the sex-shared SIA neurons, which have reduced responsiveness to octopamine in males. Furthermore, dopamine promotes exploration both inside and outside of bacterial lawn (the food source) in males and suppresses it in hermaphrodites. These results demonstrate that sexually dimorphic signaling allows the same neuromodulator to promote adaptive behavior for each sex.SIGNIFICANCE STATEMENT The mechanisms that generate sex differences in sex-shared behaviors, including locomotion, are not well understood. We show that there are sex differences in the regulation of behavioral states in the model animal Caenorhabditis elegans Dopamine promotes the high locomotor activity state in males, which must search for mates to reproduce, and suppresses it in self-fertilizing hermaphrodites through distinct molecular mechanisms. This study demonstrates that sex-specific signaling generates sex differences in the regulation of behavioral states, which in turn modulates the locomotor activity to suit reproduction for each sex.


Asunto(s)
Conducta Animal/efectos de los fármacos , Caenorhabditis elegans/fisiología , Dopamina/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Animales Modificados Genéticamente , Transportador de Aminoácidos Catiônicos 2/metabolismo , Trastornos del Desarrollo Sexual , Conducta Exploratoria/efectos de los fármacos , Femenino , Interneuronas/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Receptores de Amina Biogénica/efectos de los fármacos , Receptores de Amina Biogénica/genética , Serotonina/farmacología , Caracteres Sexuales , Conducta Sexual , Transducción de Señal/efectos de los fármacos
3.
Mol Brain ; 10(1): 2, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28057021

RESUMEN

Sleep-disordered breathing produces cognitive impairments, and is possibly associated with Alzheimer disease (AD). Intermittent hypoxia treatment (IHT), an experimental model for sleep-disordered breathing, results in cognitive impairments in animals via unknown mechanisms. Here, we exposed mice to IHT protocols, and performed biochemical analyses and microarray analyses regarding their hippocampal samples. In particular, we performed gene ontology (GO)-based microarray analysis to elucidate effects of IHT on hippocampal functioning, which were compared with the effects of various previously-reported experimental conditions on that (ref. Gene Expression Omnibus, The National Center for Biotechnology Information). Our microarray analyses revealed that IHT and aging shared alterations in some common GO, which were also observed with kainic acid treatment, Dicer ablation, or moderate glutamate excess. Mapping the altered genes using the Kyoto Encyclopedia of Genes and Genomes PATHWAY database indicated that IHT and aging affected several pathways including "MAPK signaling pathway", "PI3K-Akt signaling pathway", and "glutamatergic synapse". Consistent with the gene analyses, in vivo analyses revealed that IHT increased phosphorylated tau, reflecting an imbalance of kinases and/or phosphatases, and reduced proteins relevant to glutamatergic synapses. In addition, IHT increased phosphorylated p70 S6 kinase, indicating involvement of the mammalian target of rapamycin signaling pathway. Furthermore, IHT mice demonstrated hyperactivity in Y-maze tests, which was also observed in AD models. We obtained important data or something from the massive amount of microarray data, and confirmed the validity by in vivo analyses: the IHT-induced cognitive impairment may be partially explained by the fact that IHT increases phosphorylated tau via biological processes common to aging. Moreover, as aging is a major risk factor for AD, IHT is a novel model for investigating the pathological processes contributing to AD onset.


Asunto(s)
Envejecimiento/patología , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia/metabolismo , Hipoxia/patología , Proteínas tau/metabolismo , Animales , Ontología de Genes , Masculino , Aprendizaje por Laberinto , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Análisis de Componente Principal , Sinapsis/metabolismo
4.
Eur J Pharmacol ; 741: 17-24, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25084219

RESUMEN

Multiple sclerosis is a chronic demyelinating disease of the central nervous system leading to progressive cognitive and motor dysfunction, which is characterized by neuroinflammation, demyelination, astrogliosis, loss of oligodendrocytes, and axonal pathologies. Cyclic phosphatidic acid (cPA) is a naturally occurring phospholipid mediator with a unique cyclic phosphate ring structure at the sn-2 and sn-3 positions of the glycerol backbone. cPA elicits a neurotrophin-like action and protects hippocampal neurons from ischemia-induced delayed neuronal death. In this study, we investigated the effects of cPA on cuprizone-induced demyelination, which is a model of multiple sclerosis. Mice were fed a diet containing 0.2% cuprizone for 5 weeks, which induces severe demyelination, astrocyte and microglial activation, and motor dysfunction. Simultaneous administration of cPA effectively attenuated cuprizone-induced demyelination, glial activation, and motor dysfunction. These data indicate that cPA may be a useful treatment to reduce the extent of demyelination and the severity of motor dysfunction in multiple sclerosis. cPA is a potential lead compound in the development of drugs for the treatment of this devastating disease.


Asunto(s)
Cuprizona/toxicidad , Enfermedades Desmielinizantes/prevención & control , Modelos Animales de Enfermedad , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , Trastornos de la Destreza Motora/prevención & control , Ácidos Fosfatidicos/uso terapéutico , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Compuestos Heterocíclicos con 1 Anillo/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Trastornos de la Destreza Motora/inducido químicamente , Trastornos de la Destreza Motora/patología , Ácidos Fosfatidicos/farmacología , Resultado del Tratamiento
5.
Br J Pharmacol ; 168(7): 1570-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22639973

RESUMEN

BACKGROUND AND PURPOSE The free fatty acid FFA1 receptor and GPR120 are GPCRs whose endogenous ligands are medium- and long-chain FFAs, and they are important in regulating insulin and GLP-1 secretion respectively. Given that the ligands of FFA1 receptor and GPR120 have similar properties, selective pharmacological tools are required to study their functions further. EXPERIMENTAL APPROACH We used a docking simulation approach using homology models for each receptor. Biological activity was assessed by phosphorylation of ERK and elevation of intracellular calcium ([Ca(2+) ]i ) in cells transfected with FFA1 receptor or GPR120. Insulin secretion from murine pancreatic beta cells (MIN6) was also measured. KEY RESULTS Calculated hydrogen bonding energies between a series of synthetic carboxylic acid compounds and the homology models of the FFA1 receptor and GPR120, using docking simulations, correlated well with the effects of the compounds on ERK phosphorylation in transfected cells (R(2) = 0.65 for FFA1 receptor and 0.76 for GPR120). NCG75, the compound with the highest predicted selectivity for FFA1 receptors from this structure-activity relationship analysis, activated ERK and increased [Ca(2+) ]i as potently as the known FFA1 receptor-selective agonist, Compound 1. Site-directed mutagenesis analysis based on the docking simulation showed that different amino acid residues were important for the recognition and activation by FFA1 receptor agonists. Moreover, NCG75 strongly induced ERK and [Ca(2+) ]i responses, and promoted insulin secretion from MIN6 cells, which express endogenous FFA1 receptors. CONCLUSION AND IMPLICATIONS A docking simulation approach using FFA1 receptor and GPR120 homology models could be useful in predicting FFA1 receptor-selective agonists.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Ácidos Láuricos/química , Simulación del Acoplamiento Molecular , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Animales , Calcio/metabolismo , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Enlace de Hidrógeno , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ácidos Láuricos/farmacología , Ligandos , Ratones , Mutagénesis Sitio-Dirigida , Fosforilación , Receptores Acoplados a Proteínas G/genética , Relación Estructura-Actividad
6.
Biochem Biophys Res Commun ; 421(1): 20-6, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22465007

RESUMEN

Mitochondrial pH is a key determinant of mitochondrial energy metabolism. We have developed a new fluorescence-based ratiometric pH biosensor using a chloride-insensitive and hydrogen-sensitive probe for direct, quantitative and bleaching-free measurement in a living cell. Fusing this biosensor with a mitochondrial localization signal (MTpHGV) allowed us to determine mitochondrial pH. This new system was applied to measure mitochondrial pH in pancreatic ß-cells, in which mitochondrial function plays a pivotal role in insulin secretion. Rat INS1 cells and mouse MIN6 cells are transfected with MTpHGV stably to monitor mitochondrial pH. While carbonyl cyanide 3-chlorophenylhydrazone (CCCP) treatment rapidly decreased mitochondrial pH in cultured rat MTpHGV-INS-1 cells, MTpHGV-MIN6 cells showed a rapid increase. These data suggest that MTpHGV probe exist in matrix side in INS-1 cells, but on the outer side of mitochondrial inner membrane in MIN6 cells. Moreover, while MTpHGV-INS-1 cells showed a rapid increase of pH by glucose stimulation, mitochondrial pH decreased quickly by glucose stimulation in all MTpHGV-MIN6 cells examined and recovered smoothly. Perfusion study of glucose load in MTpHGV-MIN6 cells under aminooxyacetate (AOA) or 100µM diazoxide showed that this mitochondrial pH acidification was dependent on nicotinamide adenine dinucleotide (NADH) shuttle, but independent from KATP channel. This new system for measuring mitochondrial pH is sensitive across the range of physiologic conditions and may be a useful tool for evaluating mitochondrial function in living cells.


Asunto(s)
Técnicas Biosensibles , Células Secretoras de Insulina/química , Mitocondrias/química , Ácido Aminooxiacético/farmacología , Animales , Calcio/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Línea Celular , Diazóxido/farmacología , Glucosa/farmacología , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ionóforos de Protónes/farmacología , Ratas
7.
J Diabetes Investig ; 3(5): 449-56, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24843605

RESUMEN

UNLABELLED: Aims/Introduction: Mutations in hepatocyte nuclear factor-4α (HNF4α) lead to various diseases, among which C-terminal deletions of HNF4α are exclusively responsible for maturity onset diabetes of the young 1 (MODY1). MODY is an autosomal dominant disease characterized by a primary defect in insulin response to glucose, suggesting that the C-terminus of HNF4α is important for pancreatic ß-cell function. To clarify the role of the C-terminus of HNF4α, changes in cellular localization and the binding ability to its regulator were examined, specifically in the region containing Q268, which deletion causes MODY1. MATERIALS AND METHODS: Cellular localization of mutant HNF4α were examined in monkey kidney 7 (COS7), Chinese hamster ovary, rat insulinoma and mouse insulinoma cells, and their binding activity to other proteins were examined by fluorescence resonance energy transfer (FRET) in COS7 cells. RESULTS: Although wild-type HNF4α was localized in the nucleoplasm in transfected cultured cells, Q268X-HNF4α was located predominantly in the nucleolus. Deletion analysis of the C-terminus of HNF4α showed that the S337X-HNF4α mutant, and other mutants with shorter amino acid sequences (S337-K194), were mostly localized in the nucleolus. HNF4α mutants with amino acid sequences shorter than the W192X-HNF4α mutant gradually spread to the nucleoplasm in accordance with their lengths. The A250X-HNF4α mutant was capable of causing the accumulation of HNF4α or the small heterodimer partner (SHP), one of the HNF4α regulators, in the nucleolus. However, the R154X-HNF4α mutant did not have binding ability to wild-type HNF4α or SHP, and thus was seen in the nucleus. CONCLUSIONS: The C-terminus sites might play a key role in facilitating the nucleolar and subnucleolar localization of HNF4α. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2012.00210.x, 2012).

8.
Mol Pharmacol ; 78(5): 804-10, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20685848

RESUMEN

GPR120 is a G protein-coupled receptor expressed preferentially in the intestinal tract and adipose tissue, that has been implicated in mediating free fatty acid-stimulated glucagon-like peptide-1 (GLP-1) secretion. To develop GPR120-specific agonists, a series of compounds (denoted as NCG compounds) derived from a peroxisome proliferator-activated receptor γ agonist were synthesized, and their structure-activity relationships as GPR120 agonists were explored. To examine the agonistic activities of these newly synthesized NCG compounds, and of compounds already shown to have GPR120 agonistic activity (grifolic acid and MEDICA16), we conducted docking simulation in a GPR120 homology model that was developed on the basis of a photoactivated model derived from the crystal structure of bovine rhodopsin. We calculated the hydrogen bonding energies between the compounds and the GPR120 model. These energies correlated well with the GPR120 agonistic activity of the compounds (R(2) = 0.73). NCG21, the NCG compound with the lowest calculated hydrogen bonding energy, showed the most potent extracellular signal-regulated kinase (ERK) activation in a cloned GPR120 system. Furthermore, NCG21 potently activated ERK, intracellular calcium responses and GLP-1 secretion in murine enteroendocrine STC-1 cells that express GPR120 endogenously. Moreover, administration of NCG21 into the mouse colon caused an increase in plasma GLP-1 levels. Taken together, our present study showed that a docking simulation using a GPR120 homology model might be useful to predict the agonistic activity of compounds.


Asunto(s)
Aminopiridinas/química , Modelos Moleculares , Fenilbutiratos/química , Receptores Acoplados a Proteínas G/agonistas , Secuencia de Aminoácidos , Aminopiridinas/farmacología , Animales , Sitios de Unión , Calcio/metabolismo , Bovinos , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Enlace de Hidrógeno , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fenilbutiratos/farmacología , Fosforilación , Unión Proteica , Receptores Acoplados a Proteínas G/química , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
9.
Mol Pharmacol ; 75(1): 85-91, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18927207

RESUMEN

GPR40 is a G protein-coupled receptor (GPCR) whose endogenous ligands have recently been identified as medium- and long-chain free fatty acids (FFAs), and it is thought to play an important role in insulin release. Despite recent research efforts, much still remains unclear in our understanding of its pharmacology, mainly because the receptor-ligand interaction has not been analyzed directly. To study the pharmacology of GPR40 in a more direct fashion, we developed a flow cytometry-based binding assay. FLAG-tagged GPR40 protein was expressed in Sf9 cells, solubilized, immobilized on immunomagnetic beads, and labeled with the fluorescent probe C1-BODIPY-C12. Flow cytometry analysis showed that C1-BODIPY-C12 specifically labels a single class of binding site in a saturable and reversible manner with an apparent dissociation constant of approximately 3 microM. The FFAs that activate GPR40 competed with C1-BODIPY-C12 binding; thus, medium- to long-chain FFAs could compete, whereas short-chain FFAs and methyl linoleate had no inhibitory effect. Furthermore, ligands that are known to activate GPR40 competed for binding in a concentration-dependent manner. All the ligands that inhibited the binding promoted phosphorylation of extracellular signal-regulated kinase (ERK)-1/2 in human embryonic kidney (HEK) 293 cells that expressed GPR40 and [Ca(2+)](i) responses in mouse insulinoma (MIN6) cells that natively express GPR40; however, pioglitazone, a thiazolidinedione that failed to compete for the binding, did not activate ERK or [Ca(2+)](i) response. This study showed that a flow cytometry-based binding assay can successfully identify direct interactions between GPR40 and its ligands. This approach would be of value in studying the pharmacology of GPCRs.


Asunto(s)
Bioensayo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Baculoviridae/genética , Sitios de Unión , Unión Competitiva , Compuestos de Boro/química , Compuestos de Boro/metabolismo , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Citometría de Flujo/métodos , Colorantes Fluorescentes/metabolismo , Humanos , Concentración 50 Inhibidora , Insulinoma/metabolismo , Insulinoma/patología , Riñón/citología , Ligandos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación , Unión Proteica , Spodoptera/citología , Spodoptera/metabolismo
10.
Biol Reprod ; 78(6): 1081-90, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18322275

RESUMEN

Mouse phospholipase C, zeta 1 (PLCZ1), a strong candidate of egg-activating sperm factor, induces Ca(2+) oscillations and accumulates into formed pronucleus (PN) when expressed by cRNA injection. These activities were compared among mouse and human PLCZ1, newly cloned rat Plcz1, and medaka fish plcz1. The PLCZ1 proteins of the four species have an approximately homologous sequence of nuclear localization signal. However, the nuclear translocation ability was defective in rat, human, and medaka PLCZ1 expressed in mouse eggs. Rat PLCZ1 could not enter rat PN, whereas mouse PLCZ1 could. Mouse and human PLCZ1 translocated into the nucleus of COS-7 cells transfected with cDNA. There was little medaka PLCZ1 accumulated in the nucleus, and rat PLCZ1 was never located in the nucleus. All PLCZ1 proteins including fish could induce Ca(2+) oscillations in mouse eggs, but the activity was variable in the order of human >> mouse > medaka >> rat, estimated from minimal RNA concentration to induce Ca(2+) spikes. Ca(2+) oscillations by human PLCZ1 continued far beyond the time of PN formation (T(PN)), whereas those by mouse PLCZ1 ceased slightly before T(PN). High-frequency Ca(2+) spikes by overexpressed rat PLCZ1 stopped far before T(PN), possibly by feedback inhibition. Ca(2+) oscillations by fertilization of rat eggs stopped at T(PN), despite defective nuclear translocation of rat PLCZ1. Thus, PLCZ1 sequestration into PN participates in termination of Ca(2+) oscillations at the interphase of mouse embryos but does not always operate in other mammals, notably in rat embryos.


Asunto(s)
Señalización del Calcio/fisiología , Fosfoinositido Fosfolipasa C/metabolismo , Espermatozoides/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Cartilla de ADN/genética , Femenino , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Oryzias , Óvulo/metabolismo , Fosfoinositido Fosfolipasa C/genética , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Interacciones Espermatozoide-Óvulo/fisiología , Transfección
11.
J Biol Chem ; 281(38): 27794-805, 2006 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-16854985

RESUMEN

Sperm-specific phospholipase C-zeta (PLCzeta) causes intracellular Ca(2+) oscillations and thereby egg activation and is accumulated into the formed pronucleus (PN) when expressed in mouse eggs by injection of cRNA encoding PLCzeta, which consists of four EF-hand domains (EF1-EF4) in the N terminus, X and Y catalytic domains, and C-terminal C2 domain. Those activities were analyzed by expressing PLCzeta mutants tagged with fluorescent protein Venus by injection of cRNA into unfertilized eggs or 1-cell embryos after fertilization. Nuclear localization signal (NLS) existed at 374-381 in the X/Y linker region. Nuclear translocation was lost by replacement of Arg(376), Lys(377), Arg(378), Lys(379), or Lys(381) with glutamate, whereas Ca(2+) oscillations were conserved. Nuclear targeting was also absent for point mutation of Lys(299) and/or Lys(301) in the C terminus of X domain, or Trp(13), Phe(14), or Val(18) in the N terminus of EF1. Ca(2+) oscillation-inducing activity was lost by the former mutation and was remarkably inhibited by the latter. A short sequence 374-383 fused with Venus showed active translocation into the nucleus of COS-7 cells, but 296-309 or 1-19 did not. Despite the presence of these special regions, both activities were deprived by deletion of not only EF1 but also EF2-4 or C2 domain. Thus, PLCzeta is driven into the nucleus primarily by the aid of NLS and putative regulatory sites, but coordinated three-dimensional structure, possibly formed by a folding in the X/Y linker and close EF/C2 contact as in PLCdelta1, seems to be required not only for enzymatic activity but also for nuclear translocation ability.


Asunto(s)
Señalización del Calcio , Núcleo Celular/metabolismo , Fosfolipasas de Tipo C/química , Transporte Activo de Núcleo Celular , Animales , Dominio Catalítico , Células Cultivadas , Femenino , Ratones , Modelos Moleculares , Señales de Localización Nuclear , Óvulo/fisiología , Fosfoinositido Fosfolipasa C , Transporte de Proteínas , Fosfolipasas de Tipo C/fisiología
12.
Nat Med ; 11(1): 90-4, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15619630

RESUMEN

Diabetes, a disease in which the body does not produce or use insulin properly, is a serious global health problem. Gut polypeptides secreted in response to food intake, such as glucagon-like peptide-1 (GLP-1), are potent incretin hormones that enhance the glucose-dependent secretion of insulin from pancreatic beta cells. Free fatty acids (FFAs) provide an important energy source and also act as signaling molecules in various cellular processes, including the secretion of gut incretin peptides. Here we show that a G-protein-coupled receptor, GPR120, which is abundantly expressed in intestine, functions as a receptor for unsaturated long-chain FFAs. Furthermore, we show that the stimulation of GPR120 by FFAs promotes the secretion of GLP-1 in vitro and in vivo, and increases circulating insulin. Because GLP-1 is the most potent insulinotropic incretin, our results indicate that GPR120-mediated GLP-1 secretion induced by dietary FFAs is important in the treatment of diabetes.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Glucagón/metabolismo , Mucosa Intestinal/metabolismo , Fragmentos de Péptidos/metabolismo , Precursores de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Péptido 1 Similar al Glucagón , Humanos , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética
13.
Dev Biol ; 268(2): 245-57, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15063165

RESUMEN

Sperm-specific phospholipase C zeta (PLC zeta) is known to induce intracellular Ca(2+) oscillations and egg activation when expressed in mouse eggs by injection of RNA encoding PLC zeta. We investigated the expression level and spatial distribution of PLC zeta in the egg in real time and in relation to the initiation and termination of Ca(2+) oscillations by monitoring fluorescence of a yellow fluorescent protein 'Venus' fused with PLC zeta. Ca(2+) oscillations similar to those at fertilization were induced at 40-50 min after RNA injection, when expressed PLC zeta reached 10-40 x 10(-15) g in the egg. PLC zeta-Venus increased up to 3 h and attained a steady level at 4-5 h. Interestingly, PLC zeta-Venus is accumulated to the pronucleus (PN) formed at 5-6 h and continuously increased there. Ca(2+) oscillations stopped in most eggs before initiation of the accumulation. A variant of PLC zeta that lacks three EF hand domains was much less effective in induction of Ca(2+) oscillations and little accumulated in the pronucleus, indicating a critical role of those domains. The ability of the accumulation to the pronucleus qualifies PLC zeta for a strong candidate of the Ca(2+) oscillation-inducing sperm factor, which is introduced into the ooplasm upon sperm-egg fusion and concentrated to the pronucleus after inducing egg activation.


Asunto(s)
Calcio/metabolismo , Núcleo Celular/metabolismo , Óvulo/enzimología , Fosfolipasas de Tipo C/genética , Animales , Citoplasma/metabolismo , Genes Reporteros , Isoenzimas/metabolismo , Ratones , Óvulo/metabolismo , Fosfoinositido Fosfolipasa C , Fosfolipasa C delta , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Fosfolipasas de Tipo C/biosíntesis , Fosfolipasas de Tipo C/metabolismo
14.
Biochem Biophys Res Commun ; 292(1): 8-12, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11890664

RESUMEN

Mutations in small heterodimer partner (SHP) and hepatocyte nuclear factor 4alpha (HNF4alpha) are associated with mild obesity and diabetes mellitus, respectively. Both receptors work together to determine the normal pancreatic beta-cell function. We examined their subcellular localization and interaction in living cells by tagging them with yellow and cyan variants of green fluorescent protein (GFP) variants. Expressed SHP resided only in the cytoplasm in COS-7 cells which lacks HNF4alpha, but predominantly in the nucleus in insulinoma cells (MIN6). HNF4alpha was localized exclusively in the nuclei of both cells, coexpressed with HNF4alpha in COS-7 cells, redistributed in the nucleus, depending on the amount of HNF4alpha. We found fluorescence resonance energy transfer between GFP-tagged SHP and HNF4alpha, indicating a specific close association between them in the nucleus. The results strongly suggest that SHP exists primarily in the cytoplasm and is translocated into the nucleus on interacting with its nuclear receptor partner HNF4alpha.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN , Fosfoproteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Proteínas Fluorescentes Verdes , Factor Nuclear 4 del Hepatocito , Indicadores y Reactivos/análisis , Insulinoma , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Fosfoproteínas/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Recombinantes de Fusión/análisis , Factores de Transcripción/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...