Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Hum Genet ; 140(2): 277-287, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32617796

RESUMEN

CRISPR-Cas9 are widely used for gene targeting in mice and rats. The non-homologous end-joining (NHEJ) repair pathway, which is dominant in zygotes, efficiently induces insertion or deletion (indel) mutations as gene knockouts at targeted sites, whereas gene knock-ins (KIs) via homology-directed repair (HDR) are difficult to generate. In this study, we used a double-stranded DNA (dsDNA) donor template with Cas9 and two single guide RNAs, one designed to cut the targeted genome sequences and the other to cut both the flanked genomic region and one homology arm of the dsDNA plasmid, which resulted in 20-33% KI efficiency among G0 pups. G0 KI mice carried NHEJ-dependent indel mutations at one targeting site that was designed at the intron region, and HDR-dependent precise KIs of the various donor cassettes spanning from 1 to 5 kbp, such as EGFP, mCherry, Cre, and genes of interest, at the other exon site. These findings indicate that this combinatorial method of NHEJ and HDR mediated by the CRISPR-Cas9 system facilitates the efficient and precise KIs of plasmid DNA cassettes in mice and rats.


Asunto(s)
Sistemas CRISPR-Cas/genética , Reparación del ADN por Unión de Extremidades/genética , Técnicas de Sustitución del Gen/métodos , Plásmidos/genética , Reparación del ADN por Recombinación/genética , Animales , ADN/genética , Exones/genética , Femenino , Edición Génica/métodos , Genoma/genética , Intrones/genética , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Ratas , Ratas Long-Evans , Ratas Wistar
2.
J Biol Chem ; 288(7): 4513-21, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23269670

RESUMEN

Although amino acids are dietary nutrients that evoke the secretion of glucagon-like peptide 1 (GLP-1) from intestinal L cells, the precise molecular mechanism(s) by which amino acids regulate GLP-1 secretion from intestinal L cells remains unknown. Here, we show that the G protein-coupled receptor (GPCR), family C group 6 subtype A (GPRC6A), is involved in amino acid-induced GLP-1 secretion from the intestinal L cell line GLUTag. Application of l-ornithine caused an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) in GLUTag cells. Application of a GPRC6A receptor antagonist, a phospholipase C inhibitor, or an IP(3) receptor antagonist significantly suppressed the l-ornithine-induced [Ca(2+)](i) increase. We found that the increase in [Ca(2+)](i) stimulated by l-ornithine correlated with GLP-1 secretion and that l-ornithine stimulation increased exocytosis in a dose-dependent manner. Furthermore, depletion of endogenous GPRC6A by a specific small interfering RNA (siRNA) inhibited the l-ornithine-induced [Ca(2+)](i) increase and GLP-1 secretion. Taken together, these findings suggest that the GPRC6A receptor functions as an amino acid sensor in GLUTag cells that promotes GLP-1 secretion.


Asunto(s)
Células Enteroendocrinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Aminoácidos/metabolismo , Animales , Calcio/metabolismo , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica/métodos , Ligandos , Ratones , Ratones Transgénicos , Modelos Biológicos , Ornitina/farmacología , ARN Interferente Pequeño/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Factores de Tiempo , Transfección
3.
Biochem J ; 450(2): 365-73, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23282092

RESUMEN

Intracellular cAMP and Ca(2+) are important second messengers that regulate insulin secretion in pancreatic ß-cells; however, the molecular mechanism underlying their mutual interaction for exocytosis is not fully understood. In the present study, we investigated the interplay between intracellular cAMP and Ca(2+) concentrations ([cAMP](i) and [Ca(2+)](i) respectively) in the pancreatic ß-cell line MIN6 using total internal reflection fluorescence microscopy. For measuring [cAMP](i), we developed a genetically encoded yellow fluorescent biosensor for cAMP [Flamindo (fluorescent cAMP indicator)], which changes fluorescence intensity with cAMP binding. Application of high-KCl or glucose to MIN6 cells induced the elevation of [cAMP](i) and exocytosis. Furthermore, application of an L-type Ca(2+) channel agonist or ionomycin to induce extracellular Ca(2+) influx evoked the elevation of [cAMP](i), whereas application of carbachol or thapsigargin, which mobilize Ca(2+) from internal stores, did not evoke the elevation of [cAMP](i). We performed RT (reverse transcription)-PCR analysis and found that Ca(2+)-sensitive Adcy1 (adenylate cyclase 1) was expressed in MIN6 cells. Knockdown of endogenous ADCY1 by small interference RNA significantly suppressed glucose-induced exocytosis and the elevation of both [cAMP](i) and [Ca(2+)](i). Taken together, the findings of the present study demonstrate that ADCY1 plays an important role in the control of pancreatic ß-cell cAMP homoeostasis and insulin secretion.


Asunto(s)
Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Insulina/metabolismo , Adenilil Ciclasas/genética , Animales , Células COS , Señalización del Calcio , Células Cultivadas , Chlorocebus aethiops , AMP Cíclico/metabolismo , Glucosa/metabolismo , Secreción de Insulina , Ratones , Transfección
4.
Cell Metab ; 36(5): 1044-1058.e10, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38452767

RESUMEN

Obesity is often associated with aging. However, the mechanism of age-related obesity is unknown. The melanocortin-4 receptor (MC4R) mediates leptin-melanocortin anti-obesity signaling in the hypothalamus. Here, we discovered that MC4R-bearing primary cilia of hypothalamic neurons progressively shorten with age in rats, correlating with age-dependent metabolic decline and increased adiposity. This "age-related ciliopathy" is promoted by overnutrition-induced upregulation of leptin-melanocortin signaling and inhibited or reversed by dietary restriction or the knockdown of ciliogenesis-associated kinase 1 (CILK1). Forced shortening of MC4R-bearing cilia in hypothalamic neurons by genetic approaches impaired neuronal sensitivity to melanocortin and resulted in decreased brown fat thermogenesis and energy expenditure and increased appetite, finally developing obesity and leptin resistance. Therefore, despite its acute anti-obesity effect, chronic leptin-melanocortin signaling increases susceptibility to obesity by promoting the age-related shortening of MC4R-bearing cilia. This study provides a crucial mechanism for age-related obesity, which increases the risk of metabolic syndrome.


Asunto(s)
Cilios , Leptina , Neuronas , Obesidad , Receptor de Melanocortina Tipo 4 , Animales , Receptor de Melanocortina Tipo 4/metabolismo , Receptor de Melanocortina Tipo 4/genética , Cilios/metabolismo , Cilios/patología , Obesidad/metabolismo , Obesidad/patología , Neuronas/metabolismo , Neuronas/patología , Leptina/metabolismo , Ratas , Masculino , Transducción de Señal , Hipotálamo/metabolismo , Envejecimiento/metabolismo , Envejecimiento/patología , Ratas Sprague-Dawley , Ratones , Metabolismo Energético , Tejido Adiposo Pardo/metabolismo , Termogénesis
5.
Biochem Biophys Res Commun ; 438(1): 145-51, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23876310

RESUMEN

Recent studies have suggested that astrocytes release gliotransmitters (i.e., ATP, L-glutamate, D-serine, and peptide hormones) and participate actively in synaptic functioning. Although ATP release from astrocytes modulates the activity of neurons, the mechanisms regulating the ATP release from astrocytes and the source of ATP in astrocytes are not well understood. Recently a vesicular nucleotide transporter (VNUT)/solute carrier family 17, member 9 (SLC17A9) has been identified as a mediator of the active accumulation of ATP into vesicles. Here we show by immunocytochemical analysis under confocal microscope and live cell imaging under total internal reflection fluorescence microscope that lysosome-associated VNUT is responsible for ATP release in astrocytes. VNUT was expressed in both primary cultured cortical astrocytes and glioma cell line C6 cells, and mainly localized on lysosome in the cells. We found that VNUT-associated secretory lysosomes do not fully collapse into the plasma membrane after lysosomal exocytosis. We also found that inhibition of VNUT function by Evans Blue decreased ATP uptake into secretory lysosomes. Depletion and inhibition of endogenous VNUT by small interference RNA and Evans Blue, respectively decreased the amount of ATP release from the cells, whereas overexpression of VNUT increased it. Taken together, these findings indicate that the participation of VNUT in ATP storage in secretory lysosomes during lysosomal exocytosis of ATP from astrocytes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Astrocitos/metabolismo , Lisosomas/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Células Cultivadas , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
6.
Genes Cells ; 16(5): 608-16, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21470345

RESUMEN

Amino acids such as L-glutamate and L-arginine are potent stimuli for insulin secretion from pancreatic ß-cells. However, the precise molecular mechanisms of amino acid-induced insulin secretion have only partly understood. In this study, we show here that mouse pancreatic ß-cell line MIN6 cells expressed amino acid taste receptor (heterodimer of type 1 taste G protein-coupled receptor member 1 and member 3; Tas1R1 and Tas1R3, respectively). We found that administration of L-glutamate or L-arginine to MIN6 cells caused the increase in free intracellular concentrations of both inositol-1,4,5-triphosphate (IP(3)) and Ca(2+) , and umami substance inocinate enhanced the effects of l-glutamate. Effects of amino acids on intracellular IP(3) and Ca(2+) concentration were diminished by application of lactisole, a Tas1R3 receptor antagonist. Furthermore, we investigated the effect of amino acids on the insulin release from MIN6 cells by both ELISA and total internal reflection fluorescence microscopy. Application of L-glutamate or L-arginine significantly increased the release of insulin, whereas inhibited by the application of lactisole. Based on these findings, we propose that heterodimer of Tas1R1 and Tas1R3 is the fundamental receptor for the sensing amino acids and regulates the amino acid-induced insulin secretion in pancreatic ß-cells.


Asunto(s)
Aminoácidos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética , Aminoácidos/farmacología , Animales , Arginina/metabolismo , Arginina/farmacología , Derivados del Benceno/farmacología , Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Immunoblotting , Inositol 1,4,5-Trifosfato/metabolismo , Secreción de Insulina , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Multimerización de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Gusto
7.
Biochem J ; 429(3): 497-504, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20528772

RESUMEN

Since the fusion pore of the secretory vesicle is resealed before complete dilation during 'kiss-and-run' exocytosis, their cargoes are not completely released. Although the transient fusion pore is kept open for several seconds, the precise mechanisms that control fusion pore maintenance, and their physiological significance, are not well understood. Using dual-colour TIRF (total internal reflection fluorescence) microscopy in neuroendocrine PC12 cells, we show that myosin II regulates the fusion pore dynamics during kiss-and-run exocytosis. The release kinetics of mRFP (monomeric red fluorescent protein)-tagged tPA (tissue plasminogen activator) and Venus-tagged BDNF (brain-derived neurotrophic factor), which show slower release kinetics than NPY (neuropeptide Y)-mRFP and insulin-mRFP, were prolonged by the overexpression of a wild-type form of the RLC (myosin II regulatory light chain). In contrast, overexpression of a dominant-negative form of RLC shortened the release kinetics. Using spH (synapto-pHluorin), a green fluorescent protein-based pH sensor inside the vesicles, we confirmed that the modulation of the release kinetics by myosin II is due to changes in the duration of fusion pore opening. In addition, we revealed that the amount of hormone released into the extracellular space upon stimulation was increased by overexpression of wild-type RLC. We propose that the duration of fusion pore opening is regulated by myosin II to control the amount of hormone released from a single vesicle.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Exocitosis , Fusión de Membrana , Miosina Tipo II/fisiología , Activador de Tejido Plasminógeno/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Inmunohistoquímica , Cinética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Células PC12 , Ratas , Proteína Fluorescente Roja
8.
J Neurochem ; 114(1): 171-81, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20403080

RESUMEN

The double C2 (Doc2) family is characterized by an N-terminal Munc13-1-interacting domain and C-terminal tandem C2 domains, and it comprises three isoforms, Doc2alpha, Doc2beta, and Doc2gamma, in humans and mice. Doc2alpha, the best-characterized, brain-specific isoform, exhibits Ca(2+)-dependent phospholipid-binding activity through its C2A domain, and the Ca(2+)-binding activity is thought to be important for the regulation of Ca(2+)-dependent exocytosis. In contrast to the C2A domain, however, nothing is known about the physiological functions of the C2B domain in regulated exocytosis. In this study, we demonstrated by a mutation analysis that the polybasic sequence in the C2B domain of Doc2alpha (306 KKSKHKTCVKKK 317) is required for binding of syntaxin-1a/synaptosome-associated protein of 25 kDa (SNAP-25) heterodimer. We also investigated the effect of Lys-to-Gln (named KQ) mutations in the polybasic sequence of the C2B domain on vesicle dynamics by total internal reflection fluorescence microscopy in PC12 cells. A Doc2alpha(KQ) mutant, which lacks binding activity toward syntaxin-1a/SNAP-25 heterodimer, significantly decreased the number of plasma membrane-docked vesicles before stimulation and strongly inhibited high-KCl-induced exocytosis from the plasma membrane-docked vesicles. These results indicate that the polybasic sequence in the C2B domain functions as a binding site for syntaxin-1a/SNAP-25 heterodimer and controls the number of 'readily releasable' vesicles in neuroendocrine cells.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Exocitosis , Proteínas del Tejido Nervioso/fisiología , Vesículas Secretoras/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/genética , Membrana Celular/metabolismo , Citosol/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Proteínas del Tejido Nervioso/genética , Células PC12 , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Proteína 25 Asociada a Sinaptosomas/genética , Sintaxina 1/genética
9.
J Endocrinol ; 226(1): 25-34, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26099355

RESUMEN

Ghrelin is synthesized in X/A-like cells of the gastric mucosa, which plays an important role in the regulation of energy homeostasis. Although ghrelin secretion is known to be induced by neurotransmitters or hormones or by nutrient sensing in the ghrelin-secreting cells themselves, the mechanism of ghrelin secretion is not clearly understood. In the present study, we found that changing the extracellular glucose concentration from elevated (25  mM) to optimal (10 mM) caused an increase in the intracellular Ca2+ concentration ([Ca2+]i) in ghrelin-secreting mouse ghrelinoma 3-1 (MGN3-1) cells (n=32, P<0.01), whereas changing the glucose concentration from elevated to lowered (5 or 1 mM) had little effect on [Ca2+]i increase. Overexpression of a closed form of an ATP-sensitive K+ (KATP) channel mutant suppressed the 10  mM glucose-induced [Ca2+]i increase (n=8, P<0.01) and exocytotic events (n=6, P<0.01). We also found that a low concentration of a KATP channel opener, diazoxide, with 25  mM glucose induced [Ca2+]i increase (n=23, P<0.01) and ghrelin secretion (n≥3, P<0.05). In contrast, the application of a low concentration of a KATP channel blocker, tolbutamide, significantly induced [Ca2+]i increase (n=15, P<0.01) and ghrelin secretion (n≥3, P<0.05) under 5 mM glucose. Furthermore, the application of voltage-dependent Ca2+ channel inhibitors suppressed the 10 mM glucose-induced [Ca2+]i increase (n≥26, P<0.01) and ghrelin secretion (n≥5, P<0.05). These findings suggest that KATP and voltage-dependent Ca2+ channels are involved in glucose-dependent ghrelin secretion in MGN3-1 cells.


Asunto(s)
Ghrelina/metabolismo , Glucosa/metabolismo , Canales KATP/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Señalización del Calcio , Línea Celular , Diazóxido/farmacología , Exocitosis , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Canales KATP/efectos de los fármacos , Canales KATP/genética , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Tolbutamida/farmacología
10.
Neurotoxicology ; 37: 197-206, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23694735

RESUMEN

There has been broad concern that arsenic in the environment exerts neurotoxicity. To determine the mechanism by which arsenic disrupts neuronal development, primary cultured neurons obtained from the cerebral cortex of mouse embryos were exposed to sodium arsenite (NaAsO2) at concentrations between 0 and 2 µM from days 2 to 4 in vitro and cell survival, neurite outgrowth and expression of glutamate AMPA receptor subunits were assessed at day 4 in vitro. Cell survival was significantly decreased by exposure to 2 µM NaAsO2, whereas 0.5 µM NaAsO2 increased cell survival instead. The assessment of neurite outgrowth showed that total neurite length was significantly suppressed by 1 µM and 2 µM NaAsO2, indicating that the lower concentration of NaAsO2 impairs neuritogenesis before inducing cell death. Immunoblot analysis of AMPA receptor subunit expression showed that the protein level of GluA1, a specific subunit of the AMPA receptor, was significantly decreased by 1 µM and 2 µM NaAsO2. When immunocytochemistry was used to confirm this effect by staining for GluA1 expression in neuropeptide Y neurons, most of which contain GluA1, GluA1 expression in neuropeptide Y neurons was found to be significantly suppressed by 1 µM and 2 µM NaAsO2 but to be increased at the concentration of 0.5 µM. Finally, to determine whether neurons could be rescued from the NaAsO2-induced impairment of neuritogenesis by compensatory overexpression of GluA1, we used primary cultures of neurons transfected with a plasmid vector to overexpress either GluA1 or GluA2, and the results showed that GluA1/2 overexpression protected against the deleterious effects of NaAsO2 on neurite outgrowth. These results suggest that the NaAsO2 concentration inducing neurite suppression is lower than the concentration that induces cell death and is the same as the concentration that suppresses GluA1 expression. Consequently, the suppression of GluA1 expression by NaAsO2 seems at least partly responsible for neurite suppression induced by NaAsO2.


Asunto(s)
Arsenitos/toxicidad , Corteza Cerebral/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Neuritas/efectos de los fármacos , Receptores AMPA/efectos de los fármacos , Compuestos de Sodio/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Femenino , Ácido Glutámico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuritas/metabolismo , Neuritas/patología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Receptores AMPA/genética , Receptores AMPA/metabolismo , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA