Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Nutrients ; 16(14)2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39064713

RESUMEN

(1) Background: Proglucagon-derived peptides (PDGPs) including glucagon (Gcg), GLP-1, and GLP-2 regulate lipid metabolism in the liver, adipocytes, and intestine. However, the mechanism by which PGDPs participate in alterations in lipid metabolism induced by high-fat diet (HFD) feeding has not been elucidated. (2) Methods: Mice deficient in PGDP (GCGKO) and control mice were fed HFD for 7 days and analyzed, and differences in lipid metabolism in the liver, adipose tissue, and duodenum were investigated. (3) Results: GCGKO mice under HFD showed lower expression levels of the genes involved in free fatty acid (FFA) oxidation such as Hsl, Atgl, Cpt1a, Acox1 (p < 0.05), and Pparα (p = 0.05) mRNA in the liver than in control mice, and both FFA and triglycerides content in liver and adipose tissue weight were lower in the GCGKO mice. On the other hand, phosphorylation of hormone-sensitive lipase (HSL) in white adipose tissue did not differ between the two groups. GCGKO mice under HFD exhibited lower expression levels of Pparα and Cd36 mRNA in the duodenum as well as increased fecal cholesterol contents compared to HFD-controls. (4) Conclusions: GCGKO mice fed HFD exhibit a lesser increase in hepatic FFA and triglyceride contents and adipose tissue weight, despite reduced ß-oxidation in the liver, than in control mice. Thus, the absence of PGDP prevents dietary-induced fatty liver development due to decreased lipid uptake in the intestinal tract.


Asunto(s)
Antígenos CD36 , Dieta Alta en Grasa , Absorción Intestinal , Metabolismo de los Lípidos , Hígado , Ratones Noqueados , PPAR alfa , Proglucagón , Animales , Masculino , Dieta Alta en Grasa/efectos adversos , PPAR alfa/metabolismo , PPAR alfa/genética , Hígado/metabolismo , Proglucagón/metabolismo , Proglucagón/genética , Antígenos CD36/metabolismo , Antígenos CD36/genética , Ratones , Esterol Esterasa/metabolismo , Esterol Esterasa/genética , Triglicéridos/metabolismo , Ratones Endogámicos C57BL , Ácidos Grasos no Esterificados/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Duodeno/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Tejido Adiposo/metabolismo , Grasas de la Dieta , Péptido 2 Similar al Glucagón/metabolismo , Aciltransferasas , Lipasa
2.
J Physiol Sci ; 73(1): 19, 2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37704979

RESUMEN

Neural stem cells (NSCs) are maintained in the adult mammalian brain throughout the animal's lifespan. NSCs in the subependymal zone infrequently divide and generate transit amplifying cells, which are destined to become olfactory bulb neurons. When transit amplifying cells are depleted, they are replenished by the quiescent NSC pool. However, the cellular basis for this recovery process remains largely unknown. In this study, we traced NSCs and their progeny after transit amplifying cells were eliminated by intraventricular infusion of cytosine ß-D-arabinofuranoside. We found that although the number of neurosphere-forming NSCs decreased shortly after the treatment, they were restored to normal levels 3 weeks after the cessation of treatment. More importantly, the depletion of transit amplifying cells did not induce a significant expansion of the NSC pool by symmetric divisions. Our data suggest that the size of the NSC pool is hardly affected by brain damage due to antimitotic drug treatment.


Asunto(s)
Encéfalo , Células-Madre Neurales , Animales , Neuronas , Infusiones Intraventriculares , Longevidad , Mamíferos
3.
J Diabetes Investig ; 14(9): 1045-1055, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37300240

RESUMEN

AIMS/INTRODUCTION: Glucagon is secreted from pancreatic α-cells and plays an important role in amino acid metabolism in liver. Various animal models deficient in glucagon action show hyper-amino acidemia and α-cell hyperplasia, indicating that glucagon contributes to feedback regulation between the liver and the α-cells. In addition, both insulin and various amino acids, including branched-chain amino acids and alanine, participate in protein synthesis in skeletal muscle. However, the effect of hyperaminoacidemia on skeletal muscle has not been investigated. In the present study, we examined the effect of blockade of glucagon action on skeletal muscle using mice deficient in proglucagon-derived peptides (GCGKO mice). MATERIALS AND METHODS: Muscles isolated from GCGKO and control mice were analyzed for their morphology, gene expression and metabolites. RESULTS: GCGKO mice showed muscle fiber hypertrophy, and a decreased ratio of type IIA and an increased ratio of type IIB fibers in the tibialis anterior. The expression levels of myosin heavy chain (Myh) 7, 2, 1 and myoglobin messenger ribonucleic acid were significantly lower in GCGKO mice than those in control mice in the tibialis anterior. GCGKO mice showed a significantly higher concentration of arginine, asparagine, serine and threonine in the quadriceps femoris muscles, and also alanine, aspartic acid, cysteine, glutamine, glycine and lysine, as well as four amino acids in gastrocnemius muscles. CONCLUSIONS: These results show that hyperaminoacidemia induced by blockade of glucagon action in mice increases skeletal muscle weight and stimulates slow-to-fast transition in type II fibers of skeletal muscle, mimicking the phenotype of a high-protein diet.


Asunto(s)
Glucagón , Músculo Esquelético , Proglucagón , Animales , Ratones , Aminoácidos , Glucagón/metabolismo , Músculo Esquelético/metabolismo , Proglucagón/genética , Proglucagón/metabolismo
4.
J Diabetes Investig ; 14(5): 648-658, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36729958

RESUMEN

AIMS/INTRODUCTION: Glucagon, a peptide hormone produced from proglucagon, is involved in the pathophysiology of diabetes. Plasma glucagon levels are currently measured by sandwich enzyme-linked immunosorbent assay (ELISA), but the currently used sandwich ELISA cross-reacts with proglucagon-derived peptides, thereby providing incorrect results in subjects with elevated plasma proglucagon-derived peptide levels. We aimed to develop a more broadly reliable ELISA for measuring plasma glucagon levels. MATERIALS AND METHODS: A new sandwich ELISA was developed using newly generated monoclonal antibodies against glucagon. After its validation, plasma glucagon levels were measured with the new ELISA and the currently used ELISA in subjects who underwent laparoscopic sleeve gastrectomy (LSG) and in outpatients with suspected glucose intolerance. The ELISA results were compared with those from liquid chromatography-high resolution mass (LC-HRMS) analysis, which we previously established as the most accurate measuring system. RESULTS: The new ELISA has high specificity (<1% cross-reactivities) and high sensitivity (a lower range of 0.31 pmol/L). Plasma glucagon values in the subjects who underwent laparoscopic sleeve gastrectomy and some outpatients with suspected glucose intolerance differed between the new ELISA and the currently used ELISA. These subjects also showed markedly high plasma glicentin levels. Despite the elevated plasma glicentin levels, the new ELISA showed better positive correlation with LC-HRMS than did the currently used ELISA. CONCLUSIONS: The new ELISA enables more accurate measurement of plasma glucagon than the currently used ELISA, even in subjects with elevated proglucagon-derived peptide levels. It should be clinically useful in elucidating the pathophysiology of individual diabetic patients.


Asunto(s)
Diabetes Mellitus , Intolerancia a la Glucosa , Hormonas Peptídicas , Humanos , Glucagón , Proglucagón , Glicentina , Intolerancia a la Glucosa/diagnóstico , Glucosa , Ensayo de Inmunoadsorción Enzimática/métodos
5.
J Atheroscler Thromb ; 30(1): 74-86, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35314564

RESUMEN

AIMS: We previously reported that glucagon-like peptide-1 receptor agonists (GLP-1RAs) reduced serum low-density lipoprotein cholesterol (LDL-C) levels in patients with type 2 diabetes mellitus receiving statins, which increased LDL receptor (LDLR) expression. Nevertheless, it remains unclear how much LDLR expression contributes to the LDL-C-lowering effect of GLP-1RAs. We examined the effect of a GLP-1RA, namely, exendin-4, on serum LDL-C levels and its mechanism in Ldlr-/- and C57BL/6J mice. METHODS: Ten-week-old Ldlr-/- and C57BL/6J mice received exendin-4 or saline for 5 days, and serum lipid profiles and hepatic lipid levels were examined. Cholesterol metabolism-related gene expression and protein levels in the liver and ileum and the fecal bile acid (BA) composition were also examined. RESULTS: Exendin-4 treatment significantly decreased serum very-low-density lipoprotein cholesterol (VLDL-C) and LDL-C levels and mature hepatic SREBP2 levels and increased hepatic Insig1/2 mRNA expression in both mouse strains. In Ldlr-/- mice, exendin-4 treatment also significantly decreased hepatic cholesterol levels and fecal BA excretion, decreased hepatic Cyp7a1 mRNA expression, and increased small intestinal Fgf15 mRNA expression. In C57BL/6J mice, exendin-4 treatment significantly decreased small intestinal NPC1L1 levels. CONCLUSIONS: Our findings demonstrate that exendin-4 treatment decreased serum VLDL-C and LDL-C levels in a manner that was independent of LDLR. Exendin-4 treatment might decrease serum cholesterol levels by lowering hepatic SREBP2 levels and cholesterol absorption in Ldlr-/- and C57BL/6J mice. Exendin-4 treatment might decrease cholesterol absorption by different mechanisms in Ldlr-/- and C57BL/6J mice.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ratones , Animales , Exenatida , LDL-Colesterol , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ratones Endogámicos C57BL , Colesterol , Receptores de LDL/genética , Receptores de LDL/metabolismo , ARN Mensajero
6.
Sci Rep ; 12(1): 17530, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36266531

RESUMEN

Tissue optical clearing permits detailed evaluation of organ three-dimensional (3-D) structure as well as that of individual cells by tissue staining and autofluorescence. In this study, we evaluated intestinal morphology, intestinal epithelial cells (IECs), and enteroendocrine cells, such as incretin-producing cells, in reporter mice by intestinal 3-D imaging. 3-D intestinal imaging of reporter mice using optical tissue clearing enabled us to evaluate both detailed intestinal morphologies and cell numbers, villus length and crypt depth in the same samples. In disease mouse model of lipopolysaccharide (LPS)-injected mice, the results of 3-D imaging using tissue optical clearing in this study was consistent with those of 2-D imaging in previous reports and could added the new data of intestinal morphology. In analysis of incretin-producing cells of reporter mice, we could elucidate the number, the percentage, and the localization of incretin-producing cells in intestine and the difference of those between L cells and K cells. Thus, we established a novel method of intestinal analysis using tissue optical clearing and 3-D imaging. 3-D evaluation of intestine enabled us to clarify not only detailed intestinal morphology but also the precise number and localization of IECs and incretin-producing cells in the same samples.


Asunto(s)
Incretinas , Lipopolisacáridos , Ratones , Animales , Imagenología Tridimensional/métodos , Intestinos , Mucosa Intestinal/diagnóstico por imagen , Imagen Óptica/métodos
7.
Neurochem Res ; 47(9): 2793-2804, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35753011

RESUMEN

α1,3-Fucosyltransferase 9 (Fut9) is responsible for the synthesis of Lewis X [LeX, Galß1-4(Fucα1-3)GlcNAc] carbohydrate epitope, a marker for pluripotent or multipotent tissue-specific stem cells. Although Fut9-deficient mice show anxiety-related behaviors, structural and cellular abnormalities in the brain remain to be investigated. In this study, using in situ hybridization and immunohistochemical techniques in combination, we clarified the spatiotemporal expression of Fut9, together with LeX, in the brain and retina. We found that Fut9-expressing cells are positive for Ctip2, a marker of neurons residing in layer V/VI, and TLE4, a marker of corticothalamic projection neurons (CThPNs) in layer VI, of the cortex. A birthdating analysis using 5-ethynyl-2'-deoxyuridine at embryonic day (E)11.5, 5-bromo-2'-deoxyuridine at E12.5, and in utero electroporation of a GFP expression plasmid at E14.5 revealed a reduction in the percentage of neurons produced at E11.5 in layer VI/subplate of the cortex and in the ganglion cell layer of the retina in P0 Fut9-/- mice. Furthermore, this reduction in layer VI/subplate neurons persisted into adulthood, leading to a reduction in the number of Ctip2strong/Satb2- excitatory neurons in layer V/VI of the adult Fut9-/- cortex. These results suggest that Fut9 plays significant roles in the differentiation, migration, and maturation of neural precursor cells in the cortex and retina.


Asunto(s)
Antígeno Lewis X , Células-Madre Neurales , Animales , Corteza Cerebral/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Retina/metabolismo
8.
Nutrients ; 14(5)2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35267952

RESUMEN

(1) Background: Protein stimulates the secretion of glucagon (GCG), which can affect glucose metabolism. This study aimed to analyze the metabolic effect of a high-protein diet (HPD) in the presence or absence of proglucagon-derived peptides, including GCG and GLP-1. (2) Methods: The response to HPD feeding for 7 days was analyzed in mice deficient in proglucagon-derived peptides (GCGKO). (3) Results: In both control and GCGKO mice, food intake and body weight decreased with HPD and intestinal expression of Pepck increased. HPD also decreased plasma FGF21 levels, regardless of the presence of proglucagon-derived peptides. In control mice, HPD increased the hepatic expression of enzymes involved in amino acid metabolism without the elevation of plasma amino acid levels, except branched-chain amino acids. On the other hand, HPD-induced changes in the hepatic gene expression were attenuated in GCGKO mice, resulting in marked hyperaminoacidemia with lower blood glucose levels; the plasma concentration of glutamine exceeded that of glucose in HPD-fed GCGKO mice. (4) Conclusions: Increased plasma amino acid levels are a common feature in animal models with blocked GCG activity, and our results underscore that GCG plays essential roles in the homeostasis of amino acid metabolism in response to altered protein intake.


Asunto(s)
Dieta Rica en Proteínas , Glucagón , Animales , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Ratones , Péptidos , Proglucagón/genética , Proglucagón/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G617-G626, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33533304

RESUMEN

Glucagon-like peptide-1 (GLP-1) is an incretin secreted from enteroendocrine preproglucagon (PPG)-expressing cells (traditionally known as L cells) in response to luminal nutrients that potentiates insulin secretion. Augmentation of endogenous GLP-1 secretion might well represent a novel therapeutic target for diabetes treatment in addition to the incretin-associated drugs currently in use. In this study, we found that PPG cells substantially express carbonic anhydrase 8 (CAR8), which has been reported to inhibit inositol 1,4,5-trisphosphate (IP3) binding to the IP3 receptor and subsequent Ca2+ efflux from the endoplasmic reticulum in neuronal cells. In vitro experiments using STC-1 cells demonstrated that Car8 knockdown increases long-chain fatty acid (LCFA)-stimulated GLP-1 secretion. This effect was reduced in the presence of phospholipase C (PLC) inhibitor; in addition, Car8 knockdown increased the intracellular Ca2+ elevation caused by α-linolenic acid, indicating that CAR8 exerts its effect on GLP-1 secretion via the PLC/IP3/Ca2+ pathway. Car8wdl null mutant mice showed significant increase in GLP-1 response to oral corn oil administration compared with that in wild-type littermates, with no significant change in intestinal GLP-1 content. These results demonstrate that CAR8 negatively regulates GLP-1 secretion from PPG cells in response to LCFAs, suggesting the possibility of augmentation of postprandial GLP-1 secretion by CAR8 inhibition.NEW & NOTEWORTHY This study focused on the physiological significance of carbonic anhydrase 8 (CAR8) in GLP-1 secretion from enteroendocrine preproglucagon (PPG)-expressing cells. We found an inhibitory role of CAR8 in LCFA-induced GLP-1 secretion in vitro and in vivo, suggesting a novel therapeutic approach to diabetes and obesity through augmentation of postprandial GLP-1 secretion by CAR8 inhibition.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Aceite de Maíz/farmacología , Células Enteroendocrinas/efectos de los fármacos , Ácidos Grasos/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Biomarcadores de Tumor/genética , Señalización del Calcio , Línea Celular , Células Enteroendocrinas/enzimología , Glucagón/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Vías Secretoras , Fosfolipasas de Tipo C/metabolismo
10.
J Diabetes Investig ; 12(1): 32-34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32506830

RESUMEN

Glucagon promotes hepatic glucose production, lipolysis and amino acid catabolism. Inhibition of glucagon activity, or glucagon resistance, not only lowers blood glucose levels, but also induces non-alcoholic fatty liver disease and hyperaminoacidemia.


Asunto(s)
Glucagón , Lipólisis , Ácidos Grasos , Humanos , Receptores de Inositol 1,4,5-Trifosfato , Hígado
12.
Cereb Cortex ; 30(12): 6415-6425, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-32766673

RESUMEN

The origin and life-long fate of quiescent neural stem cells (NSCs) in the adult mammalian brain remain largely unknown. A few neural precursor cells in the embryonic brain elongate their cell cycle time and subsequently become quiescent postnatally, suggesting the possibility that life-long NSCs are selected at an early embryonic stage. Here, we utilized a GFP-expressing lentivirus to investigate the fate of progeny from individual lentivirus-infected NSCs by identifying the lentiviral integration site. Our data suggest that NSCs become specified to two or more lineages prior to embryonic day 13.5 in mice: one NSC lineage produces cells only for the cortex and another provides neurons to the olfactory bulb. The majority of neurosphere-forming NSCs in the adult brain are relatively dormant and generate very few cells, if any, in the olfactory bulb or cortex, and this NSC population could serve as a reservoir that is occasionally reactivated later in life.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Linaje de la Célula , Células-Madre Neurales/fisiología , Animales , Vectores Genéticos , Lentivirus/fisiología , Ratones Transgénicos
13.
Biochem Biophys Res Commun ; 532(1): 47-53, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32826056

RESUMEN

Although diabetic polyneuropathy (DPN) is the commonest diabetic complication, its pathology remains to be clarified. As previous papers have suggested the neuroprotective effects of glucagon-like peptide-1 in DPN, the current study investigated the physiological indispensability of glucagon gene-derived peptides (GCGDPs) including glucagon-like peptide-1 in the peripheral nervous system (PNS). Neurological functions and neuropathological changes of GCGDP deficient (gcg-/-) mice were examined. The gcg-/- mice showed tactile allodynia and thermal hyperalgesia at 12-18 weeks old, followed by tactile and thermal hypoalgesia at 36 weeks old. Nerve conduction studies revealed a decrease in sensory nerve conduction velocity at 36 weeks old. Pathological findings showed a decrease in intraepidermal nerve fiber densities. Electron microscopy revealed a decrease in circularity and an increase in g-ratio of myelinated fibers and a decrease of unmyelinated fibers in the sural nerves of the gcg-/- mice. Effects of glucagon on neurite outgrowth were examined using an ex vivo culture of dorsal root ganglia. A supraphysiological concentration of glucagon promoted neurite outgrowth. In conclusion, the mice with deficiency of GCGDPs developed peripheral neuropathy with age. Furthermore, glucagon might have neuroprotective effects on the PNS of mice. GCGDPs might be involved in the pathology of DPN.


Asunto(s)
Neuropatías Diabéticas/etiología , Péptidos Similares al Glucagón/deficiencia , Animales , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/patología , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Glucagón/deficiencia , Glucagón/genética , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/deficiencia , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Péptidos Similares al Glucagón/genética , Péptidos Similares al Glucagón/metabolismo , Hiperalgesia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Nerviosas Mielínicas/patología , Conducción Nerviosa , Proyección Neuronal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo
14.
J Diabetes Investig ; 10(6): 1391-1393, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31254453

RESUMEN

Glucagon plays an essential role in robust feedback regulation between the liver and α-cells, and exerts suppressive or static effects on the plasma concentration of amino acids, especially glutamine. Thereby, "glutaminostatin" might be an alternative name in recognition of another facet of glucagon as a suppressor of plasma glutamine levels.


Asunto(s)
Aminoácidos/sangre , Fármacos Gastrointestinales/farmacología , Células Secretoras de Glucagón/metabolismo , Glucagón/farmacología , Hígado/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos
15.
Nutrients ; 11(5)2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31083314

RESUMEN

Long-term exposure to a high starch, low-protein diet (HSTD) induces body weight gain and hyperinsulinemia concomitantly with an increase in ß-cell mass (BCM) and pancreatic islets number in mice; however, the effect of short-term exposure to HSTD on BCM and islet number has not been elucidated. In the present study, we investigated changes in body weight, plasma insulin levels, BCM and islet number in mice fed HSTD for 5 weeks followed by normal chow (NC) for 2 weeks. BCM and islet number were increased in mice fed HSTD for 5 weeks compared with those in mice fed NC. On the other hand, mice fed HSTD for 5 weeks followed by NC for 2 weeks (SN) showed decreased BCM and insulin levels, compared to mice fed HSTD for 7 weeks, and no significant differences in these parameters were observed between SN and the control NC at 7 weeks. No significant difference in body weight was observed among HSTD, NC and SN fed groups. These results suggest that a high-starch diet induces an increase in BCM in a manner independent of body weight gain, and that 2 weeks of NC feeding is sufficient for the reversal of the morphological changes induced in islets by HSTD feeding.


Asunto(s)
Dieta con Restricción de Proteínas , Células Secretoras de Insulina/efectos de los fármacos , Almidón/farmacología , Aumento de Peso , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Almidón/administración & dosificación , Factores de Tiempo
16.
FASEB J ; 33(5): 6239-6253, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30789757

RESUMEN

Our previous study demonstrated that sphingosine kinase 1-interacting protein (SKIP, or Sphkap) is expressed in pancreatic ß-cells, and depletion of SKIP enhances glucose-stimulated insulin secretion. We find here that SKIP is also expressed in intestinal K- and L-cells and that secretion of gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) as well as insulin are significantly increased, and blood glucose levels are decreased in SKIP-deficient (SKIP-/-) mice compared with those in wild-type mice. Plasma triglyceride (Tg), LDL cholesterol, and mRNA levels of proinflammatory cytokines in adipose tissues, livers, and intestines were found to be significantly decreased in SKIP-/- mice. The phenotypic characteristics of SKIP-/- mice, including adiposity and attenuation of basal inflammation, were abolished by genetic depletion of GIP. The improvement of glucose tolerance and lipid profiles in SKIP-/- mice were cancelled by GLP-1 receptor antagonist exendin-(9-39) treatment. In summary, depletion of SKIP ameliorates glucose tolerance by enhancing secretion of insulin and incretins, improves lipid metabolism, and reduces basal inflammation levels. Thus, inhibition of SKIP action may emerge as a new option for treatment of type 2 diabetes mellitus with metabolic dysfunction.-Liu, Y., Harashima, S., Wang, Y., Suzuki, K., Tokumoto, S., Usui, R., Tatsuoka, H., Tanaka, D., Yabe, D., Harada, N., Hayashi, Y., Inagaki, N. Sphingosine kinase 1-interacting protein is a dual regulator of insulin and incretin secretion.


Asunto(s)
Glucemia/metabolismo , Incretinas/metabolismo , Secreción de Insulina , Monoéster Fosfórico Hidrolasas/metabolismo , Tejido Adiposo/metabolismo , Animales , Colesterol/sangre , Citocinas/genética , Citocinas/metabolismo , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Masculino , Ratones , Monoéster Fosfórico Hidrolasas/genética
17.
Thyroid ; 29(4): 607-608, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30672388

RESUMEN

The co-occurrence of resistance to thyroid hormone beta (RTHß) and myotonic dystrophy type 1 (DM1) was observed in a Japanese family. Two mutations, P453A and C36Y, were identified in the thyroid hormone receptor beta (THRB) gene. Whereas family members with THRBP453A exhibited RTHß, two members with THRBC36Y but without THRBP453A had normal thyroid function. Two members, one with RTHß and the other without, had a triplet expansion in the dystrophia myotonia protein kinase gene, a hallmark of DM1. The member with both RTHß and DM1 developed atrial fibrillation at the age of 16 years, suggesting a synergistic impact on the heart.


Asunto(s)
Mutación , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Receptores beta de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/genética , Repeticiones de Trinucleótidos , Adolescente , Adulto , Fibrilación Atrial/etiología , Aleteo Atrial/etiología , Femenino , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Persona de Mediana Edad , Distrofia Miotónica/complicaciones , Distrofia Miotónica/diagnóstico , Linaje , Fenotipo , Síndrome de Resistencia a Hormonas Tiroideas/complicaciones , Síndrome de Resistencia a Hormonas Tiroideas/diagnóstico
18.
J Diabetes Investig ; 10(2): 238-250, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30084544

RESUMEN

AIMS/INTRODUCTION: A high-carbohydrate diet is known to increase insulin secretion and induce obesity. However, whether or not a high-carbohydrate diet affects ß-cell mass (BCM) has been little investigated. MATERIALS AND METHODS: Both wild-type (WT) mice and adenosine triphosphate-sensitive potassium channel-deficient (Kir6.2KO) mice were fed normal chow or high-starch (ST) diets for 22 weeks. BCM and the numbers of islets were analyzed by immunohistochemistry, and gene expression levels in islets were investigated by quantitative real-time reverse transcription polymerase chain reaction. MIN6-K8 ß-cells were stimulated in solution containing various concentrations of glucose combined with nifedipine and glimepiride, and gene expression was analyzed. RESULTS: Both WT and Kir6.2KO mice fed ST showed hyperinsulinemia and body weight gain. BCM, the number of islets and the expression levels of cyclinD2 messenger ribonucleic acid were increased in WT mice fed ST compared with those in WT mice fed normal chow. In contrast, no significant difference in BCM, the number of islets or the expression levels of cyclinD2 messenger ribonucleic acid were observed between Kir6.2KO mice fed normal chow and those fed ST. Incubation of MIN6-K8 ß-cells in high-glucose media or with glimepiride increased cyclinD2 expression, whereas nifedipine attenuated a high-glucose-induced increase in cyclinD2 expression. CONCLUSIONS: These results show that a high-starch diet increases BCM in an adenosine triphosphate-sensitive potassium channel-dependent manner, which is mediated through upregulation of cyclinD2 expression.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ciclina D2/metabolismo , Carbohidratos de la Dieta/efectos adversos , Células Secretoras de Insulina/patología , Canales de Potasio de Rectificación Interna/fisiología , Aumento de Peso , Animales , Glucemia/análisis , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
19.
J Neurochem ; 147(5): 584-594, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30028510

RESUMEN

In the adult mammalian brain, neural stem cells (NSCs) reside in two neurogenic regions, the walls of the lateral ventricles, and the subgranular zone of the hippocampus, which generate new neurons for the olfactory bulb and dentate gyrus, respectively. These adult NSCs retain their self-renewal ability and capacity to differentiate into neurons and glia as demonstrated by in vitro studies. However, their contribution to tissue repair in disease and injury is limited, lending credence to the claim by prominent neuropathologist Ramón y Cajal that 'once development was ended, the founts of growth and regeneration of the axons and dendrites dried up irrevocably'. However, recent progress toward understanding the fundamental biology of adult NSCs and their role in pathological conditions has provided new insight into the potential therapeutic utility of endogenous NSCs. In this short review, we highlight two topics: the altered behavior of NSCs after brain damage and the dysfunction of NSCs and oligodendrocyte precursor cells, another type of undifferentiated cell in the adult brain, in mood affective disorders.


Asunto(s)
Lesiones Encefálicas/patología , Encéfalo/patología , Trastornos Mentales/patología , Células-Madre Neurales/fisiología , Neurogénesis , Humanos , Trastornos del Humor/patología , Células-Madre Neurales/patología , Oligodendroglía/patología , Oligodendroglía/fisiología
20.
Am J Physiol Endocrinol Metab ; 314(6): E572-E583, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29406782

RESUMEN

Both high-fat (HFD) and high-carbohydrate (ST) diets are known to induce weight gain. Glucose-dependent insulinotropic polypeptide (GIP) is secreted mainly from intestinal K cells upon stimuli by nutrients such as fat and glucose, and it potentiates glucose-induced insulin secretion. GIP is well known to contribute to HFD-induced obesity. In this study, we analyzed the effect of ST feeding on GIP secretion and metabolic parameters to explore the role of GIP in ST-induced weight gain. Both wild-type (WT) and GIP receptor deficient ( GiprKO) mice were fed normal chow (NC), ST, or moderate (m)HFD for 22 wk. Body weight was measured, and then glucose tolerance tests were performed. Insulin secretion from isolated islets also was analyzed. WT mice fed ST or mHFD displayed weight gain concomitant with increased plasma GIP levels compared with WT mice fed NC. WT mice fed mHFD showed improved glucose tolerance due to enhanced insulin secretion during oral glucose tolerance tests compared with WT mice fed NC or ST. GiprKO mice fed mHFD did not display weight gain. On the other hand, GiprKO mice fed ST showed weight gain and did not display obvious glucose intolerance. Glucose-induced insulin secretion was enhanced during intraperitoneal glucose tolerance tests and from isolated islets in both WT and GiprKO mice fed ST compared with those fed NC. In conclusion, enhanced GIP secretion induced by mHFD-feeding contributes to increased insulin secretion and body weight gain, whereas GIP is marginally involved in weight gain induced by ST-feeding.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/farmacología , Polipéptido Inhibidor Gástrico/fisiología , Aumento de Peso/efectos de los fármacos , Animales , Carbohidratos de la Dieta/efectos adversos , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Insulina/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de la Hormona Gastrointestinal/genética , Receptores de la Hormona Gastrointestinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA