Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 621(7978): 389-395, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37648852

RESUMEN

Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes1,2. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance3-9. In particular, carbohydrate metabolism of commensals has been proposed to contribute up to 10% of the host's overall energy extraction10, thereby playing a role in the pathogenesis of obesity and prediabetes3,4,6. Nevertheless, the underlying mechanism remains unclear. Here we investigate this relationship using a comprehensive multi-omics strategy in humans. We combine unbiased faecal metabolomics with metagenomics, host metabolomics and transcriptomics data to profile the involvement of the microbiome in insulin resistance. These data reveal that faecal carbohydrates, particularly host-accessible monosaccharides, are increased in individuals with insulin resistance and are associated with microbial carbohydrate metabolisms and host inflammatory cytokines. We identify gut bacteria associated with insulin resistance and insulin sensitivity that show a distinct pattern of carbohydrate metabolism, and demonstrate that insulin-sensitivity-associated bacteria ameliorate host phenotypes of insulin resistance in a mouse model. Our study, which provides a comprehensive view of the host-microorganism relationships in insulin resistance, reveals the impact of carbohydrate metabolism by microbiota, suggesting a potential therapeutic target for ameliorating insulin resistance.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Microbioma Gastrointestinal , Resistencia a la Insulina , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Microbioma Gastrointestinal/fisiología , Resistencia a la Insulina/fisiología , Monosacáridos/metabolismo , Insulina/metabolismo , Síndrome Metabólico/metabolismo , Heces/química , Heces/microbiología , Metabolómica
2.
Immunity ; 44(6): 1422-33, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27317261

RESUMEN

Obesity has been shown to increase the morbidity of infections, however, the underlying mechanisms remain largely unknown. Here we demonstrate that obesity caused adiponectin deficiency in the bone marrow (BM), which led to an inflamed BM characterized by increased tumor necrosis factor (TNF) production from bone marrow macrophages. Hematopoietic stem and progenitor cells (HSPCs) chronically exposed to excessive TNF in obese marrow aberrantly expressed cytokine signaling suppressor SOCS3, impairing JAK-STAT mediated signal transduction and cytokine-driven cell proliferation. Accordingly, both obese and adiponectin-deficient mice showed attenuated clearance of infected Listeria monocytogenes, indicating that obesity or loss of adiponectin is critical for exacerbation of infection. Adiponectin treatment restored the defective HSPC proliferation and bacterial clearance of obese and adiponectin-deficient mice, affirming the importance of adiponectin against infection. Taken together, our findings demonstrate that obesity impairs hematopoietic response against infections through a TNF-SOCS3-STAT3 axis, highlighting adiponectin as a legitimate target against obesity-related infections.


Asunto(s)
Adiponectina/metabolismo , Células Madre Hematopoyéticas/fisiología , Inflamación/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Obesidad/inmunología , Adiponectina/genética , Animales , Bacteriólisis , Médula Ósea/inmunología , Células Cultivadas , Dieta , Regulación de la Expresión Génica , Hematopoyesis , Movilización de Célula Madre Hematopoyética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Stem Cells ; 35(7): 1835-1848, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28480607

RESUMEN

Myelotoxic injury, such as chemotherapeutic agents and ionizing radiation, unlocks the vigorous power of hematopoietic stem cells (HSCs) to replenish the hematopoietic system, making quiescent HSCs enter the cell cycle. Considering that both HSC-intrinsic and -extrinsic mechanisms enforce quiescence of HSCs, the drastic change in bone marrow (BM) environment after injury, represented by massive expansion of BM adipocytes, might trigger HSC activation. BM adipocytes, the major cellular component in the ablated marrow, however, reportedly suppress proliferation of hematopoietic cells, which may indicate the BM adipocytogenesis is an irrational response of injured organism. Given that adipose tissue is an endocrine organ with pleiotropic functions, we hypothesized that adipocyte-derived factors, especially adiponectin, an anti-inflammatory adipokine involved in regulation of granulopoiesis, are implicated in HSC activation. Myeloablative intervention increased BM adiponectin by multiple mechanisms, including adipocyte expansion and increased diffusion from the blood. Adiponectin-null (Adipoq -/- ) mice showed delayed hematopoietic recovery after BM injury, with Adipoq-/- HSCs more quiescent and defective in mammalian target of rapamycin complex 1 (mTORC1) activation. Recombinant adiponectin promoted not only HSC activation in vivo but cytokine-induced activation in vitro, and shortened the time for exit from quiescence in an mTORC1-dependent manner. These data illustrate a scarcely-reported example of a cell-extrinsic factor, adiponectin, enhancing quiescence exit of HSCs, and subsequent hematopoietic recovery. Our findings also highlight adipocytes as a source of adiponectin to ensure the proliferative burst of hematopoietic cells in ablated marrow. Stem Cells 2017;35:1835-1848.


Asunto(s)
Adiponectina/genética , Médula Ósea/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Adiponectina/deficiencia , Animales , Compuestos de Bencidrilo/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Ciclofosfamida/farmacología , Citarabina/farmacología , Compuestos Epoxi/farmacología , Fluorouracilo/farmacología , Regulación de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Agonistas Mieloablativos/farmacología , Poli I-C/farmacología , Transducción de Señal , Sirolimus/farmacología , Irradiación Corporal Total
4.
Proc Natl Acad Sci U S A ; 112(10): E1067-76, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713368

RESUMEN

Increase in the concentration of plasma L-cysteine is closely associated with defective insulin secretion from pancreatic ß-cells, which results in type 2 diabetes (T2D). In this study, we investigated the effects of prolonged L-cysteine treatment on glucose-stimulated insulin secretion (GSIS) from mouse insulinoma 6 (MIN6) cells and from mouse pancreatic islets, and found that the treatment reversibly inhibited glucose-induced ATP production and resulting GSIS without affecting proinsulin and insulin synthesis. Comprehensive metabolic analyses using capillary electrophoresis time-of-flight mass spectrometry showed that prolonged L-cysteine treatment decreased the levels of pyruvate and its downstream metabolites. In addition, methyl pyruvate, a membrane-permeable form of pyruvate, rescued L-cysteine-induced inhibition of GSIS. Based on these results, we found that both in vitro and in MIN6 cells, L-cysteine specifically inhibited the activity of pyruvate kinase muscle isoform 2 (PKM2), an isoform of pyruvate kinases that catalyze the conversion of phosphoenolpyruvate to pyruvate. L-cysteine also induced PKM2 subunit dissociation (tetramers to dimers/monomers) in cells, which resulted in impaired glucose-induced ATP production for GSIS. DASA-10 (NCGC00181061, a substituted N,N'-diarylsulfonamide), a specific activator for PKM2, restored the tetramer formation and the activity of PKM2, glucose-induced ATP production, and biphasic insulin secretion in L-cysteine-treated cells. Collectively, our results demonstrate that impaired insulin secretion due to exposure to L-cysteine resulted from its direct binding and inactivation of PKM2 and suggest that PKM2 is a potential therapeutic target for T2D.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Proteínas Portadoras/antagonistas & inhibidores , Cisteína/farmacología , Glucosa/farmacología , Insulina/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Animales , Línea Celular , Secreción de Insulina , Ratones , Hormonas Tiroideas , Proteínas de Unión a Hormona Tiroide
5.
Diabetologia ; 59(11): 2426-2434, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27525648

RESUMEN

AIMS/HYPOTHESIS: Recently, incretin-related agents have been reported to attenuate insulin resistance in animal models, although the underlying mechanisms remain unclear. In this study, we investigated whether anagliptin, the dipeptidyl peptidase 4 (DPP-4) inhibitor, attenuates skeletal muscle insulin resistance through endothelial nitric oxide synthase (eNOS) activation in the endothelial cells. We used endothelium-specific Irs2-knockout (ETIrs2KO) mice, which show skeletal muscle insulin resistance resulting from a reduction of insulin-induced skeletal muscle capillary recruitment as a consequence of impaired eNOS activation. METHODS: In vivo, 8-week-old male ETIrs2KO mice were fed regular chow with or without 0.3% (wt/wt) DPP-4 inhibitor for 8 weeks to assess capillary recruitment and glucose uptake by the skeletal muscle. In vitro, human coronary arterial endothelial cells (HCAECs) were used to explore the effect of glucagon-like peptide 1 (GLP-1) on eNOS activity. RESULTS: Treatment with anagliptin ameliorated the impaired insulin-induced increase in capillary blood volume, interstitial insulin concentration and skeletal muscle glucose uptake in ETIrs2KO mice. This improvement in insulin-induced glucose uptake was almost completely abrogated by the GLP-1 receptor (GLP-1R) antagonist exendin-(9-39). Moreover, the increase in capillary blood volume with anagliptin treatment was also completely inhibited by the NOS inhibitor. GLP-1 augmented eNOS phosphorylation in HCAECs, with the effect completely disappearing after exposure to the protein kinase A (PKA) inhibitor H89. These data suggest that anagliptin treatment enhances insulin-induced capillary recruitment and interstitial insulin concentrations, resulting in improved skeletal muscle glucose uptake by directly acting on the endothelial cells via NO- and GLP-1-dependent mechanisms in vivo. CONCLUSIONS/INTERPRETATION: Anagliptin may be a promising agent to ameliorate skeletal muscle insulin resistance in obese patients with type 2 diabetes.


Asunto(s)
Insulina/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Óxidos de Nitrógeno/metabolismo , Pirimidinas/farmacología , Animales , Dipeptidil Peptidasa 4/sangre , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas Sustrato del Receptor de Insulina/deficiencia , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Pirimidinas/sangre , Espectrometría de Masas en Tándem
6.
Nature ; 464(7293): 1313-9, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20357764

RESUMEN

Adiponectin is an anti-diabetic adipokine. Its receptors possess a seven-transmembrane topology with the amino terminus located intracellularly, which is the opposite of G-protein-coupled receptors. Here we provide evidence that adiponectin induces extracellular Ca(2+) influx by adiponectin receptor 1 (AdipoR1), which was necessary for subsequent activation of Ca(2+)/calmodulin-dependent protein kinase kinase beta (CaMKKbeta), AMPK and SIRT1, increased expression and decreased acetylation of peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha), and increased mitochondria in myocytes. Moreover, muscle-specific disruption of AdipoR1 suppressed the adiponectin-mediated increase in intracellular Ca(2+) concentration, and decreased the activation of CaMKK, AMPK and SIRT1 by adiponectin. Suppression of AdipoR1 also resulted in decreased PGC-1alpha expression and deacetylation, decreased mitochondrial content and enzymes, decreased oxidative type I myofibres, and decreased oxidative stress-detoxifying enzymes in skeletal muscle, which were associated with insulin resistance and decreased exercise endurance. Decreased levels of adiponectin and AdipoR1 in obesity may have causal roles in mitochondrial dysfunction and insulin resistance seen in diabetes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/metabolismo , Calcio/metabolismo , Mitocondrias/metabolismo , Receptores de Adiponectina/metabolismo , Sirtuina 1/metabolismo , Transactivadores/metabolismo , Animales , Señalización del Calcio , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina , Ratones , Células Musculares/citología , Células Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Oocitos/metabolismo , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Receptores de Adiponectina/deficiencia , Factores de Transcripción , Xenopus laevis
7.
Diabetologia ; 57(3): 542-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24317852

RESUMEN

AIMS/HYPOTHESIS: Common genetic variations of the transcription factor 7-like 2 gene (encoded by TCF7L2), one of the T cell factor/lymphoid enhancer-binding factor transcription factors for the converging wingless-type MMTV integration site family (Wnt)/ß-catenin signalling pathway, are known to be associated with type 2 diabetes. Individuals with at-risk alleles of TCF7L2 exhibit impaired insulin secretion. Although previous studies using animal models have revealed the existence of a relationship between the Wnt/ß-catenin signalling pathway and glucose homeostasis, it remains unclear whether TCF7L2 in the pancreatic beta cells might be causally involved in insulin secretion in vivo. In this study, we investigated the role of TCF7L2 expressed in the pancreatic beta cells in glucose homeostasis. METHODS: Three independent groups of genetically engineered mice (DN mice) were generated, in which expression of the dominant-negative form of Tcf7l2 was driven under a rat insulin promoter. Phenotypes of both adult and newborn mice were evaluated. The levels of genes and proteins expressed in isolated islets were determined by reverse transcription-quantitative PCR and western blot analysis, respectively. RESULTS: Adult DN mice showed impaired glucose tolerance and decreased insulin secretion in both oral and intraperitoneal glucose tolerance tests. Marked reduction of the beta cell area and whole-pancreas insulin content was observed in both the adult and newborn DN mice. Islets from the DN mice showed decreased gene expressions of Ccnd1, Ccnd2, Irs1, Irs2, Ins1, Ins2 and Mafa, consistent with the deleterious effects of the dominant-negative form of Tcf7l2 on beta cell proliferation and insulin production. CONCLUSIONS/INTERPRETATION: TCF7L2 expressed in the pancreatic beta cells plays a crucial role in glucose metabolism through regulation of the beta cell mass.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Western Blotting , Células Cultivadas , Regulación de la Expresión Génica , Homeostasis , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Ratones , Páncreas/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción TCF/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/genética , Vía de Señalización Wnt
8.
Nat Med ; 13(3): 332-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17268472

RESUMEN

Adiponectin plays a central role as an antidiabetic and antiatherogenic adipokine. AdipoR1 and AdipoR2 serve as receptors for adiponectin in vitro, and their reduction in obesity seems to be correlated with reduced adiponectin sensitivity. Here we show that adenovirus-mediated expression of AdipoR1 and R2 in the liver of Lepr(-/-) mice increased AMP-activated protein kinase (AMPK) activation and peroxisome proliferator-activated receptor (PPAR)-alpha signaling pathways, respectively. Activation of AMPK reduced gluconeogenesis, whereas expression of the receptors in both cases increased fatty acid oxidation and lead to an amelioration of diabetes. Alternatively, targeted disruption of AdipoR1 resulted in the abrogation of adiponectin-induced AMPK activation, whereas that of AdipoR2 resulted in decreased activity of PPAR-alpha signaling pathways. Simultaneous disruption of both AdipoR1 and R2 abolished adiponectin binding and actions, resulting in increased tissue triglyceride content, inflammation and oxidative stress, and thus leading to insulin resistance and marked glucose intolerance. Therefore, AdipoR1 and R2 serve as the predominant receptors for adiponectin in vivo and play important roles in the regulation of glucose and lipid metabolism, inflammation and oxidative stress in vivo.


Asunto(s)
Adiponectina/metabolismo , Marcación de Gen , Receptores de Superficie Celular/genética , Adiponectina/antagonistas & inhibidores , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Femenino , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Unión Proteica/genética , Receptores de Adiponectina , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Receptores de Leptina
9.
Diabetes ; 73(3): 474-489, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38064504

RESUMEN

Genome-wide association studies have identified several gene polymorphisms, including UBE2E2, associated with type 2 diabetes. Although UBE2E2 is one of the ubiquitin-conjugating enzymes involved in the process of ubiquitin modifications, the pathophysiological roles of UBE2E2 in metabolic dysfunction are not yet understood. Here, we showed upregulated UBE2E2 expression in the islets of a mouse model of diet-induced obesity. The diabetes risk allele of UBE2E2 (rs13094957) in noncoding regions was associated with upregulation of UBE2E2 mRNA in the human pancreas. Although glucose-stimulated insulin secretion was intact in the isolated islets, pancreatic ß-cell-specific UBE2E2-transgenic (TG) mice exhibited reduced insulin secretion and decreased ß-cell mass. In TG mice, suppressed proliferation of ß-cells before the weaning period and while receiving a high-fat diet was accompanied by elevated gene expression levels of p21, resulting in decreased postnatal ß-cell mass expansion and compensatory ß-cell hyperplasia, respectively. In TG islets, proteomic analysis identified enhanced formation of various types of polyubiquitin chains, accompanied by increased expression of Nedd4 E3 ubiquitin protein ligase. Ubiquitination assays showed that UBE2E2 mediated the elongation of ubiquitin chains by Nedd4. The data suggest that UBE2E2-mediated ubiquitin modifications in ß-cells play an important role in regulating glucose homeostasis and ß-cell mass.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Células Secretoras de Insulina , Ratones , Animales , Humanos , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estudio de Asociación del Genoma Completo , Proteómica , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Ratones Transgénicos , Dieta Alta en Grasa/efectos adversos , Ubiquitinas/genética , Ubiquitinas/metabolismo , Insulina/metabolismo
10.
Cell Metab ; 6(1): 55-68, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17618856

RESUMEN

Adiponectin has been shown to stimulate fatty acid oxidation and enhance insulin sensitivity through the activation of AMP-activated protein kinase (AMPK) in the peripheral tissues. The effects of adiponectin in the central nervous system, however, are still poorly understood. Here, we show that adiponectin enhances AMPK activity in the arcuate hypothalamus (ARH) via its receptor AdipoR1 to stimulate food intake; this stimulation of food intake by adiponectin was attenuated by dominant-negative AMPK expression in the ARH. Moreover, adiponectin also decreased energy expenditure. Adiponectin-deficient mice showed decreased AMPK phosphorylation in the ARH, decreased food intake, and increased energy expenditure, exhibiting resistance to high-fat-diet-induced obesity. Serum and cerebrospinal fluid levels of adiponectin and expression of AdipoR1 in the ARH were increased during fasting and decreased after refeeding. We conclude that adiponectin stimulates food intake and decreases energy expenditure during fasting through its effects in the central nervous system.


Asunto(s)
Adiponectina/fisiología , Ingestión de Alimentos , Hipotálamo/enzimología , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Adenoviridae/genética , Adiponectina/líquido cefalorraquídeo , Adiponectina/genética , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético , Femenino , Hipotálamo/patología , Técnicas para Inmunoenzimas , Hibridación in Situ , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Sondas ARN , Receptores de Adiponectina , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Leptina
12.
Nihon Rinsho ; 69(3): 563-72, 2011 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-21400857

RESUMEN

As the number of type 2 diabetes patients is still increasing in Japan, careful management and appropriate treatment of diabetes with oral hypoglycemic agents are required for all general physicians. Ultimate aim of treatment should be maintenance of a quality of life not different from that of persons without diabetes. Recently various new oral hypoglycemic agents have been developed and six different classes of agents are available now: sulfonylureas, biguanides, alpha-glucosidase inhibitors, glinides, thiazolidinediones, and dipeptidyl peptidase-4 inhibitors. In this article we describe the specific mechanisms of actions, side effects, and important points in clinical usage. Every drug therapy should be supported by lifestyle changes and the agents to be used should be selected after consideration of patient's patho-physiological state of diabetes, the presence of any complications, age, functional disorders of the liver, kidney, heart and lung, and the risk for hypoglycemia.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Administración Oral , Humanos
13.
Diabetes ; 70(8): 1640-1653, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33980693

RESUMEN

Insulin receptor substrate-1 (Irs1) is one of the major substrates for insulin receptor and insulin-like growth factor-1 (IGF-1) receptor tyrosine kinases. Systemic Irs1-deficient mice show growth retardation, with resistance to insulin and IGF-1, although the underlying mechanisms remain poorly understood. For this study, we generated mice with brain-specific deletion of Irs1 (NIrs1KO mice). The NIrs1KO mice exhibited lower body weights, shorter bodies and bone lengths, and decreased bone density. Moreover, the NIrs1KO mice exhibited increased insulin sensitivity and glucose utilization in the skeletal muscle. Although the ability of the pituitary to secrete growth hormone (GH) remained intact, the amount of hypothalamic growth hormone-releasing hormone (GHRH) was significantly decreased and, accordingly, the pituitary GH mRNA expression levels were impaired in these mice. Plasma GH and IGF-1 levels were also lower in the NIrs1KO mice. The expression levels of GHRH protein in the median eminence, where Irs1 antibody staining is observed, were markedly decreased in the NIrs1KO mice. In vitro, neurite elongation after IGF-1 stimulation was significantly impaired by Irs1 downregulation in the cultured N-38 hypothalamic neurons. In conclusion, brain Irs1 plays important roles in the regulation of neurite outgrowth of GHRH neurons, somatic growth, and glucose homeostasis.


Asunto(s)
Encéfalo/metabolismo , Trastornos del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/genética , Hipotálamo/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Resistencia a la Insulina/fisiología , Tejido Adiposo Blanco/metabolismo , Animales , Glucosa/metabolismo , Trastornos del Crecimiento/metabolismo , Hormona del Crecimiento/sangre , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Homeostasis/fisiología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Neuronas/metabolismo
14.
J Diabetes Investig ; 12(2): 266-276, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32621777

RESUMEN

AIMS/INTRODUCTION: To prevent diabetic complications, strict glucose control and frequent monitoring of blood glucose levels with invasive methods are necessary. We considered the monitoring of tear glucose levels might be a possible method for non-invasive glucose monitoring. To develop tear glucose monitoring for clinical application, we investigated the precise correlation between the blood and tear glucose concentrations. MATERIALS AND METHODS: A total of 10 participants and 20 participants with diabetes were admitted, and blood and tear samples were collected. Before statistical analysis, we eliminated tear samples contaminated with blood. We observed the daily blood and tear glucose dynamics, and carried out a random intercept model analysis to examine the association between the blood and tear glucose concentrations. RESULTS: Tear occult blood tests showed that the tear glucose concentrations and their variation increased in both participants with and without diabetes as contamination of blood increased. In both participants with and without diabetes, fluctuations of the plasma glucose concentrations were observed depending on the timing of collection of the samples, and the dynamics of the tear glucose concentrations paralleled those of the plasma glucose concentrations. The random intercept model analysis showed a significant association between the plasma and tear glucose concentrations in participants with diabetes (P < 0.001). This association still existed even after adjusting for the glycated hemoglobin levels and the prandial state (P < 0.001). CONCLUSIONS: It is important to eliminate the tear samples contaminated with blood. Tear glucose monitoring might be a reliable and non-invasive substitute method for monitoring the blood glucose concentrations for diabetes patients, irrespective of glycated hemoglobin levels and timing of sample collection.


Asunto(s)
Biomarcadores/análisis , Glucemia/análisis , Diabetes Mellitus/diagnóstico , Hemoglobina Glucada/análisis , Modelos Estadísticos , Sangre Oculta , Lágrimas/metabolismo , Adulto , Automonitorización de la Glucosa Sanguínea , Estudios de Casos y Controles , Diabetes Mellitus/epidemiología , Diabetes Mellitus/metabolismo , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Lágrimas/química
15.
J Am Heart Assoc ; 10(23): e020760, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34796738

RESUMEN

Background It is uncertain whether risk classification under the nationwide program on screening and lifestyle modification for metabolic syndrome captures well high-risk individuals who could benefit from lifestyle interventions. We examined the validity of risk classification by linking the incidence of cardiovascular disease (CVD). Methods and Results Individual-level data of 29 288 Japanese individuals aged 40 to 74 years without a history of CVD from 10 prospective cohort studies were used. Metabolic syndrome was defined as the presence of high abdominal obesity and/or overweight plus risk factors such as high blood pressure, high triglyceride or low high-density lipoprotein cholesterol levels, and high blood glucose levels. The risk categories for lifestyle intervention were information supply only, motivation-support intervention, and intensive support intervention. Sex- and age-specific hazard ratios and population attributable fractions of CVD, which were also further adjusted to consider non-high density lipoprotein cholesterol levels, were estimated with reference to nonobese/overweight individuals, using Cox proportional hazard regression. Since the reference category included those with risk factors, we set a supernormal group (nonobese/overweight with no risk factor) as another reference. We documented 1023 incident CVD cases (565 men and 458 women). The adjusted CVD risk was 60% to 70% higher in men and women aged 40 to 64 years receiving an intensive support intervention, and 30% higher in women aged 65 to 74 years receiving a motivation-support intervention, compared with nonobese/overweight individuals. The population attributable fractions in men and women aged 40 to 64 years receiving an intensive support intervention were 17.7% and 6.6%, respectively, while that in women aged 65 to 74 years receiving a motivation-support intervention was 9.4%. Compared with the supernormal group, nonobese/overweight individuals with risk factors had similar hazard ratios and population attributable fractions as individuals with metabolic syndrome. Conclusions Similar CVD excess and attributable risks among individuals with metabolic syndrome components in the absence and presence of obesity/overweight imply the need for lifestyle modification in both high-risk groups.


Asunto(s)
Enfermedades Cardiovasculares , Síndrome Metabólico , Obesidad , Adulto , Anciano , Enfermedades Cardiovasculares/epidemiología , Femenino , Humanos , Incidencia , Japón/epidemiología , Masculino , Síndrome Metabólico/epidemiología , Persona de Mediana Edad , Obesidad/epidemiología , Prevalencia , Estudios Prospectivos , Medición de Riesgo
16.
J Clin Invest ; 117(1): 246-57, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17200721

RESUMEN

Glucokinase (Gck) functions as a glucose sensor for insulin secretion, and in mice fed standard chow, haploinsufficiency of beta cell-specific Gck (Gck(+/-)) causes impaired insulin secretion to glucose, although the animals have a normal beta cell mass. When fed a high-fat (HF) diet, wild-type mice showed marked beta cell hyperplasia, whereas Gck(+/-) mice demonstrated decreased beta cell replication and insufficient beta cell hyperplasia despite showing a similar degree of insulin resistance. DNA chip analysis revealed decreased insulin receptor substrate 2 (Irs2) expression in HF diet-fed Gck(+/-) mouse islets compared with wild-type islets. Western blot analyses confirmed upregulated Irs2 expression in the islets of HF diet-fed wild-type mice compared with those fed standard chow and reduced expression in HF diet-fed Gck(+/-) mice compared with those of HF diet-fed wild-type mice. HF diet-fed Irs2(+/-) mice failed to show a sufficient increase in beta cell mass, and overexpression of Irs2 in beta cells of HF diet-fed Gck(+/-) mice partially prevented diabetes by increasing beta cell mass. These results suggest that Gck and Irs2 are critical requirements for beta cell hyperplasia to occur in response to HF diet-induced insulin resistance.


Asunto(s)
Grasas de la Dieta/farmacología , Glucoquinasa/fisiología , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/patología , Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/fisiología , Animales , Glucoquinasa/deficiencia , Glucoquinasa/genética , Humanos , Hiperplasia , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Transducción de Señal
17.
Endocr J ; 55(3): 515-22, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18446001

RESUMEN

Adiponectin has been proposed to act as an antidiabetic adipokine, suppressing gluconeogenesis and stimulating fatty acid oxidation in the liver and skeletal muscle. Although adiponectin-knockout (adipo(-/-)) mice are known to exhibit insulin resistance, the degrees of insulin resistance and glucose intolerance are unexpectedly only moderate. In this study, the adipo(-/-) mice showed hepatic, but not muscle, insulin resistance. insulin-stimulated phosphorylation of IRS-1 and IRS-2 was impaired, the IRS-2 protein level was decreased, and insulin-stimulated phosphorylation of Akt was decreased in the liver of the adipo(-/-) mice. However, the triglyceride content in the liver was not increased in these mice, despite the decrease in the PPARalpha expression involved in lipid combustion, since the expressions of lipogenic genes such as SREBP-1 and SCD-1 were decreased in association with the increased leptin sensitivity. Consistent with this, the down-regulation SREBP-1 and SCD-1 observed in the adipo(-/-) mice was no longer observed, and the hepatic triglyceride content was significantly increased in the adiponectin leptin double-knockout (adipo(-/-)ob/ob) mice. On the other hand, the triglyceride content in the skeletal muscle was significantly decreased in the adipo(-/-) mice, probably due to up-regulated AMPK activity associated with the increased leptin sensitivity. In fact, these phenotypes in the skeletal muscle of these mice were no longer observed in the adipo(-/-)ob/ob mice. In conclusion, adipo(-/-) mice showed impaired insulin signaling in the liver to cause hepatic insulin resistance, however, no increase in the triglyceride content was observed in either the liver or the skeletal muscle, presumably on account of the increased leptin sensitivity.


Asunto(s)
Resistencia a la Insulina/genética , Adiponectina/genética , Animales , Regulación hacia Abajo , Técnica de Clampeo de la Glucosa , Insulina/metabolismo , Leptina/genética , Lipogénesis/genética , Hígado/química , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Transducción de Señal/genética , Transducción de Señal/fisiología , Triglicéridos/análisis
18.
Nat Commun ; 9(1): 4863, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451856

RESUMEN

M2a-subtype macrophage activation is known to be impaired in obesity, although the underlying mechanisms remain poorly understood. Herein, we demonstrate that, the IL-4/Irs2/Akt pathway is selectively impaired, along with decreased macrophage Irs2 expression, although IL-4/STAT6 pathway is maintained. Indeed, myeloid cell-specific Irs2-deficient mice show impairment of IL-4-induced M2a-subtype macrophage activation, as a result of stabilization of the FoxO1/HDAC3/NCoR1 corepressor complex, resulting in insulin resistance under the HF diet condition. Moreover, the reduction of macrophage Irs2 expression is mediated by hyperinsulinemia via the insulin receptor (IR). In myeloid cell-specific IR-deficient mice, the IL-4/Irs2 pathway is preserved in the macrophages, which results in a reduced degree of insulin resistance, because of the lack of IR-mediated downregulation of Irs2. We conclude that downregulation of Irs2 in macrophages caused by hyperinsulinemia is responsible for systemic insulin resistance via impairment of M2a-subtype macrophage activation in obesity.


Asunto(s)
Hiperinsulinismo/genética , Proteínas Sustrato del Receptor de Insulina/genética , Interleucina-4/genética , Macrófagos/metabolismo , Obesidad/genética , Células 3T3-L1 , Animales , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Dieta Alta en Grasa/efectos adversos , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Proteínas Sustrato del Receptor de Insulina/deficiencia , Resistencia a la Insulina , Interleucina-4/metabolismo , Activación de Macrófagos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal
19.
J Clin Invest ; 114(7): 917-27, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467830

RESUMEN

We previously demonstrated that insulin receptor substrate 2 (Irs2) KO mice develop diabetes associated with hepatic insulin resistance, lack of compensatory beta cell hyperplasia, and leptin resistance. To more precisely determine the roles of Irs2 in beta cells and the hypothalamus, we generated beta cell-specific Irs2 KO and hypothalamus-specific Irs2 knockdown (betaHT-IRS2) mice. Expression of Irs2 mRNA was reduced by approximately 90% in pancreatic islets and was markedly reduced in the arcuate nucleus of the hypothalamus. By contrast, Irs2 expression in liver, muscle, and adipose tissue of betaHT-IRS2 mice was indistinguishable from that of control mice. The betaHT-IRS2 mice displayed obesity and leptin resistance. At 4 weeks of age, the betaHT-IRS2 mice showed normal insulin sensitivity, but at 8 and 12 weeks, they were insulin resistant with progressive obesity. Despite their normal insulin sensitivity at 8 weeks with caloric restriction, the betaHT-IRS2 mice exhibited glucose intolerance and impaired glucose-induced insulin secretion. beta Cell mass and beta cell proliferation in the betaHT-IRS2 mice were reduced significantly at 8 and 12 weeks but not at 10 days. Insulin secretion, normalized by cell number per islet, was significantly increased at high glucose concentrations in the betaHT-IRS2 mice. We conclude that, in beta cells and the hypothalamus, Irs2 is crucially involved in the regulation of beta cell mass and leptin sensitivity.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Islotes Pancreáticos/metabolismo , Fosfoproteínas/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Glucemia/metabolismo , Peso Corporal , Restricción Calórica , División Celular/fisiología , Dieta , Femenino , Intolerancia a la Glucosa/metabolismo , Hiperlipidemias/metabolismo , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/citología , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Fosfoproteínas/genética , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Distribución Tisular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Soc Sci Med ; 182: 45-51, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28412640

RESUMEN

BACKGROUND: Although living in a hilly environment may promote muscular activity in the daily lives of residents, and such activity may prevent diabetes mellitus, few studies have focused on the impact of living in a hilly environment on diabetes mellitus. The purpose of this study was to investigate the impact of a hilly neighborhood environment on DM in older people. METHODS: We used data from the Japan Gerontological Evaluation Study, a population-based, cross-sectional study of individuals aged 65 or older without long-term care needs in Japan, which was conducted in 2010. A total of 8904 participants in 46 neighborhoods had responded to the questionnaire and undergone a health check. Diabetes mellitus was diagnosed as HbA1c ≥ 6.5% and those undergoing treatment for diabetes mellitus. Poorly controlled diabetes mellitus was diagnosed in those without other chronic diseases who had an HbA1c > 7.5%, and in those with other chronic diseases if their HbA1c was >8.0%. Neighborhood environment was evaluated based on the percentage of positive responses in the questionnaire and geographical information system data. A multilevel analysis was performed, adjusted for individual-level risk factors. Furthermore, sensitivity analysis was conducted for those who were undergoing treatment for diabetes mellitus (n = 1007). RESULTS: After adjustment for other physical environmental and individual covariates, a 1 interquartile range increase (1.48°) in slope in the neighborhood decreased the risk of poorly controlled diabetes mellitus by 18% (odds ratio [OR]: 0.82, 95% confidence interval [CI]: 0.70-0.97). Sensitivity analysis confirmed that larger slopes in the neighborhood showed a significant protective effect against diabetes mellitus among those who were undergoing treatment for diabetes mellitus (OR: 0.73, 95% CI: 0.59-0.90). CONCLUSION: A hilly neighborhood environment was not associated with diabetes mellitus, but was protective against poorly controlled diabetes mellitus.


Asunto(s)
Diabetes Mellitus/epidemiología , Ambiente , Mapeo Geográfico , Características de la Residencia/estadística & datos numéricos , Anciano , Anciano de 80 o más Años , Estudios Transversales , Escolaridad , Empleo/métodos , Empleo/estadística & datos numéricos , Femenino , Hemoglobina Glucada/análisis , Humanos , Renta/estadística & datos numéricos , Japón/epidemiología , Masculino , Análisis Multinivel , Oportunidad Relativa , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA