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










Base de datos
Intervalo de año de publicación
1.
Endocrinology ; 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38878275

RESUMEN

Genes regulating body fat are shared with high fidelity by mice and humans, indicating that mouse knockout (KO) phenotyping might identify valuable anti-obesity drug targets. Male Mrs2 magnesium transporter (Mrs2) KO mice were recently reported as thin when fed high-fat diet (HFD). They also exhibited increased energy expenditure (EE)/body weight and had beiged adipocytes that, along with isolated hepatocytes, demonstrated increased oxygen consumption suggesting increased EE drove the thin phenotype. Here we provide our data on these and additional assays in Mrs2 KO mice. We generated Mrs2 KO mice by homologous recombination. HFD-fed male and female Mrs2 KO mice had significantly less body fat, measured by QMR, than WT littermates. HFD-fed Mrs2 KO mice did not demonstrate increased EE by indirect calorimetry and could not maintain body temperature at 4°C, consistent with their decreased BAT stores but despite increased beige WAT. Instead, when provided a choice between HFD and low-fat diet (LFD), Mrs2 KO mice showed a significant 15% decrease in total energy intake resulting from significantly lower HFD intake that offset numerically increased LFD intake. Food restriction studies performed using WT mice suggested that this decrease in energy intake could explain the loss of body fat. OGTT studies revealed significantly improved insulin sensitivity in Mrs2 KO mice. We conclude that HFD-fed Mrs2 KO mice are thin with improved insulin sensitivity, and that this favorable metabolic phenotype is driven by hypophagia. Further evaluation is warranted to determine the suitability of MRS2 as a drug target for anti-obesity therapeutics.

2.
Diabetes Metab Syndr Obes ; 15: 45-58, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35023939

RESUMEN

PURPOSE: Humans with haploinsufficiency of GPR75, an orphan GPCR, are thin. Gpr75 knockout (KO) mice are also thin with improved glucose homeostasis. We wanted to confirm these findings in Gpr75 KO mice and determine whether decreased energy intake and/or increased energy expenditure contributed to the thin phenotype. METHODS: Gpr75 KO mice were generated by homologous recombination. All studies compared female and male Gpr75 KO mice to their wild type (WT) littermates. Body composition was measured by DXA and QMR technologies. Glucose homeostasis was evaluated by measuring glucose and insulin levels during oral glucose tolerance tests (OGTTs). Food intake was measured in group-housed mice. In singly housed mice, energy expenditure was measured in Oxymax indirect calorimetry chambers, and locomotor activity was measured in Oxymax and Photobeam Activity System chambers. RESULTS: In all 12 cohorts of adult female or male mice, Gpr75 KO mice had less body fat; pooled data showed that, compared to WT littermates (n = 103), Gpr75 KO mice (n = 118) had 49% less body fat and 4% less LBM (P < 0.001 for each). KO mice also had 8% less body fat at weaning (P < 0.05), and during the month after weaning as the thin phenotype became more exaggerated, Gpr75 KO mice ate significantly less than, but had energy expenditure and activity levels comparable to, their WT littermates. During OGTTs, Gpr75 KO mice showed improved glucose tolerance (glucose AUC 23% lower in females, P < 0.05, and 26% lower in males, P < 0.001), accompanied by significantly decreased insulin levels and significantly increased insulin sensitivity indices. CONCLUSION: Gpr75 KO mice are thin at weaning, are hypophagic as the thin phenotype becomes more exaggerated, and exhibit improved glucose tolerance and insulin sensitivity as healthy-appearing adults. These results suggest that inhibiting GPR75 in obese humans may safely decrease energy intake and body fat while improving glucose tolerance and insulin sensitivity.

3.
Diabetes Metab Syndr Obes ; 14: 3753-3785, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34483672

RESUMEN

PURPOSE: Obesity is a major public health problem. Understanding which genes contribute to obesity may better predict individual risk and allow development of new therapies. Because obesity of a mouse gene knockout (KO) line predicts an association of the orthologous human gene with obesity, we reviewed data from the Lexicon Genome5000TM high throughput phenotypic screen (HTS) of mouse gene KOs to identify KO lines with high body fat. MATERIALS AND METHODS: KO lines were generated using homologous recombination or gene trapping technologies. HTS body composition analyses were performed on adult wild-type and homozygous KO littermate mice from 3758 druggable mouse genes having a human ortholog. Body composition was measured by either DXA or QMR on chow-fed cohorts from all 3758 KO lines and was measured by QMR on independent high fat diet-fed cohorts from 2488 of these KO lines. Where possible, comparisons were made to HTS data from the International Mouse Phenotyping Consortium (IMPC). RESULTS: Body fat data are presented for 75 KO lines. Of 46 KO lines where independent external published and/or IMPC KO lines are reported as obese, 43 had increased body fat. For the remaining 29 novel high body fat KO lines, Ksr2 and G2e3 are supported by data from additional independent KO cohorts, 6 (Asnsd1, Srpk2, Dpp8, Cxxc4, Tenm3 and Kiss1) are supported by data from additional internal cohorts, and the remaining 21 including Tle4, Ak5, Ntm, Tusc3, Ankk1, Mfap3l, Prok2 and Prokr2 were studied with HTS cohorts only. CONCLUSION: These data support the finding of high body fat in 43 independent external published and/or IMPC KO lines. A novel obese phenotype was identified in 29 additional KO lines, with 27 still lacking the external confirmation now provided for Ksr2 and G2e3 KO mice. Undoubtedly, many mammalian obesity genes remain to be identified and characterized.

4.
Bone Res ; 7: 2, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30622831

RESUMEN

The disability, mortality and costs caused by non-vertebral osteoporotic fractures are enormous. Existing osteoporosis therapies are highly effective at reducing vertebral but not non-vertebral fractures. Cortical bone is a major determinant of non-vertebral bone strength. To identify novel osteoporosis drug targets, we phenotyped cortical bone of 3 366 viable mouse strains with global knockouts of druggable genes. Cortical bone thickness was substantially elevated in Notum -/- mice. NOTUM is a secreted WNT lipase and we observed high NOTUM expression in cortical bone and osteoblasts but not osteoclasts. Three orally active small molecules and a neutralizing antibody inhibiting NOTUM lipase activity were developed. They increased cortical bone thickness and strength at multiple skeletal sites in both gonadal intact and ovariectomized rodents by stimulating endocortical bone formation. Thus, inhibition of NOTUM activity is a potential novel anabolic therapy for strengthening cortical bone and preventing non-vertebral fractures.

5.
Endocrinology ; 157(12): 4534-4541, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27754787

RESUMEN

Polymorphisms in the SLC30A8 gene, which encodes the ZnT8 zinc transporter, are associated with altered susceptibility to type 2 diabetes (T2D), and SLC30A8 haploinsufficiency is protective against the development of T2D in obese humans. SLC30A8 is predominantly expressed in pancreatic islet ß-cells, but surprisingly, multiple knockout mouse studies have shown little effect of Slc30a8 deletion on glucose tolerance or glucose-stimulated insulin secretion (GSIS). Multiple other Slc30a isoforms are expressed at low levels in pancreatic islets. We hypothesized that functional compensation by the Slc30a7 isoform, which encodes ZnT7, limits the impact of Slc30a8 deletion on islet function. We therefore analyzed the effect of Slc30a7 deletion alone or in combination with Slc30a8 on in vivo glucose metabolism and GSIS in isolated islets. Deletion of Slc30a7 alone had complex effects in vivo, impairing glucose tolerance and reducing the glucose-stimulated increase in plasma insulin levels, hepatic glycogen levels, and pancreatic insulin content. Slc30a7 deletion also affected islet morphology and increased the ratio of islet α- to ß-cells. However, deletion of Slc30a7 alone had no effect on GSIS in isolated islets, whereas combined deletion of Slc30a7 and Slc30a8 abolished GSIS. These data demonstrate that the function of ZnT8 in islets can be unmasked by removal of ZnT7 and imply that ZnT8 may affect T2D susceptibility through actions in other tissues where it is expressed at low levels rather than through effects on pancreatic islet function.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Peso Corporal/genética , Proteínas de Transporte de Catión/genética , Femenino , Células Secretoras de Glucagón/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Factores Sexuales , Transportador 8 de Zinc
6.
J Pharmacol Exp Ther ; 345(2): 250-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23487174

RESUMEN

LX4211 [(2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(methylthio)tetrahydro-2H-pyran-3,4,5-triol], a dual sodium/glucose cotransporter 1 (SGLT1) and SGLT2 inhibitor, is thought to decrease both renal glucose reabsorption by inhibiting SGLT2 and intestinal glucose absorption by inhibiting SGLT1. In clinical trials in patients with type 2 diabetes mellitus (T2DM), LX4211 treatment improved glycemic control while increasing circulating levels of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). To better understand how LX4211 increases GLP-1 and PYY levels, we challenged SGLT1 knockout (-/-) mice, SGLT2-/- mice, and LX4211-treated mice with oral glucose. LX4211-treated mice and SGLT1-/- mice had increased levels of plasma GLP-1, plasma PYY, and intestinal glucose during the 6 hours after a glucose-containing meal, as reflected by area under the curve (AUC) values, whereas SGLT2-/- mice showed no response. LX4211-treated mice and SGLT1-/- mice also had increased GLP-1 AUC values, decreased glucose-dependent insulinotropic polypeptide (GIP) AUC values, and decreased blood glucose excursions during the 6 hours after a challenge with oral glucose alone. However, GLP-1 and GIP levels were not increased in LX4211-treated mice and were decreased in SGLT1-/- mice, 5 minutes after oral glucose, consistent with studies linking decreased intestinal SGLT1 activity with reduced GLP-1 and GIP levels 5 minutes after oral glucose. These data suggest that LX4211 reduces intestinal glucose absorption by inhibiting SGLT1, resulting in net increases in GLP-1 and PYY release and decreases in GIP release and blood glucose excursions. The ability to inhibit both intestinal SGLT1 and renal SGLT2 provides LX4211 with a novel dual mechanism of action for improving glycemic control in patients with T2DM.


Asunto(s)
Péptido 1 Similar al Glucagón/sangre , Glucosa/metabolismo , Glicósidos/farmacología , Hipoglucemiantes/farmacología , Absorción Intestinal/efectos de los fármacos , Péptido YY/sangre , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Área Bajo la Curva , Glucemia/metabolismo , Dieta , Polipéptido Inhibidor Gástrico/metabolismo , Prueba de Tolerancia a la Glucosa , Glucosuria/metabolismo , Metilglucósidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodo Posprandial/fisiología , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/fisiología , Transportador 2 de Sodio-Glucosa/genética
7.
Am J Physiol Endocrinol Metab ; 304(2): E117-30, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23149623

RESUMEN

Sodium-glucose cotransporter 2 (SGLT2) is the major, and SGLT1 the minor, transporter responsible for renal glucose reabsorption. Increasing urinary glucose excretion (UGE) by selectively inhibiting SGLT2 improves glycemic control in diabetic patients. We generated Sglt1 and Sglt2 knockout (KO) mice, Sglt1/Sglt2 double-KO (DKO) mice, and wild-type (WT) littermates to study their relative glycemic control and to determine contributions of SGLT1 and SGLT2 to UGE. Relative to WTs, Sglt2 KOs had improved oral glucose tolerance and were resistant to streptozotocin-induced diabetes. Sglt1 KOs fed glucose-free high-fat diet (G-free HFD) had improved oral glucose tolerance accompanied by delayed intestinal glucose absorption and increased circulating glucagon-like peptide-1 (GLP-1), but had normal intraperitoneal glucose tolerance. On G-free HFD, Sglt2 KOs had 30%, Sglt1 KOs 2%, and WTs <1% of the UGE of DKOs. Consistent with their increased UGE, DKOs had lower fasting blood glucose and improved intraperitoneal glucose tolerance than Sglt2 KOs. In conclusion, 1) Sglt2 is the major renal glucose transporter, but Sglt1 reabsorbs 70% of filtered glucose if Sglt2 is absent; 2) mice lacking Sglt2 display improved glucose tolerance despite UGE that is 30% of maximum; 3) Sglt1 KO mice respond to oral glucose with increased circulating GLP-1; and 4) DKO mice have improved glycemic control over mice lacking Sglt2 alone. These data suggest that, in patients with type 2 diabetes, combining pharmacological SGLT2 inhibition with complete renal and/or partial intestinal SGLT1 inhibition may improve glycemic control over that achieved by SGLT2 inhibition alone.


Asunto(s)
Glucemia/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/genética , Animales , Glucemia/genética , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/orina , Femenino , Péptido 1 Similar al Glucagón/farmacología , Prueba de Tolerancia a la Glucosa , Glucosuria/genética , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transportador 1 de Sodio-Glucosa/fisiología , Transportador 2 de Sodio-Glucosa/fisiología , Estreptozocina
8.
PLoS One ; 7(7): e40972, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22829903

RESUMEN

OBJECTIVE: The SLC30A8 gene encodes the islet-specific transporter ZnT-8, which is hypothesized to provide zinc for insulin-crystal formation. A polymorphic variant in SLC30A8 is associated with altered susceptibility to type 2 diabetes. Several groups have examined the effect of global Slc30a8 gene deletion but the results have been highly variable, perhaps due to the mixed 129SvEv/C57BL/6J genetic background of the mice studied. We therefore sought to remove the conflicting effect of 129SvEv-specific modifier genes. METHODS: The impact of Slc30a8 deletion was examined in the context of the pure C57BL/6J genetic background. RESULTS: Male C57BL/6J Slc30a8 knockout (KO) mice had normal fasting insulin levels and no change in glucose-stimulated insulin secretion (GSIS) from isolated islets in marked contrast to the ∼50% and ∼35% decrease, respectively, in both parameters observed in male mixed genetic background Slc30a8 KO mice. This observation suggests that 129SvEv-specific modifier genes modulate the impact of Slc30a8 deletion. In contrast, female C57BL/6J Slc30a8 KO mice had reduced (∼20%) fasting insulin levels, though this was not associated with a change in fasting blood glucose (FBG), or GSIS from isolated islets. This observation indicates that gender also modulates the impact of Slc30a8 deletion, though the physiological explanation as to why impaired insulin secretion is not accompanied by elevated FBG is unclear. Neither male nor female C57BL/6J Slc30a8 KO mice showed impaired glucose tolerance. CONCLUSIONS: Our data suggest that, despite a marked reduction in islet zinc content, the absence of ZnT-8 does not have a substantial impact on mouse physiology.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Ayuno/sangre , Insulina/sangre , Animales , Glucemia/metabolismo , Proteínas de Transporte de Catión/genética , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Insulina/genética , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Sexuales , Zinc/metabolismo , Transportador 8 de Zinc
9.
Mol Cell Biol ; 31(1): 63-80, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20974805

RESUMEN

An early lesion in many kidney diseases is damage to podocytes, which are critical components of the glomerular filtration barrier. A number of proteins are essential for podocyte filtration function, but the signaling events contributing to development of nephrotic syndrome are not well defined. Here we show that class II phosphoinositide 3-kinase C2α (PI3KC2α) is expressed in podocytes and plays a critical role in maintaining normal renal homeostasis. PI3KC2α-deficient mice developed chronic renal failure and exhibited a range of kidney lesions, including glomerular crescent formation and renal tubule defects in early disease, which progressed to diffuse mesangial sclerosis, with reduced podocytes, widespread effacement of foot processes, and modest proteinuria. These findings were associated with altered expression of nephrin, synaptopodin, WT-1, and desmin, indicating that PI3KC2α deficiency specifically impacts podocyte morphology and function. Deposition of glomerular IgA was observed in knockout mice; importantly, however, the development of severe glomerulonephropathy preceded IgA production, indicating that nephropathy was not directly IgA mediated. PI3KC2α deficiency did not affect immune responses, and bone marrow transplantation studies also indicated that the glomerulonephropathy was not the direct consequence of an immune-mediated disease. Thus, PI3KC2α is critical for maintenance of normal glomerular structure and function by supporting normal podocyte function.


Asunto(s)
Glomérulos Renales/anatomía & histología , Glomérulos Renales/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Animales , Antígenos de Superficie/metabolismo , Trasplante de Médula Ósea , Glomerulonefritis/etiología , Glomerulonefritis/patología , Glomerulonefritis/fisiopatología , Glomerulonefritis por IGA/etiología , Glomerulonefritis por IGA/patología , Glomerulonefritis por IGA/fisiopatología , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina G/metabolismo , Glomérulos Renales/enzimología , Glomérulos Renales/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Podocitos/enzimología , Podocitos/patología , Podocitos/fisiología , Insuficiencia Renal/etiología , Insuficiencia Renal/patología , Insuficiencia Renal/fisiopatología , Quimera por Trasplante
10.
Obesity (Silver Spring) ; 19(5): 1010-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21127480

RESUMEN

The kinase suppressor of ras 2 (KSR2) gene resides at human chromosome 12q24, a region linked to obesity and type 2 diabetes (T2D). While knocking out and phenotypically screening mouse orthologs of thousands of druggable human genes, we found KSR2 knockout (KSR2(-/-)) mice to be more obese and glucose intolerant than melanocortin 4 receptor(-/-) (MC4R(-/-)) mice. The obesity and T2D of KSR2(-/-) mice resulted from hyperphagia which was unresponsive to leptin and did not originate downstream of MC4R. The kinases AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) are each linked to food intake regulation, but only mTOR had increased activity in KSR2(-/-) mouse brain, and the ability of rapamycin to inhibit food intake in KSR2(-/-) mice further implicated mTOR in this process. The metabolic phenotype of KSR2 heterozygous (KSR2(+/minus;)) and KSR2(-/-) mice suggests that human KSR2 variants may contribute to a similar phenotype linked to human chromosome 12q24.


Asunto(s)
Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hiperfagia/metabolismo , Obesidad/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Ingestión de Alimentos/genética , Leptina/metabolismo , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética
11.
J Pharmacol Exp Ther ; 333(3): 844-53, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20190014

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are involved in the regulation of lipid and glucose metabolism. PPARgamma agonists improve insulin sensitivity and hyperglycemia and are effective in treating type 2 diabetes mellitus (T2DM), whereas PPARalpha agonists are used to treat dyslipidemia and atherosclerosis. The goal here was to examine the efficacy of a selective PPARalpha agonist {(S)-3-[3-(1-carboxy-1-methyl-ethoxy)-phenyl]-piperidine-1-carboxylic acid 4-trifluoromethyl-benzyl ester; CP-900691} on lipid, glycemic, and inflammation indices in 14 cynomolgus monkeys with spontaneous T2DM maintained on daily insulin therapy. Monkeys were dosed orally with either vehicle (n = 7) or CP-900691 (3 mg/kg, n = 7) daily for 6 weeks. CP-900691 treatment increased plasma high-density lipoprotein cholesterol (HDLC) (33 +/- 3 to 60 +/- 4 mg/dL, p < 0.001) and apolipoprotein A1 (96 +/- 5 to 157 +/- 5 mg/dL, p < 0.001), reduced plasma triglycerides (547 +/- 102 to 356 +/- 90 mg/dL, p < 0.01), and apolipoprotein B (62 +/- 3 to 45 +/- 3 mg/dL, p < 0.01), improved the lipoprotein index (HDL to non-HDLC ratio; 0.28 +/- 0.06 to 0.79 +/- 0.16, p < 0.001), decreased body weight (p < 0.01) and C-reactive protein (CRP) (1700 +/- 382 to 304 +/- 102 ng/ml, p < 0.01), and increased adiponectin (1697 +/- 542 to 4242 +/- 1070 ng/ml, p < 0.001) compared with baseline. CP-900691 treatment reduced exogenous insulin requirements by approximately 25% (p < 0.04) while lowering plasma fructosamine from 2.87 +/- 0.09 to 2.22 +/- 0.17 mM (p < 0.05), indicative of improved glycemic control. There were no changes in any of the aforementioned parameters in the vehicle group. Because low HDLC and high triglycerides are well established risk factors for cardiovascular disease, the marked improvements in these parameters, and in glycemic control, body weight, and CRP, suggest that CP-900691 may be of benefit in diabetic and obese or hyperlipidemic populations.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Hipolipemiantes , Lípidos/sangre , Lipoproteínas/sangre , PPAR alfa/agonistas , Piperidinas/farmacología , Propionatos/farmacología , Adiponectina/sangre , Animales , Área Bajo la Curva , Proteína C-Reactiva/metabolismo , Diabetes Mellitus Tipo 2/genética , Relación Dosis-Respuesta a Droga , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Macaca fascicularis , Pérdida de Peso/efectos de los fármacos
12.
J Med Chem ; 52(20): 6201-4, 2009 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-19785435

RESUMEN

The prevalence of diabetes throughout the world continues to increase and has become a major health issue. Recently there have been several reports of inhibitors directed toward the sodium-dependent glucose cotransporter 2 (SGLT2) as a method of maintaining glucose homeostasis in diabetic patients. Herein we report the discovery of the novel O-xyloside 7c that inhibits SGLT2 in vitro and urinary glucose reabsorption in vivo.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Xilosa/análogos & derivados , Xilosa/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/orina , Descubrimiento de Drogas , Glucosa/metabolismo , Humanos , Ratones , Especificidad por Sustrato , Xilosa/administración & dosificación , Xilosa/uso terapéutico
13.
Biochem J ; 421(3): 371-6, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19450229

RESUMEN

The Slc30a8 gene encodes the islet-specific zinc transporter ZnT-8, which provides zinc for insulin-hexamer formation. Polymorphic variants in amino acid residue 325 of human ZnT-8 are associated with altered susceptibility to Type 2 diabetes and ZnT-8 autoantibody epitope specificity changes in Type 1 diabetes. To assess the physiological importance of ZnT-8, mice carrying a Slc30a8 exon 3 deletion were analysed histologically and phenotyped for energy metabolism and pancreatic hormone secretion. No gross anatomical or behavioural changes or differences in body weight were observed between wild-type and ZnT-8-/- mice, and ZnT-8-/- mouse islets were indistinguishable from wild-type in terms of their numbers, size and cellular composition. However, total zinc content was markedly reduced in ZnT-8-/- mouse islets, as evaluated both by Timm's histochemical staining of pancreatic sections and direct measurements in isolated islets. Blood glucose levels were unchanged in 16-week-old, 6 h fasted animals of either gender; however, plasma insulin concentrations were reduced in both female (approximately 31%) and male (approximately 47%) ZnT-8-/- mice. Intraperitoneal glucose tolerance tests demonstrated no impairment in glucose clearance in male ZnT-8-/- mice, but glucose-stimulated insulin secretion from isolated islets was reduced approximately 33% relative to wild-type littermates. In summary, Slc30a8 gene deletion is accompanied by a modest impairment in insulin secretion without major alterations in glucose metabolism.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Eliminación de Secuencia , Animales , Glucemia , Diabetes Mellitus/genética , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Zinc/metabolismo , Transportador 8 de Zinc
14.
Nutr Res ; 28(6): 368-76, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19083434

RESUMEN

Consuming soy and soy isoflavones has been shown to cause modest improvements in plasma lipids, lipoproteins, and indices of insulin sensitivity in postmenopausal women. The effect of soy on such end points is attributed often to estrogen receptor agonism by isoflavones. Recent in vitro studies suggest that isoflavones, in combination with high estrogen concentrations (within the range seen circulating in premenopausal women), function as estrogen receptor antagonists that potentially may counteract the beneficial effects seen with soy consumption. We studied insulin sensitivity in 15 premenopausal nonhuman primates consuming either a high isoflavone soy diet or a soy-free casein/lactalbumin diet for 4 months. Insulin sensitivity was measured by intravenous glucose tolerance testing, hyperinsulinemic-euglycemic clamps, and insulin-stimulated insulin receptor and protein kinase B phosphorylation levels in muscle. In addition, plasma lipids, adiponectin, thyroid hormone, and body weights are reported. We show that high isoflavones do not adversely affect insulin sensitivity but do significantly alter insulin secretion to glucose stimulation. Small but significant increases in thyroxine and increased high-density lipoprotein cholesterol were observed as has been reported commonly with soy intake. These study results demonstrate that consumption of soy containing high isoflavone levels is not associated with changes in insulin sensitivity in the high estrogen milieu of the premenopausal female.


Asunto(s)
Moduladores de los Receptores de Estrógeno/uso terapéutico , Glycine max , Hiperinsulinismo/dietoterapia , Insulina/metabolismo , Isoflavonas/uso terapéutico , Fitoterapia , Extractos Vegetales/uso terapéutico , Proteínas de Soja/uso terapéutico , Adiponectina/análisis , Adiponectina/sangre , Animales , Glucemia/análisis , Peso Corporal , HDL-Colesterol/sangre , Femenino , Prueba de Tolerancia a la Glucosa , Haplorrinos , Resistencia a la Insulina/fisiología , Secreción de Insulina , Lípidos/análisis , Lípidos/sangre , Premenopausia/fisiología , Semillas , Tiroxina/sangre
15.
Metabolism ; 57(7 Suppl 1): S24-31, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18555850

RESUMEN

Isoflavones may influence insulin action by means of their well-known receptor-mediated estrogenic activity. However, isoflavones also bind to peroxisome proliferator-activated receptors (PPARs) that are strongly associated with insulin action. Soy protein with its isoflavones has previously been shown to improve glycemic control in diabetic postmenopausal women and to improve insulin sensitivity in ovariectomized monkeys. The purpose of the current report was to extend our studies of dietary soy protein to male monkeys and determine effects of the soy isoflavones on insulin resistance. Two studies are reported here. Study one involved 91 male monkeys consuming 3 diets differing only by the source of protein (casein-lactalbumin, soy protein with a low isoflavone concentration, or soy protein with a high isoflavone concentration). Intravenous glucose tolerance tests were done, and plasma adiponectin and lipoprotein concentrations were determined after 25 months of study. Samples of visceral fat were obtained at 31 months for assessment of adiponectin and PPARgamma expression. The second study involved 8 monkeys in a Latin-square design that compared the effects of diets with casein/lactalbumin, soy protein with a high isoflavone concentration, or soy protein that was alcohol-washed to deplete the isoflavones. After 8 weeks of treatment, insulin sensitivity and plasma lipoproteins were assessed. At 10 weeks, a biopsy of the skeletal muscle was performed for determination of insulin receptor, PPARalpha, and PPARgamma content. The major findings were that consumption of isoflavone-containing soy protein dose-dependently increased insulin responses to the glucose challenge and decreased plasma adiponectin, whereas isoflavone-depleted soy protein decreased body weight and had no effect on plasma adiponectin concentrations. Muscle PPARalpha and gamma expression was also increased with the isoflavone-depleted soy relative to either casein or soy protein containing the isoflavones. Further studies are needed to determine the mechanisms involved in these effects of a high-soy isoflavone diet and to optimize dietary isoflavone content for maximal health benefits in male subjects.


Asunto(s)
Adiponectina/sangre , Resistencia a la Insulina , Isoflavonas/farmacología , Proteínas de Soja/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Isoflavonas/uso terapéutico , Macaca fascicularis , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Proteínas de Soja/uso terapéutico
16.
Metabolism ; 56(6): 830-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17512317

RESUMEN

The purpose of this study was to determine if the insulin resistance we have previously reported in surgically postmenopausal primates treated with combined hormone therapy (HT) is due in part to effects on adipose tissue. Eighty-seven ovariectomized monkeys were fed a moderately atherogenic diet (0.28 mg cholesterol per kilocalorie [0.07 mg/kJ]) and randomized to receive no hormones (control, n = 29), estrogen therapy (ET, conjugated equine estrogens, 0.625 mg/d human equivalent; n = 29), or HT (ET + medroxyprogesterone acetate, 2.5 mg/d human equivalent; n = 29) in the diet for 2 years. Fasting glycemic measures were made at baseline and at the end of treatment. Circulating adiponectin measures, insulin tolerance tests, glucose tolerance tests, and isolated adipocyte glucose uptake assays were performed at the end of the trial. Hormone therapy-treated animals were insulin resistant, as determined by greater fasting insulin concentrations (P = .008), greater homeostasis model assessment of insulin resistance (HOMA-R) value (P = .005) and slower glucose disposal after insulin administration (K(ITT); P = .02) when compared with controls. Subcutaneous adipocytes from HT-treated monkeys had a greater ED(50) for insulin (P = .04) and lower maximal glucose uptake per cell (P < .001) compared with controls, suggesting impaired adipocyte insulin sensitivity. Adipocytes were smaller (P = .001) and adiponectin concentrations were greatest in the ET group (P = .02), with no difference between controls and HT-treated monkeys. In conclusion, estrogen therapy resulted in smaller adipocyte size and greater adiponectin concentrations than control or HT. Hormone therapy resulted in impaired insulin sensitivity and adipocyte glucose uptake compared with controls, whereas there was no difference between ET and controls. Because no adverse effects were found with ET alone, it is likely that the progestin, medroxyprogesterone acetate, resulted in the negative effects of the combined HT regimen on whole-body insulin sensitivity, which were mediated, in part, by reductions in adipose tissue responses to insulin.


Asunto(s)
Tejido Adiposo/metabolismo , Terapia de Reemplazo de Estrógeno , Estrógenos Conjugados (USP)/farmacología , Resistencia a la Insulina , Acetato de Medroxiprogesterona/farmacología , Adipocitos/metabolismo , Animales , Composición Corporal , Peso Corporal , Femenino , Macaca fascicularis
17.
Gastroenterology ; 128(5): 1381-90, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15887119

RESUMEN

BACKGROUND & AIMS: Little is known about the role of mitochondrial beta-oxidation in development of nonalcoholic fatty liver disease (NAFLD). Mitochondrial trifunctional protein (MTP) catalyzes long-chain fatty acid oxidation. Recently, we generated a mouse model for MTP deficiency and reported that homozygous (MTPa-/-) mice suffer neonatal death. In this study, we investigated effects of heterozygosity for the MTP defect on hepatic oxidative stress, insulin resistance, and development of NAFLD in mice. METHODS: We evaluated liver histopathology, serum alanine aminotransferase (ALT), glucose, fatty acids, and insulin levels in MTPa+/- and MTPa+/+ littermates. Insulin resistance was evaluated using glucose tolerance test (GTT) and insulin tolerance test (ITT). Liver tissues were used to measure triglyceride and fatty acid content, activity of superoxide dismutases (SOD) and glutathione peroxidase (GPx), glutathione (GSH), and cytochrome P-450 2E1 expression. RESULTS: Aging but not young MTPa+/- mice developed hepatic steatosis with elevated ALT, basal hyperinsulinemia, and increased insulin area under curve (AUC) on GTT compared with MTPa+/+ littermates. In response to insulin challenge, aging MTPa+/- mice had slower rate of glucose disappearance and increased glucose AUC. Significant hepatic steatosis and insulin resistance developed concomitantly in the MTPa+/- mice at 9-10 months of age. Aging MTPa+/- mice had higher antioxidant activity of total SOD and GPx, lower GSH, and increased expression of cytochrome P-450 2E1, consistent with increased hepatic oxidative stress. CONCLUSIONS: Heterozygosity for beta-oxidation defects predisposes to NAFLD and insulin resistance in aging mice. Impairment of mitochondrial beta-oxidation may play an important role in pathogenesis of NAFLD.


Asunto(s)
Hígado Graso/genética , Hígado Graso/metabolismo , Resistencia a la Insulina/genética , Complejos Multienzimáticos/genética , Animales , Ácidos Grasos/metabolismo , Hígado Graso/patología , Heterocigoto , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Proteína Trifuncional Mitocondrial , Complejos Multienzimáticos/metabolismo , Estrés Oxidativo/fisiología
18.
Steroids ; 69(5): 313-8, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15219409

RESUMEN

We have previously shown that hormone therapy (HT) with medroxyprogesterone acetate (MPA) alone or in combination with conjugated equine estrogens (CEE) impairs insulin sensitivity. In the current study, we sought to determine if the effect of MPA on whole body insulin sensitivity is associated with alterations in insulin signaling proteins in skeletal muscle. Ovariectomized cynomolgus monkeys were treated for 2 years with either no hormones (n = 10), CEE (0.625 mg/day human equivalent, n = 11) or CEE + MPA (2.5 mg/day human equivalent, n = 12). At the end of the study, biopsies of rectus femoris muscle were flash frozen in the basal and insulin-stimulated (10 min post-intravenous insulin injection) state. Immunoblotting revealed that CEE + MPA monkeys had significantly less glucose transporter 4 (GLUT4) expression (ANOVA P = 0.001), but there was no significant treatment effect on expression of insulin receptor, insulin receptor substrate (IRS)-1, IRS-2, or the p85 subunit of phosphatidylinositol 3-kinase (PI 3-K). There was a tendency for decreased insulin receptor tyrosine phosphorylation with CEE + MPA treatment (ANOVA P = 0.14). These deficiencies in skeletal muscle insulin signaling likely contribute to the unfavorable changes in whole body insulin sensitivity associated with CEE + MPA treatment.


Asunto(s)
Terapia de Reemplazo de Hormonas , Insulina/metabolismo , Acetato de Medroxiprogesterona/farmacología , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos Hormonales/farmacología , Estrógenos Conjugados (USP)/farmacología , Femenino , Transportador de Glucosa de Tipo 4 , Humanos , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Macaca fascicularis , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiología , Ovariectomía , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Distribución Aleatoria , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología
19.
Arterioscler Thromb Vasc Biol ; 23(12): 2241-6, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14576074

RESUMEN

OBJECTIVE: We sought to determine if arterial LDL metabolism contributes to the decreased atherosclerosis seen with soy and if isolated isoflavones would have similar effects. METHODS AND RESULTS: Ovariectomized monkeys were fed an atherogenic diet for 20 weeks with a protein source of (1) casein/lactalbumin (CAS, n=20), (2) soy protein isolate (SOY, n=20), or (3) casein/lactalbumin with isolated soy isoflavones (ISO, n=17). Plasma lipoprotein concentrations were improved with SOY but not ISO. Arterial LDL metabolism was characterized with one subset (n=12/group) injected with dual-labeled tyramine-cellobiose (TC)-LDL (125I-TC-131I-LDL) 24 hours before necropsy to determine LDL degradation and accumulation, while another subset (n=8/group) was injected with 125I-TC-LDL 1 hour before necropsy to determine LDL permeability and delivery. CONCLUSIONS: Coronary artery LDL degradation was reduced by 50% (P=0.02) with SOY but not with ISO compared with CAS. Neither treatment altered arterial permeability. Reduced LDL degradation with SOY was due to decreased arterial LDL delivery (P=0.02). Carotid artery cholesterol ester was also decreased with SOY, but not with ISO. Plasma isoprostanes or plasma markers of inflammation did not differ among treatment groups. Thus, the decreased arterial LDL delivery and subsequent LDL degradation may explain, in part, the atheroprotective effects of soy.


Asunto(s)
Arteriosclerosis/prevención & control , Suplementos Dietéticos , Isoflavonas/administración & dosificación , Isoflavonas/uso terapéutico , Lipoproteínas LDL/sangre , Lipoproteínas LDL/metabolismo , Proteínas de Soja/uso terapéutico , Animales , Arteriosclerosis/dietoterapia , Biomarcadores/sangre , Colesterol/sangre , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Isoflavonas/metabolismo , Isoprostanos/sangre , Lípidos/sangre , Lipoproteínas/sangre , Macaca fascicularis
20.
Metabolism ; 52(9): 1085-91, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14506611

RESUMEN

The effects of tibolone on body weight, body composition, and fasting carbohydrate measures in surgically postmenopausal cynomolgus monkeys were compared to those of conjugated equine estrogens (CEE) with and without medroxyprogesterone acetate (MPA). Monkeys were fed a moderately atherogenic diet with either no hormones (control n = 29), CEE (0.042 mg/kg, n = 27), CEE + MPA (0.167 mg/kg, n = 29), low-dose tibolone (LoTib, 0.05 mg/kg, n = 30), or high-dose tibolone (HiTib, 0.20 mg/kg, n = 31) daily for 2 years. Body weight (BW) was measured throughout the study, and dual-energy x-ray absorptiometry (DEXA) scans of the abdominal region (lumbar vertebrae 1 through 5) were performed at the end of the trial to assess abdominal body composition. Fasting carbohydrate measures (glucose, insulin, C-peptide, and fructosamine) were determined at baseline and after 2 years of treatment. Compared to controls, BW significantly increased and abdominal soft tissue mass was greater (analysis of variance [ANOVA], P <.001, P = 0.003, respectively) in all but the CEE-treated group (P =.78, P =.94, respectively). HiTib-treated monkeys had greater abdominal lean mass compared to controls (P =.008), while there was no significant treatment effect on abdominal fat mass (analysis of covariance [ANCOVA], P =.29). Fasting insulin concentrations and fasting insulin/glucose ratios were greater in CEE + MPA- (P =.002, P =.03, respectively) and HiTib-treated monkeys (P =.03, P =.02, respectively) compared to controls. There was a strong trend for a treatment effect on fasting blood glucose concentration (ANCOVA, P =.06) with CEE + MPA-treated animals having the greatest values, despite no difference in fructosamine concentration (ANCOVA, P =.57). Using these fasting measures, the homeostasis model assessment (HOMA-IR) revealed significant insulin resistance with CEE + MPA treatment compared to controls (P =.008), while the quantitative insulin sensitivity check index (QUICKI) showed significantly impaired insulin sensitivity in all hormone replacement therapy (HRT) groups (all P values <.03), except CEE (P =.12). In conclusion, HRT with CEE + MPA or tibolone results in greater BW, abdominal soft tissue, and insulin resistance (CEE + MPA and HiTib) compared to control-treated monkeys.


Asunto(s)
Composición Corporal/efectos de los fármacos , Carbohidratos/sangre , Estrógenos Conjugados (USP)/farmacología , Acetato de Medroxiprogesterona/farmacología , Norpregnenos/farmacología , Posmenopausia , Animales , Arteriosclerosis/etiología , Arteriosclerosis/prevención & control , Glucemia/análisis , Peso Corporal , Péptido C/sangre , Grasas de la Dieta/administración & dosificación , Ingestión de Energía , Terapia de Reemplazo de Estrógeno , Ayuno , Femenino , Fructosamina/sangre , Insulina/sangre , Resistencia a la Insulina , Macaca fascicularis , Ovariectomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...