Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 435
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(14): 8166-8176, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32188779

RESUMEN

Multiple insulin-regulated enzymes participate in hepatic glycogen synthesis, and the rate-controlling step responsible for insulin stimulation of glycogen synthesis is unknown. We demonstrate that glucokinase (GCK)-mediated glucose phosphorylation is the rate-controlling step in insulin-stimulated hepatic glycogen synthesis in vivo, by use of the somatostatin pancreatic clamp technique using [13C6]glucose with metabolic control analysis (MCA) in three rat models: 1) regular chow (RC)-fed male rats (control), 2) high fat diet (HFD)-fed rats, and 3) RC-fed rats with portal vein glucose delivery at a glucose infusion rate matched to the control. During hyperinsulinemia, hyperglycemia dose-dependently increased hepatic glycogen synthesis. At similar levels of hyperinsulinemia and hyperglycemia, HFD-fed rats exhibited a decrease and portal delivery rats exhibited an increase in hepatic glycogen synthesis via the direct pathway compared with controls. However, the strong correlation between liver glucose-6-phosphate concentration and net hepatic glycogen synthetic rate was nearly identical in these three groups, suggesting that the main difference between models is the activation of GCK. MCA yielded a high control coefficient for GCK in all three groups. We confirmed these findings in studies of hepatic GCK knockdown using an antisense oligonucleotide. Reduced liver glycogen synthesis in lipid-induced hepatic insulin resistance and increased glycogen synthesis during portal glucose infusion were explained by concordant changes in translocation of GCK. Taken together, these data indicate that the rate of insulin-stimulated hepatic glycogen synthesis is controlled chiefly through GCK translocation.


Asunto(s)
Hígado Graso/patología , Glucoquinasa/metabolismo , Glucosa/metabolismo , Glucógeno Hepático/biosíntesis , Hígado/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/etiología , Técnicas de Silenciamiento del Gen , Glucoquinasa/genética , Glucosa/administración & dosificación , Glucosa-6-Fosfato/análisis , Glucosa-6-Fosfato/metabolismo , Humanos , Hiperglucemia/etiología , Hiperglucemia/patología , Hiperinsulinismo/etiología , Hiperinsulinismo/patología , Insulina/metabolismo , Resistencia a la Insulina , Hígado/patología , Masculino , Metabolómica , Fosforilación , Ratas
2.
Carcinogenesis ; 43(5): 504-516, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-35104315

RESUMEN

Metabolic syndrome (MetS) and its four clinical entities, central obesity, insulin resistance, hypertension and dyslipidemia, are implicated in increasing the risk and mortality of cancer in several organs. However, it is unclear how they are associated with increased risk of prostate cancer. To elucidate the mechanistic link between MetS and prostate carcinogenesis, we characterized the development of MetS and prostate adenocarcinoma in prostate-specific Pten-/- (Ptenp-/-) mice fed a high-fat (HF) diet. We found that male Ptenp-/- mice on an HF diet gained excess body weight and elevated blood glucose, insulin and insulin-like growth factor 1 (IGF1) levels at 20 weeks of age and were obese at 40 weeks. Prostate adenocarcinoma multiplicity at 40 weeks was significantly higher in the mice on an HF diet, suggesting that the HF diet promotes the development of prostate adenocarcinoma. Increased cell proliferation and enhanced AKT activation were found in the prostates of mice on an HF diet. Further transcriptome study revealed that receptor tyrosine kinase regulation, which mediates insulin/IGF1 signaling, was one of the top enriched pathways by HF diet-induced transcriptome changes. Together, our results suggest that HF diet-induced hyperinsulinemia leads to increased activation of insulin/IGF1/AKT signaling in lesioned prostates, promoting the development of adenocarcinoma.


Asunto(s)
Adenocarcinoma , Hiperinsulinismo , Resistencia a la Insulina , Neoplasias de la Próstata , Adenocarcinoma/genética , Animales , Dieta Alta en Grasa/efectos adversos , Humanos , Hiperinsulinismo/complicaciones , Hiperinsulinismo/patología , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Fosfohidrolasa PTEN/genética , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
3.
FASEB J ; 35(7): e21687, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34089273

RESUMEN

Apart from its role in inflammation and immunity, chemerin is also involved in white adipocyte biology. To study the role of chemerin in adipocyte metabolism, we examined the function of chemerin in brown adipose tissue. Brown and white adipocyte precursors were differentiated into adipocytes in the presence of Chemerin siRNA. Chemerin-deficient (Chem-/- ) mice were compared to wild-type mice when fed a high-fat diet. Chemerin is expressed during brown adipocyte differentiation and knock down of chemerin mRNA results in decreased brown adipocyte differentiation with reduced fatty acid uptake in brown adipocytes. Chem-/- mice are leaner than wild-type mice but gain more weight when challenged with high-fat diet feeding, resulting in a larger increase in fat deposition. Chem-/- mice develop insulin resistance when on a high-fat diet or due to age. Brown adipose depots in Chem-/- mice weigh more than in wild-type mice, but with decreased mitochondrial content and function. Compared to wild-type mice, male Chem-/- mice have decreased oxygen consumption, CO2 production, energy expenditure, and a lower respiratory exchange ratio. Additionally, body temperature of Chem-/- mice is lower than that of wild-type mice. These results revealed that chemerin is expressed during brown adipocyte differentiation and has a pivotal role in energy metabolism through brown adipose tissue thermogenesis.


Asunto(s)
Tejido Adiposo Pardo/patología , Envejecimiento/patología , Quimiocinas/fisiología , Dieta Alta en Grasa , Metabolismo Energético , Hiperinsulinismo/patología , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intercelular/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Femenino , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno , Termogénesis
4.
J Biol Chem ; 295(48): 16359-16369, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-32943552

RESUMEN

The incidence of diabetes, obesity, and metabolic diseases has reached an epidemic status worldwide. Insulin resistance is a common link in the development of these conditions, and hyperinsulinemia is a central hallmark of peripheral insulin resistance. However, how hyperinsulinemia leads to systemic insulin resistance is less clear. We now provide evidence that hyperinsulinemia promotes the release of soluble pro-inflammatory mediators from macrophages that lead to systemic insulin resistance. Our observations suggest that hyperinsulinemia induces sirtuin1 (SIRT1) repression and stimulates NF-κB p65 nuclear translocation and transactivation of NF-κB to promote the extracellular release of pro-inflammatory mediators. We further showed that low-dose naltrexone (LDN) abrogates hyperinsulinemia-mediated SIRT1 repression and prevents NF-κB p65 nuclear translocation. This, in turn, attenuates the hyperinsulinemia-induced release of pro-inflammatory cytokines and reinstates insulin sensitivity both in in vitro and in vivo diet-induced hyperinsulinemic mouse model. Notably, our data indicate that Sirt1 knockdown or inhibition blunts the anti-inflammatory properties of LDN in vitro Using numerous complementary in silico and in vitro experimental approaches, we demonstrated that LDN can bind to SIRT1 and increase its deacetylase activity. Together, these data support a critical role of SIRT1 in inflammation and insulin resistance in hyperinsulinemia. LDN improves hyperinsulinemia-induced insulin resistance by reorienting macrophages toward anti-inflammation. Thus, LDN treatment may provide a novel therapeutic approach against hyperinsulinemia-associated insulin resistance.


Asunto(s)
Hiperinsulinismo/tratamiento farmacológico , Resistencia a la Insulina , Naltrexona/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Células RAW 264.7 , Sirtuina 1/genética , Sirtuina 1/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1105-L1118, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34668415

RESUMEN

Increased insulin is associated with obesity-related airway hyperreactivity and asthma. We tested whether the use of metformin, an antidiabetic drug used to reduce insulin resistance, can reduce circulating insulin, thereby preventing airway hyperreactivity in rats with dietary obesity. Male and female rats were fed a high- or low-fat diet for 5 wk. Some male rats were simultaneously treated with metformin (100 mg/kg orally). In separate experiments, after 5 wk of a high-fat diet, some rats were switched to a low-fat diet, whereas others continued a high-fat diet for an additional 5 wk. Bronchoconstriction and bradycardia in response to bilateral electrical vagus nerve stimulation or to inhaled methacholine were measured in anesthetized and vagotomized rats. Body weight, body fat, caloric intake, fasting glucose, and insulin were measured. Vagally induced bronchoconstriction was potentiated only in male rats on a high-fat diet. Males gained more body weight, body fat, and had increased levels of fasting insulin compared with females. Metformin prevented development of vagally induced airway hyperreactivity in male rats on high-fat diet, in addition to inhibiting weight gain, fat gain, and increased insulin. In contrast, switching rats to a low-fat diet for 5 wk reduced body weight and body fat, but it did not reverse fasting glucose, fasting insulin, or potentiation of vagally induced airway hyperreactivity. These data suggest that medications that target insulin may be effective treatment for obesity-related asthma.


Asunto(s)
Asma/tratamiento farmacológico , Hiperreactividad Bronquial/tratamiento farmacológico , Broncoconstricción , Dieta Alta en Grasa/efectos adversos , Hiperinsulinismo/prevención & control , Metformina/farmacología , Obesidad/complicaciones , Animales , Asma/inducido químicamente , Asma/metabolismo , Asma/patología , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Broncoconstrictores/toxicidad , Femenino , Glucosa/metabolismo , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Hipoglucemiantes/farmacología , Masculino , Cloruro de Metacolina/toxicidad , Ratas , Ratas Sprague-Dawley , Nervio Vago/efectos de los fármacos , Aumento de Peso
6.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L236-L247, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34009030

RESUMEN

Obesity-related asthma often presents with more severe symptoms than non-obesity-related asthma and responds poorly to current treatments. Both insulin resistance and hyperinsulinemia are common in obesity. We have shown that increased insulin mediates airway hyperreactivity in diet-induced obese rats by causing neuronal M2 muscarinic receptor dysfunction, which normally inhibits acetylcholine release from parasympathetic nerves. Decreasing insulin with streptozotocin prevented airway hyperreactivity and M2 receptor dysfunction. The objective of the present study was to investigate whether pioglitazone, a hypoglycemic drug, prevents airway hyperreactivity and M2 receptor dysfunction in obese rats. Male rats fed a low- or high-fat diet were treated with pioglitazone or PBS by daily gavage. Body weight, body fat, fasting insulin, and bronchoconstriction and bradycardia in response to electrical stimulation of vagus nerves and to aerosolized methacholine were recorded. Pilocarpine, a muscarinic receptor agonist, was used to measure M2 receptor function. Rats on a high-fat diet had potentiated airway responsiveness to vagal stimulation and dysfunctional neuronal M2 receptors, whereas airway responsiveness to methacholine was unaffected. Pioglitazone reduced fasting insulin and prevented airway hyperresponsiveness and M2 receptor dysfunction but did not change inflammatory cytokine mRNA expression in alveolar macrophages. High-fat diet, with and without pioglitazone, had tissue-specific effects on insulin receptor mRNA expression. In conclusion, pioglitazone prevents vagally mediated airway hyperreactivity and protects neuronal M2 muscarinic receptor function in obese rats.


Asunto(s)
Hiperreactividad Bronquial/tratamiento farmacológico , Hiperinsulinismo/tratamiento farmacológico , Insulina/metabolismo , Neuronas/efectos de los fármacos , Obesidad/complicaciones , Pioglitazona/farmacología , Receptor Muscarínico M2/metabolismo , Animales , Hiperreactividad Bronquial/etiología , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Dieta Alta en Grasa/efectos adversos , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Hipoglucemiantes/farmacología , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M2/genética
7.
Hum Genomics ; 14(1): 9, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32143698

RESUMEN

BACKGROUND: Gain-of-function mutations in the GLUD1 gene, encoding for glutamate dehydrogenase (GDH), result in the hyperinsulinism/hyperammonemia HI/HA syndrome. HI/HA patients present with harmful hypoglycemia secondary to protein-induced HI and elevated plasma ammonia levels. These symptoms may be accompanied by seizures and mental retardation. GDH is a mitochondrial enzyme that catalyzes the oxidative deamination of glutamate to α-ketoglutarate, under allosteric regulations mediated by its inhibitor GTP and its activator ADP. The present study investigated the functional properties of the GDH-G446V variant (alias c.1496G > T, p.(Gly499Val) (NM_005271.4)) in patient-derived lymphoblastoid cells. RESULTS: The calculated energy barrier between the opened and closed state of the enzyme was 41% lower in GDH-G446V compared to wild-type GDH, pointing to altered allosteric regulation. Computational analysis indicated conformational changes of GDH-G446V in the antenna region that is crucial for allosteric regulators. Enzymatic activity measured in patient-derived lymphoblastoid cells showed impaired allosteric responses of GDH-G446V to both regulators GTP and ADP. In particular, as opposed to control lymphoblastoid cells, GDH-G446V cells were not responsive to GTP in the lower range of ADP concentrations. Assessment of the metabolic rate revealed higher mitochondrial respiration in response to GDH-dependent substrates in the GDH-G446V lymphoblastoid cells compared to control cells. This indicates a shift toward glutaminolysis for energy provision in cells carrying the GDH-G446V variant. CONCLUSIONS: Substitution of the small amino acid glycine for the hydrophobic branched-chain valine altered the allosteric sensitivity to both inhibitory action of GTP and activation by ADP, rendering cells metabolically responsive to glutamine.


Asunto(s)
Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Guanosina Trifosfato/metabolismo , Hiperinsulinismo/patología , Linfocitos/patología , Mutación , Adulto , Regulación Alostérica , Estudios de Casos y Controles , Femenino , Glutamato Deshidrogenasa/química , Humanos , Hiperinsulinismo/genética , Recién Nacido , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Conformación Proteica
8.
Am J Med Genet A ; 185(4): 1251-1255, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33442921

RESUMEN

Rubinstein-Taybi syndrome (RSTS) is an autosomal dominant genetic syndrome characterized by distinct facial features, broad thumbs, growth restriction, microcephaly, intellectual disability, and developmental delay. Pathogenic variants in both CREBBP and EP300 have been associated with RSTS. Here we present a case of a female with hyperinsulinism and features consistent with RSTS, found to have a pathogenic variant in EP300. While there have been a few rare case reports of hyperinsulinism in RSTS, we suggest that hyperinsulinism might be a more prominent feature in EP300 variant RSTS than previously recognized.


Asunto(s)
Proteína p300 Asociada a E1A/genética , Predisposición Genética a la Enfermedad , Hiperinsulinismo/genética , Síndrome de Rubinstein-Taybi/genética , Femenino , Variación Genética/genética , Genotipo , Humanos , Hiperinsulinismo/patología , Lactante , Recién Nacido , Mutación/genética , Fenotipo , Síndrome de Rubinstein-Taybi/patología , Eliminación de Secuencia/genética
9.
Proc Natl Acad Sci U S A ; 115(5): 1027-1032, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339498

RESUMEN

The ß-cell-enriched MAFA transcription factor plays a central role in regulating glucose-stimulated insulin secretion while also demonstrating oncogenic transformation potential in vitro. No disease-causing MAFA variants have been previously described. We investigated a large pedigree with autosomal dominant inheritance of diabetes mellitus or insulinomatosis, an adult-onset condition of recurrent hyperinsulinemic hypoglycemia caused by multiple insulin-secreting neuroendocrine tumors of the pancreas. Using exome sequencing, we identified a missense MAFA mutation (p.Ser64Phe, c.191C>T) segregating with both phenotypes of insulinomatosis and diabetes. This mutation was also found in a second unrelated family with the same clinical phenotype, while no germline or somatic MAFA mutations were identified in nine patients with sporadic insulinomatosis. In the two families, insulinomatosis presented more frequently in females (eight females/two males) and diabetes more often in males (12 males/four females). Four patients from the index family, including two homozygotes, had a history of congenital cataract and/or glaucoma. The p.Ser64Phe mutation was found to impair phosphorylation within the transactivation domain of MAFA and profoundly increased MAFA protein stability under both high and low glucose concentrations in ß-cell lines. In addition, the transactivation potential of p.Ser64Phe MAFA in ß-cell lines was enhanced compared with wild-type MAFA. In summary, the p.Ser64Phe missense MAFA mutation leads to familial insulinomatosis or diabetes by impacting MAFA protein stability and transactivation ability. The human phenotypes associated with the p.Ser64Phe MAFA missense mutation reflect both the oncogenic capacity of MAFA and its key role in islet ß-cell activity.


Asunto(s)
Diabetes Mellitus/genética , Hiperinsulinismo/genética , Insulinoma/genética , Factores de Transcripción Maf de Gran Tamaño/genética , Proteínas Mutantes/genética , Mutación Missense , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Femenino , Genes Dominantes , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulinoma/metabolismo , Insulinoma/patología , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Proteínas Mutantes/metabolismo , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Linaje , Estabilidad Proteica , Activación Transcripcional , Secuenciación del Exoma
10.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203572

RESUMEN

Type 2 diabetes mellitus (T2D) is one of the prominent risk factors for the development and progression of calcific aortic valve disease. Nevertheless, little is known about molecular mechanisms of how T2D affects aortic valve (AV) remodeling. In this study, the influence of hyperinsulinemia and hyperglycemia on degenerative processes in valvular tissue is analyzed in intact AV exposed to an either static or dynamic 3D environment, respectively. The complex native dynamic environment of AV is simulated using a software-governed bioreactor system with controlled pulsatile flow. Dynamic cultivation resulted in significantly stronger fibrosis in AV tissue compared to static cultivation, while hyperinsulinemia and hyperglycemia had no impact on fibrosis. The expression of key differentiation markers and proteoglycans were altered by diabetic conditions in an environment-dependent manner. Furthermore, hyperinsulinemia and hyperglycemia affect insulin-signaling pathways. Western blot analysis showed increased phosphorylation level of protein kinase B (AKT) after acute insulin stimulation, which was lost in AV under hyperinsulinemia, indicating acquired insulin resistance of the AV tissue in response to elevated insulin levels. These data underline a complex interplay of diabetic conditions on one hand and biomechanical 3D environment on the other hand that possesses an impact on AV tissue remodeling.


Asunto(s)
Enfermedad de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Diabetes Mellitus/patología , Hiperglucemia/patología , Hiperinsulinismo/patología , Insulina/metabolismo , Animales , Enfermedad de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/genética , Diabetes Mellitus/metabolismo , Humanos , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo
11.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34360753

RESUMEN

Activation of innate immunity and low-grade inflammation contributes to hyperglycemia and an onset of Type 2 Diabetes Mellitus (T2DM). Interleukin-2 (IL-2), leptin, High Mobility Group Box-1 (HMGB-1), and increased glucose concentrations are mediators of these processes also by modulating peripheral blood mononuclear cells (PBMCs) response. The aim of this study was to investigate if HMGB-1 and IL-2 turn on PBMCs and their leptin secretion. In isolated human PBMCs and their subpopulations from healthy individuals and naïve T2DM patients, leptin release, pro-inflammatory response and Toll-like Receptors (TLRs) activation was measured. After treatment with IL-2 and HMGB1, NK (Natural Killer) have the highest amount of leptin secretion, whilst NK-T have the maximal release in basal conditions. TLR4 (TAK242) and/or TLR2 (TLR2-IgA) inhibitors decreased leptin secretion after IL-2 and HMGB1 treatment. A further non-significant increase in leptin secretion was reported in PBMCs of naive T2DM patients in response to IL-2 and HMGB-1 stimulation. Finally, hyperglycemia or hyperinsulinemia might stimulate leptin secretion from PBMCs. The amount of leptin released from PBMCs after the different treatments was enough to stimulate the secretion of IL-1ß from monocytes. Targeting leptin sera levels and secretion from PBMCs could represent a new therapeutic strategy to counteract metabolic diseases such as T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Proteína HMGB1/farmacología , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Interleucina-2/farmacología , Leptina/metabolismo , Leucocitos Mononucleares/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Hiperglucemia/patología , Hiperinsulinismo/patología , Leucocitos Mononucleares/patología
12.
Proc Natl Acad Sci U S A ; 114(45): 11926-11931, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29078297

RESUMEN

Insulin resistance, a key etiological factor in metabolic syndrome, is closely linked to ectopic lipid accumulation and increased intracellular Ca2+ concentrations in muscle and liver. However, the mechanism by which dysregulated intracellular Ca2+ homeostasis causes insulin resistance remains elusive. Here, we show that increased intracellular Ca2+ acts as a negative regulator of insulin signaling. Chronic intracellular Ca2+ overload in hepatocytes during obesity and hyperlipidemia attenuates the phosphorylation of protein kinase B (Akt) and its key downstream signaling molecules by inhibiting membrane localization of pleckstrin homology (PH) domains. Pharmacological approaches showed that elevated intracellular Ca2+ inhibits insulin-stimulated Akt phosphorylation and abrogates membrane localization of various PH domain proteins such as phospholipase Cδ and insulin receptor substrate 1, suggesting a common mechanism inhibiting the membrane targeting of PH domains. PH domain-lipid overlay assays confirmed that Ca2+ abolishes the binding of various PH domains to phosphoinositides (PIPs) with two adjacent phosphate groups, such as PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3 Finally, thermodynamic analysis of the binding interaction showed that Ca2+-mediated inhibition of targeting PH domains to the membrane resulted from the tight binding of Ca2+ rather than PH domains to PIPs forming Ca2+-PIPs. Thus, Ca2+-PIPs prevent the recognition of PIPs by PH domains, potentially due to electrostatic repulsion between positively charged side chains in PH domains and the Ca2+-PIPs. Our findings provide a mechanistic link between intracellular Ca2+ dysregulation and Akt inactivation in insulin resistance.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Resistencia a la Insulina/fisiología , Fosfatidilinositoles/metabolismo , Dominios Homólogos a Pleckstrina/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Dieta Alta en Grasa , Intolerancia a la Glucosa/patología , Hiperinsulinismo/patología , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Fosfolipasa C delta/metabolismo , Fosforilación , Unión Proteica
13.
Molecules ; 25(16)2020 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-32796637

RESUMEN

d-allulose is an uncommon sugar that provides almost no calories when consumed. Its sweetness is 70% that of sucrose. d-allulose is a metabolic regulator of glucose and lipid metabolism. However, few reports concerning its effect on diabetes and related metabolic disturbances in db/db mice are available. In this study, we evaluated d-allulose's effect on hyperglycemia, hyperinsulinemia, diabetes and inflammatory responses in C57BL/KsJ-db/db mice. Mice were divided into normal diet, erythritol supplemented (5% w/w), and d-allulose supplemented (5% w/w) groups. Blood glucose and plasma glucagon levels and homeostatic model assessment (HOMA-IR) were significantly lower in the d-allulose group than in the normal diet group, and plasma insulin level was significantly increased. Further, d-allulose supplement significantly increased hepatic glucokinase activity and decreased hepatic phosphoenolpyruvate carboxykinase and glucose-6-phosphatase activity. Expression of glucose transporter 4, insulin receptor substrate 1, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha and AKT serine/threonine kinase 2 were also upregulated by d-allulose supplement in adipocyte and muscle. Finally, d-allulose effectively lowered plasma and hepatic triglyceride and free fatty acid levels, and simultaneously reduced hepatic fatty acid oxidation and carnitine palmitoyl transferase activity. These changes are likely attributable to suppression of hepatic fatty acid synthase and glucose-6-phosphate dehydrogenase activity. Notably, d-allulose also reduced pro-inflammatory adipokine and cytokine levels in plasma. Our results indicate that d-allulose is an effective sugar substitute for improving lipid and glucose metabolism.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Fructosa/farmacología , Hiperglucemia/tratamiento farmacológico , Hiperinsulinismo/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Inflamación/metabolismo , Inflamación/patología , Insulina/sangre , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos
14.
Biochem J ; 475(3): 561-569, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29170160

RESUMEN

The purpose of the present study was to determine the effects of prolonged hyperinsulinemia on mitochondrial respiration and uncoupling in distinct adipose tissue depots. Sixteen-week-old male mice were injected daily with placebo or insulin to induce an artificial hyperinsulinemia for 28 days. Following the treatment period, mitochondrial respiration and degree of uncoupling were determined in permeabilized perirenal, inguinal, and interscapular adipose tissue. White adipose tissue (WAT) mitochondria (inguinal and perirenal) respire at substantially lower rates compared with brown adipose tissue (BAT). Insulin treatment resulted in a significant reduction in mitochondrial respiration in inguinal WAT (iWAT) and interscapular BAT (iBAT), but not in perirenal WAT (pWAT). Furthermore, these changes were accompanied by an insulin-induced reduction in UCP-1 (uncoupling protein 1) and PGC-1α in iWAT and iBAT only, but not in pWAT or skeletal muscle. Compared with adipose tissue mitochondria in placebo conditions, adipose tissue from hyperinsulinemic mice manifested a site-specific reduction in mitochondrial respiration probably as a result of reduced uncoupling. These results may help explain weight gain so commonly seen with insulin treatment in type 2 diabetes mellitus.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/farmacología , Mitocondrias/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulina/metabolismo , Ratones , Mitocondrias/patología , Proteínas Desacopladoras Mitocondriales/genética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Respiración/efectos de los fármacos , Proteína Desacopladora 1/genética
15.
Mediators Inflamm ; 2019: 1491083, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30983877

RESUMEN

AIM: The development of type 2 diabetes (T2DM) is associated with disturbances of immune status that may be reflected by alterations of the profile of circulating immune cells. In order to study whether there exists genetic predisposition to these alterations, we investigated the relative content of circulating monocyte and lymphocyte subpopulations at fasting condition and upon stimulation by short-term hyperinsulinemia in nondiabetic first-degree relatives (FDR) of T2DM patients and in control subjects. MATERIALS AND METHODS: 19 nondiabetic (FDR) and 19 control subjects without a family history of diabetes (all men) matched for age and BMI underwent 2-hour hyperinsulinemic-euglycemic clamp. Blood samples taken before and at the end of the clamp were used for the flow cytometry analysis of lymphocyte and monocyte populations and for the assessment of cytokine levels. RESULTS: At fasting conditions, FDR showed a higher CD4/CD8 ratio of peripheral lymphocytes, a higher percentage of Th17 lymphocytes, and a lower content of intermediate monocytes when compared to controls. The CD4/CD8 ratio correlated with fat mass, insulin, and HOMA-IR in the entire group of subjects. Hyperinsulinemia decreased a relative content of peripheral CD4+ and increased a relative content of CD8+ T lymphocytes, thus decreasing the CD4/CD8 ratio by 18-22% in both groups of subjects. In FDR but not in controls, the decrease of CD4+ T lymphocyte content was partially based on the decrease of TH2 and TH17 lymphocyte subpopulations. In control subjects but not in FDR, the number of intermediate monocytes has declined in response to hyperinsulinemia. CONCLUSION: The alterations of the CD4/CD8 lymphocyte ratio, relative content of TH17 cells, and intermediate monocytes in FDR are features of genetic predisposition to T2DM and may play a role in pathogenesis of T2DM. Short-term hyperinsulinemia affected mostly the immune cell populations deregulated in FDR subjects, which suggests important interplay between immune system homeostasis and insulin levels.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Ayuno/sangre , Hiperinsulinismo/sangre , Subgrupos Linfocitarios/metabolismo , Monocitos/metabolismo , Adulto , Glucemia/metabolismo , Índice de Masa Corporal , Relación CD4-CD8 , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Resistencia a la Insulina/fisiología , Masculino , Células Th17/metabolismo , Células Th2/metabolismo
16.
J Physiol ; 596(19): 4597-4609, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30089335

RESUMEN

KEY POINTS: A ketogenic diet is known to lead to weight loss and is considered metabolically healthy; however there are conflicting reports on its effect on hepatic insulin sensitivity. KD fed animals appear metabolically healthy in the fasted state after 3 days of dietary challenge, whereas obesogenic high-fat diet (HFD) fed animals show elevated insulin levels. A glucose challenge reveals that both KD and HFD fed animals are glucose intolerant. Glucose intolerance correlates with increased lipid oxidation and lower respiratory exchange ratio (RER); however, all animals respond to glucose injection with an increase in RER. Hyperinsulinaemic-euglycaemic clamps with double tracer show that the effect of KD is a result of hepatic insulin resistance and increased glucose output but not impaired glucose clearance or tissue glucose uptake in other tissues. ABSTRACT: Despite being a relevant healthcare issue and heavily investigated, the aetiology of type 2 diabetes (T2D) is still incompletely understood. It is well established that increased endogenous glucose production (EGP) leads to a progressive increase in glucose levels, causing insulin resistance and eventual loss of glucose homeostasis. The consumption of high carbohydrate, high-fat, western style diet (HFD) is linked to the development of T2D and obesity, whereas the consumption of a low carbohydrate, high-fat, ketogenic diet (KD) is considered healthy. However, several days of carbohydrate restriction are known to cause selective hepatic insulin resistance. In the present study, we compare the effects of short-term HFD and KD feeding on glucose homeostasis in mice. We show that, even though KD fed animals appear to be healthy in the fasted state, they exhibit decreased glucose tolerance to a greater extent than HFD fed animals. Furthermore, we show that this effect originates from blunted suppression of hepatic glucose production by insulin, rather than impaired glucose clearance and tissue glucose uptake. These data suggest that the early effects of HFD consumption on EGP may be part of a normal physiological response to increased lipid intake and oxidation, and that systemic insulin resistance results from the addition of dietary glucose to EGP-derived glucose.


Asunto(s)
Diabetes Mellitus/fisiopatología , Dieta Alta en Grasa/efectos adversos , Dieta Cetogénica/efectos adversos , Intolerancia a la Glucosa/etiología , Resistencia a la Insulina , Hígado/patología , Obesidad/etiología , Animales , Glucemia/metabolismo , Intolerancia a la Glucosa/patología , Hiperinsulinismo/etiología , Hiperinsulinismo/patología , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Inanición
17.
J Biol Chem ; 292(30): 12449-12459, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28487366

RESUMEN

The onset of type 2 diabetes is characterized by transition from successful to failed insulin secretory compensation to obesity-related insulin resistance and dysmetabolism. Energy-rich diets in rodents are commonly studied models of compensatory increases in both insulin secretion and ß cell mass. However, the mechanisms of these adaptive responses are incompletely understood, and it is also unclear why these responses eventually fail. We measured the temporal trends of glucose homeostasis, insulin secretion, ß cell morphometry, and islet gene expression in C57BL/6NTac mice fed a 60% high-fat diet (HFD) or control diet for up to 16 weeks. A 2-fold increased hyperinsulinemia was maintained for the first 4 weeks of HFD feeding and then further increased through 16 weeks. ß cell mass increased progressively starting at 4 weeks, principally through nonproliferative growth. Insulin sensitivity was not significantly perturbed until 11 weeks of HFD feeding. Over the first 8 weeks, we observed two distinct waves of increased expression of ß cell functional and prodifferentiation genes. This was followed by activation of the unfolded protein response at 8 weeks and overt ß cell endoplasmic reticulum stress at 12-16 weeks. In summary, ß cell adaptation to an HFD in C57BL/6NTac mice entails early insulin hypersecretion and a robust growth phase along with hyperexpression of related genes that begin well before the onset of observed insulin resistance. However, continued HFD exposure results in cessation of gene hyperexpression, ß cell functional failure, and endoplasmic reticulum stress. These data point to a complex but not sustainable integration of ß cell-adaptive responses to nutrient overabundance, obesity development, and insulin resistance.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hiperinsulinismo/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Estrés del Retículo Endoplásmico , Hiperinsulinismo/patología , Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Factores de Tiempo
18.
Biochem Biophys Res Commun ; 503(1): 71-78, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-29852165

RESUMEN

Understanding the function and molecular relevance of distinct miRNAs in endothelial cells (ECs) paves avenues for possible therapeutic intervention by targeting epigenetic mechanisms in vascular endothelial dysfunction, one of the major complications of type 2 diabetes mellitus (T2DM). MiR-342-3p, an obesity-associated miRNA, has recently been shown to be significantly upregulated in human angiosarcoma compared to benign hemangioma, indicating its potential involvement as a proangiogenic factor. Herein, we show that endothelial miR-342-3p expression was significantly compromised in T2DM organisms and this inhibition powerfully blocked vasculogenesis in vivo by repressing endothelial proliferation and migration. From a mechanistic standpoint, miR-342-3p promoted the transactivation of fibroblast growth factor 11 (FGF11) by directly targeting its 3' untranslated regions (3'UTRs). Functionally, overexpression of exogenous FGF11 successfully rescued miR-342-3p deficiency-impaired endothelial proliferation and migration. Thus, perturbation of miR-342-3p/FGF11 cascade by hyperinsulinemia plays a causative role in the induction of vascular dysfunction in T2DM. Overall, the current study underscore an endothelial facet of miR-342-3p, which may operate as a novel epigenetic integrator linking adipogenic homeostasis and angiogenesis.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Factores de Crecimiento de Fibroblastos/genética , MicroARNs/genética , Regiones no Traducidas 3' , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/patología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/patología , Angiopatías Diabéticas/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/patología , Hiperinsulinismo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Transducción de Señal , Activación Transcripcional
19.
Lipids Health Dis ; 17(1): 122, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29793481

RESUMEN

BACKGROUND: Insulin resistant and the progression of cancer is closely related. The aim of this study was to  investigate the effect of insulin on the proliferation and migration of colon cancer cells and its underlying mechanism. METHODS: Colon carcinoma tissues from the 80 cases of colon cancer patients were collected. Immunohistochemistry was used to detect the expression of acyl coenzyme A: cholesterol acyltransferase1 (ACAT1), and we analyzed the correlation between hyperglycemia and ACAT1, hyperglycemia and metastasis. CCK8 assay and transwell assay were used to investigate the effect of different concentrations of insulin and ACAT1siRNA on human colon cancer cell line HT-29. ACAT1 mRNA expression and protein level in HT-29 cells were determined by real-time quantitative PCR and western blotting, respectively. RESULTS: Biopsies from patients with colon carcinoma showed hyperglycemia links ACAT1, lymph nodes metastasis and distant metastasis. Insulin markedly promoted cell proliferation and migration in human colon cancer HT-29 cells. Moreover, ACAT1mRNA expression and protein level were increased by insulin. ACAT1siRNA resulted in a complete inhibition of the ACAT1 mRNA expression. Consequently insulin-triggered cell proliferation and migration on colon cancer cells were inhibited. CONCLUSION: The progression of colon cancer has a positive correlation with hyperinsulinemia. Insulin-triggered cell proliferation and metastatic effects on colorectal cancer cells are mediated by ACAT1. Therefore, insulin could promote colon cancer progression by upregulation of ACAT1, which maybe is a potential therapeutic target for colon cancer.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Hiperglucemia/genética , Hiperinsulinismo/genética , Insulina/farmacología , Acetil-CoA C-Acetiltransferasa/antagonistas & inhibidores , Acetil-CoA C-Acetiltransferasa/metabolismo , Movimiento Celular , Proliferación Celular , Colesterol/metabolismo , Neoplasias del Colon/complicaciones , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Progresión de la Enfermedad , Femenino , Células HT29 , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/enzimología , Hiperglucemia/patología , Hiperinsulinismo/complicaciones , Hiperinsulinismo/enzimología , Hiperinsulinismo/patología , Insulina/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
20.
Int J Mol Sci ; 19(4)2018 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-29662006

RESUMEN

Hyperinsulinemia could have a role in the growing incidence of esophageal adenocarcinoma (EAC) and its pre-cancerous lesion, Barrett's Esophagus, a possible consequence of Gastro-Esophageal Reflux Disease. Obesity is known to mediate esophageal carcinogenesis through different mechanisms including insulin-resistance leading to hyperinsulinemia, which may mediate cancer progression via the insulin/insulin-like growth factor axis. We used the hyperinsulinemic non-obese FVB/N (Friend leukemia virus B strain) MKR (muscle (M)-IGF1R-lysine (K)-arginine (R) mouse model to evaluate the exclusive role of hyperinsulinemia in the pathogenesis of EAC related to duodeno-esophageal reflux. FVB/N wild-type (WT) and MKR mice underwent jejunum-esophageal anastomosis side-to end with the exclusion of the stomach. Thirty weeks after surgery, the esophagus was processed for histological, immunological and insulin/Insulin-like growth factor 1 (IGF1) signal transduction analyses. Most of the WT mice (63.1%) developed dysplasia, whereas most of the MKR mice (74.3%) developed squamous cell and adenosquamous carcinomas, both expressing Human Epidermal growth factor receptor 2 (HER2). Hyperinsulinemia significantly increased esophageal cancer incidence in the presence of duodenal-reflux. Insulin receptor (IR) and IGF1 receptor (IGF1R) were overexpressed in the hyperinsulinemic condition. IGF1R, through ERK1/2 mitogenic pattern activation, seems to be involved in cancer onset. Hyperinsulinemia-induced IGF1R and HER2 up-regulation could also increase the possibility of forming of IGF1R/HER2 heterodimers to support cell growth/proliferation/progression in esophageal carcinogenesis.


Asunto(s)
Reflujo Duodenogástrico/complicaciones , Neoplasias Esofágicas/etiología , Esófago/patología , Hiperinsulinismo/complicaciones , Animales , Modelos Animales de Enfermedad , Reflujo Duodenogástrico/metabolismo , Reflujo Duodenogástrico/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Esófago/metabolismo , Femenino , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulina/análisis , Insulina/metabolismo , Masculino , Ratones , Receptor ErbB-2/análisis , Receptor ErbB-2/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda