Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes Ther ; 12(7): 1871-1886, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34047962

RESUMO

INTRODUCTION: While continuous glucose monitoring (CGM) has been shown to decrease both hyper- and hypoglycemia in insulin-treated diabetes, its value in non-insulin-treated type 2 diabetes (T2D) and prediabetes is unclear. Studies examining the reduction in hyperglycemia with the use of CGM in non-insulin-treated T2D are limited. METHODS: We investigated the potential benefit of CGM combined with a mobile app that links each individual's glucose tracing to meal composition, heart rate, and physical activity in a cohort of 1022 individuals, ranging from nondiabetic to non-insulin-treated T2D, spanning a wide range of demographic, geographic, and socioeconomic characteristics. The primary endpoint was the change in time in range (TIR), defined as 54-140 mg/dL for healthy and prediabetes, and 54-180 mg/dL for T2D, from the beginning to end of a 10-day period of use of the Freestyle Libre CGM. Logged food intake, physical activity, continuous glucose, and heart rate data were captured by a smartphone-based app that continuously provided feedback to participants, overlaying daily glucose patterns with activity and food intake, including macronutrient breakdown, glycemic load (GL), and glycemic index (GI). RESULTS: A total of 665 participants meeting eligibility and data requirements were included in the final analysis. Among self-reported nondiabetic participants, CGM identified glucose excursions in the diabetic range among 15% of healthy and 36% of those with prediabetes. In the group as a whole, TIR improved significantly (p < 0.001). Among the 51.4% of participants who improved, TIR increased by an average of 6.4% (p < 0.001). Of those with poor baseline TIR, defined as TIR below comparable A1c thresholds for T2D and prediabetes, 58.3% of T2D and 91.7% of healthy/prediabetes participants improved their TIR by an average of 22.7% and 23.2%, respectively. Predictors of improved response included no prior diagnosis of T2D and lower BMI. CONCLUSIONS: These results indicate that 10-day use of CGM as a part of multimodal data collection, with synthesis and feedback to participants provided by a mobile health app, can significantly reduce hyperglycemia in non-insulin-treated individuals, including those with early stages of glucose dysregulation.

2.
J Immunol ; 205(11): 3191-3204, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33148717

RESUMO

IL-10 is a potent anti-inflammatory cytokine capable of suppressing a number of proinflammatory signals associated with intestinal inflammatory diseases, such as ulcerative colitis and Crohn's disease. Clinical use of human IL-10 (hIL-10) has been limited by anemia and thrombocytopenia following systemic injection, side effects that might be eliminated by a gut-restricted distribution. We have identified a transcytosis pathway used by cholix, an exotoxin secreted by nonpandemic forms of the intestinal pathogen Vibrio cholerae A nontoxic fragment of the first 386 aa of cholix was genetically fused to hIL-10 to produce recombinant AMT-101. In vitro and in vivo characterization of AMT-101 showed it to efficiently cross healthy human intestinal epithelium (SMI-100) by a vesicular transcytosis process, activate hIL-10 receptors in an engineered U2OS osteosarcoma cell line, and increase cellular phospho-STAT3 levels in J774.2 mouse macrophage cells. AMT-101 was taken up by inflamed intestinal mucosa and activated pSTAT3 in the lamina propria with limited systemic distribution. AMT-101 administered to healthy mice by oral gavage or to cynomolgus monkeys (nonhuman primates) by colonic spray increased circulating levels of IL-1R antagonist (IL-1Ra). Oral gavage of AMT-101 in two mouse models of induced colitis prevented associated pathological events and plasma cytokine changes. Overall, these studies suggest that AMT-101 can efficiently overcome the epithelial barrier to focus biologically active IL-10 to the intestinal lamina propria.


Assuntos
Colite/metabolismo , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Animais , Células Cultivadas , Colo/metabolismo , Doença de Crohn/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Macaca fascicularis , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Mucosa/metabolismo , Ratos , Ratos Wistar , Transcitose/fisiologia
3.
Diabetes ; 63(8): 2866-75, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24727435

RESUMO

Hypoglycemic detection at the portal-mesenteric vein (PMV) appears mediated by spinal afferents and is critical for the counter-regulatory response (CRR) to slow-onset, but not rapid-onset, hypoglycemia. Since rapid-onset hypoglycemia induces Fos protein expression in discrete brain regions, we hypothesized that denervation of the PMV or lesioning spinal afferents would suppress Fos expression in the dorsal medulla during slow-onset hypoglycemia, revealing a central nervous system reliance on PMV glucosensors. Rats undergoing PMV deafferentation via capsaicin, celiac-superior mesenteric ganglionectomy (CSMG), or total subdiaphragmatic vagotomy (TSV) were exposed to hyperinsulinemic-hypoglycemic clamps where glycemia was lowered slowly over 60-75 min. In response to hypoglycemia, control animals demonstrated a robust CRR along with marked Fos expression in the area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus. Fos expression was suppressed by 65-92% in capsaicin-treated animals, as was epinephrine (74%), norepinephrine (33%), and glucagon (47%). CSMG also suppressed Fos expression and CRR during slow-onset hypoglycemia, whereas TSV failed to impact either. In contrast, CSMG failed to impact upon Fos expression or the CRR during rapid-onset hypoglycemia. Peripheral glucosensory input from the PMV is therefore required for activation of hindbrain neurons and the full CRR during slow-onset hypoglycemia.


Assuntos
Hipoglicemia/metabolismo , Veias Mesentéricas/fisiologia , Veia Porta/fisiologia , Receptores de Superfície Celular/fisiologia , Rombencéfalo/citologia , Animais , Capsaicina , Ganglionectomia , Regulação da Expressão Gênica/fisiologia , Técnica Clamp de Glucose , Masculino , Proteínas Oncogênicas v-fos/genética , Proteínas Oncogênicas v-fos/metabolismo , Ratos , Ratos Wistar , Vagotomia
4.
Diabetes ; 62(4): 1074-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23248171

RESUMO

Gastrointestinal bypass surgeries that result in rerouting and subsequent exclusion of nutrients from the duodenum appear to rapidly alleviate hyperglycemia and hyperinsulinemia independent of weight loss. While the mechanism(s) responsible for normalization of glucose homeostasis remains to be fully elucidated, this rapid normalization coupled with the well-known effects of vagal inputs into glucose homeostasis suggests a neurohormonally mediated mechanism. Our results show that duodenal bypass surgery on obese, insulin-resistant Zucker fa/fa rats restored insulin sensitivity in both liver and peripheral tissues independent of body weight. Restoration of normoglycemia was attributable to an enhancement in key insulin-signaling molecules, including insulin receptor substrate-2, and substrate metabolism through a multifaceted mechanism involving activation of AMP-activated protein kinase and downregulation of key regulatory genes involved in both lipid and glucose metabolism. Importantly, while central nervous system-derived vagal nerves were not essential for restoration of insulin sensitivity, rapid normalization in hepatic gluconeogenic capacity and basal hepatic glucose production required intact vagal innervation. Lastly, duodenal bypass surgery selectively altered the tissue concentration of intestinally derived glucoregulatory hormone peptides in a segment-specific manner. The present data highlight and support the significance of vagal inputs and intestinal hormone peptides toward normalization of glucose and lipid homeostasis after duodenal bypass surgery.


Assuntos
Glicemia/metabolismo , Duodeno/cirurgia , Derivação Gástrica/métodos , Intolerância à Glucose/metabolismo , Tecido Adiposo/metabolismo , Animais , Calorimetria Indireta , Glucose/metabolismo , Hiperinsulinismo , Fígado/metabolismo , Ratos , Ratos Zucker , Redução de Peso
5.
Mol Endocrinol ; 24(7): 1413-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20444886

RESUMO

Recent findings denote an important contribution of macrophage inflammatory pathways in causing obesity-related insulin resistance. Inducible nitric oxide synthase (iNOS) is activated in proinflammatory macrophages and modestly elevated in insulin-responsive tissues. Although the benefits of systemic iNOS inhibition in insulin-resistant models have been demonstrated, the role of macrophage iNOS in metabolic disorders is not clear. In the current work, we used bone marrow transplantation (BMT) to generate mice with myeloid iNOS deficiency [iNOS BMT knockout (KO)]. Interestingly, disruption of iNOS in myeloid cells did not protect mice from high-fat diet-induced obesity and insulin resistance. When mice were treated with the iNOS inhibitor, N6-(1-Iminoethyl)-L-lysine hydrochloride (L-NIL), we observed a significant and comparable improvement of glucose homeostasis and insulin sensitivity in both wild-type and iNOS BMT KO mice. We further demonstrated that absence of iNOS in primary macrophages did not affect acute TLR4 signaling pathways and had only a modest and mixed effect on inflammatory gene expression. With respect to TNFalpha treatment, iNOS KO macrophages showed, if anything, a greater inflammatory response. In summary, we conclude that iNOS inhibition in tissues other than myeloid cells is responsible for the beneficial effects in obesity/insulin resistance.


Assuntos
Resistência à Insulina/fisiologia , Células Mieloides/metabolismo , Óxido Nítrico Sintase Tipo II/fisiologia , Animais , Western Blotting , Transplante de Medula Óssea , Células Cultivadas , Gorduras na Dieta/efeitos adversos , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica , Resistência à Insulina/genética , Lisina/análogos & derivados , Lisina/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/enzimologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Obesidade/induzido quimicamente , Obesidade/genética
6.
Proc Natl Acad Sci U S A ; 106(52): 22504-9, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20018750

RESUMO

Although peroxisome proliferator-activated receptor gamma (PPARgamma) agonists such as thiazolidinediones (TZDs) are widely used to treat type 2 diabetes, how its activation in individual tissues contributes to TZD's therapeutic action remains controversial. As TZDs are known to have receptor-independent effects, we sought to establish gain-of-function animal models to delineate the receptor's insulin-sensitizing actions. Unexpectedly, we find that selective activation of PPARgamma in adipocytes, but not in macrophages, is sufficient for whole-body insulin sensitization equivalent to systemic TZD treatment. In addition to improved adipokine, inflammatory, and lipid profiles, PPARgamma activation in mature adipocytes normalizes serum insulin without increased adipogenesis. Co-culture studies indicated that PPARgamma-activated adipocytes broadly suppress induction of inflammatory cytokines and C-X-C family chemokines in macrophages. Collectively, these data describe an "adipocentric" model in which adipose activation of PPARgamma is sufficient for complete insulin sensitization and suggest a specific application for fat selective PPARgamma modulators in diabetic therapy.


Assuntos
Adipócitos Brancos/metabolismo , Insulina/metabolismo , PPAR gama/metabolismo , Células 3T3-L1 , Adipócitos Brancos/efeitos dos fármacos , Animais , Linhagem Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Expressão Gênica , Humanos , Hipoglicemiantes/farmacologia , Mediadores da Inflamação/metabolismo , Insulina/sangue , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , PPAR gama/agonistas , PPAR gama/genética , Pioglitazona , Ratos , Ratos Zucker , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Tiazolidinedionas/farmacologia
7.
Cell Metab ; 10(5): 419-29, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19883619

RESUMO

Chronic low-grade inflammation, particularly in adipose tissue, is an important modulator of obesity-induced insulin resistance. The Toll-like receptor 4 (Tlr4) is a key initiator of inflammatory responses in macrophages. We performed bone marrow transplantation (BMT) of Tlr4lps-del or control C57Bl/10J donor cells into irradiated wild-type C57Bl6 recipient mice to generate hematopoietic cell-specific Tlr4 deletion mutant (BMT-Tlr4(-/-)) and control (BMT-WT) mice. After 16 weeks of a high-fat diet (HFD), BMT-WT mice developed obesity, hyperinsulinemia, glucose intolerance, and insulin resistance. In contrast, BMT-Tlr4(-/-) mice became obese but did not develop fasting hyperinsulinemia and had improved hepatic and adipose insulin sensitivity during euglycemic clamp studies, compared to HFD BMT-WT controls. HFD BMT-Tlr4(-/-) mice also showed markedly reduced adipose tissue inflammatory markers and macrophage content. In summary, our results indicate that Tlr4 signaling in hematopoietic-derived cells is important for the development of hepatic and adipose tissue insulin resistance in obese mice.


Assuntos
Tecido Adiposo/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Obesidade/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo , Tecido Adiposo/patologia , Animais , Gorduras na Dieta/administração & dosagem , Técnicas de Inativação de Genes , Técnica Clamp de Glucose , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Obesos , Obesidade/etiologia , Obesidade/patologia , Especificidade de Órgãos , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/genética
8.
Am J Physiol Endocrinol Metab ; 297(5): E1137-46, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19706791

RESUMO

The transcription factor TORC2 [transducer of regulated cAMP-responsive element-binding protein (CREB) activity 2] is a major regulator of hepatic gluconeogenesis and is increased in hyperglycemic rodent models. Because chronic hyperglycemia and increased hepatic glucose production, via increased gluconeogenesis, is a key feature of type 2 diabetes, an effective in vivo method to efficiently knock down TORC2 could provide a potential therapy for treating hyperglycemia and type 2 diabetes. To assess this, primary mouse hepatocytes, high-fat diet (HFD)-fed mice, and Zucker diabetic fatty (ZDF) rats were treated with a siRNA against TORC2 (siTORC2), which was delivered via a novel lipid nanoparticle system, or control siRNA (siCON). Compared with siCON, administration of siTORC2 resulted in highly efficient, sustained (1-3 wk) knockdown of TORC2 and its gluconeogenic target genes phosphoenolpyruvate carboxykinase and glucose-6-phophatase in primary mouse hepatocytes and in the livers of HFD-fed mice. In mice, this knockdown was specific to the liver and did not occur in kidney, skeletal muscle, or adipose tissue. In HFD-fed mice, siTORC2 reduced in vivo gluconeogenic capacity, fasting hepatic glucose production, and hyperglycemia, and led to improved hepatic and skeletal muscle insulin sensitivity. siTORC2 treatment also improved systemic hyperglycemia in ZDF rats. In conclusion, these results demonstrate the importance of TORC2 in modulating HGP in vivo and highlight a novel, liver-specific siRNA approach for the potential treatment of hyperglycemia and type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Hiperglicemia/tratamento farmacológico , Fígado/metabolismo , RNA Interferente Pequeno/farmacologia , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Animais , Glicemia/metabolismo , Western Blotting , Células Cultivadas , Química Farmacêutica , Gluconeogênese/efeitos dos fármacos , Técnica Clamp de Glucose , Hepatócitos/metabolismo , Homeostase/efeitos dos fármacos , Insulina/sangue , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Ácido Pirúvico/metabolismo , RNA Interferente Pequeno/efeitos adversos , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Zucker , Transativadores/antagonistas & inibidores , Transativadores/biossíntese , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/biossíntese
9.
J Biol Chem ; 284(42): 28498-509, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19706599

RESUMO

Chromogranin A (CHGA/Chga), a proprotein, widely distributed in endocrine and neuroendocrine tissues (not expressed in muscle, liver, and adipose tissues), generates at least four bioactive peptides. One of those peptides, pancreastatin (PST), has been reported to interfere with insulin action. We generated a Chga knock-out (KO) mouse by the targeted deletion of the Chga gene in neuroendocrine tissues. KO mice displayed hypertension, higher plasma catecholamine, and adipokine levels and lower IL-6 and lipid levels compared with wild type mice. Liver glycogen content was elevated, but the nitric oxide (NO) level was diminished. Glucose, insulin, and pyruvate tolerance tests and hyperinsulinemic-euglycemic clamp studies established increased insulin sensitivity in liver but decreased glucose disposal in muscle. Despite higher catecholamine and ketone body levels and muscle insulin resistance, KO mice maintained euglycemia due to increased liver insulin sensitivity. Suppressed mRNA abundance of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase (G6Pase) in KO mice further support this conclusion. PST administration in KO mice stimulated phosphoenolpyruvate carboxykinase and G6Pase mRNA abundance and raised the blood glucose level. In liver cells transfected with G6Pase promoter, PST caused transcriptional activation in a protein kinase C (PKC)- and NO synthase-dependent manner. Thus, PST action may be mediated by suppressing IRS1/2-phosphatidylinositol 3-kinase-Akt-FOXO-1 signaling and insulin-induced maturation of SREBP1c by PKC and a high level of NO. The combined effects of conventional PKC and endothelial NO synthase activation by PST can suppress insulin signaling. The rise in blood PST level with age and in diabetes suggests that PST is a negative regulator of insulin sensitivity and glucose homeostasis.


Assuntos
Cromogranina A/genética , Cromogranina A/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Hormônios Pancreáticos/fisiologia , Adipócitos/metabolismo , Animais , Composição Corporal , Homeostase , Lipídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/química , Hormônios Pancreáticos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
10.
Cell Metab ; 9(3): 277-86, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19254572

RESUMO

Increases in adiposity trigger metabolic and inflammatory changes that interfere with insulin action in peripheral tissues, culminating in beta cell failure and overt diabetes. We found that the cAMP Response Element Binding protein (CREB) is activated in adipose cells under obese conditions, where it promotes insulin resistance by triggering expression of the transcriptional repressor ATF3 and thereby downregulating expression of the adipokine hormone adiponectin as well as the insulin-sensitive glucose transporter 4 (GLUT4). Transgenic mice expressing a dominant-negative CREB transgene in adipocytes displayed increased whole-body insulin sensitivity in the contexts of diet-induced and genetic obesity, and they were protected from the development of hepatic steatosis and adipose tissue inflammation. These results indicate that adipocyte CREB provides an early signal in the progression to type 2 diabetes.


Assuntos
Adipócitos/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Resistência à Insulina/fisiologia , Obesidade/fisiopatologia , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Adenilato Quinase/metabolismo , Adipócitos/citologia , Adipócitos/patologia , Adiponectina/metabolismo , Animais , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Progressão da Doença , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Gluconeogênese/fisiologia , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Fatores de Transcrição/metabolismo
11.
J Clin Invest ; 118(9): 2992-3002, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18769626

RESUMO

Insulin resistance is a major metabolic feature of obesity and is a key factor in the etiology of a number of diseases, including type 2 diabetes. In this review, we discuss potential mechanisms by which brief nutrient excess and obesity lead to insulin resistance and propose that these mechanisms of action are different but interrelated. We discuss how pathways that "sense" nutrients within skeletal muscle are readily able to regulate insulin action. We then discuss how obesity leads to insulin resistance via a complex interplay among systemic fatty acid excess, microhypoxia in adipose tissue, ER stress, and inflammation. In particular, we focus on the hypothesis that the macrophage is an important cell type in the propagation of inflammation and induction of insulin resistance in obesity. Overall, we provide our integrative perspective regarding how nutrients and obesity interact to regulate insulin sensitivity.


Assuntos
Inflamação/complicações , Insulina/metabolismo , Tecido Adiposo/metabolismo , Animais , Peso Corporal , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Resistência à Insulina , Secreção de Insulina , Fígado/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , Músculo Esquelético/metabolismo , Obesidade/complicações
12.
Diabetes ; 57(5): 1380-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18292347

RESUMO

OBJECTIVE: To ascertain whether portal glucose sensing extends beyond the portal vein to the superior mesenteric vein and then test whether the role of portal-superior mesenteric glucose sensors varies with the rate of fall in glycemia. RESEARCH DESIGN AND METHODS: Chronically cannulated rats underwent afferent ablation of the portal vein (PV) or portal and superior mesenteric veins (PMV) or sham operation (control). One week later, animals underwent hyperinsulinemic-hypoglycemic clamps in which the hypoglycemic nadir, 2.48 +/- 0.06 mmol/l, was reached at a rate of decline in glucose of -0.09 or -0.21 mmol x l(-1) x min(-1) (PMV and control only). Additional PMV and control animals received an intravenous injection of the glucopenic agent 2-deoxyglucose. RESULTS; Inducing hypoglycemia slowly, at a rate of -0.09 mmol x l(-1) x min(-1), resulted in a 26-fold increase in epinephrine (23.39 +/- 0.62 nmol/l) and 12-fold increase in norepinephrine (11.42 +/- 0.92 nmol/l) for controls (P < 0.001). The epinephrine response to hypoglycemia was suppressed by 91% in PMV (2.09 +/- 0.07 nmol/l) vs. 61% in PV (9.05 +/- 1.59 nmol/l) (P < 0.001). The norepinephrine response to hypoglycemia was suppressed by 94 and 80% in PMV and PV, respectively, compared with that in controls. In contrast, when arterial glucose was lowered to 2.49 +/- 0.06 mmol/l within 20 min, no significant differences were observed in the catecholamine responses for PMV and controls over the first 45 min of hypoglycemia (20-65 min). Only at min 105 were catecholamines significantly lower for PMV vs. controls. Injection of 2-deoxyglucose induced a very rapid sympathoadrenal response with no significant differences between PMV and controls. CONCLUSIONS: The critical locus for hypoglycemic detection shifts away from the portal-mesenteric vein to some other loci (e.g., the brain) when hypoglycemia develops rapidly.


Assuntos
Glicemia/metabolismo , Hipoglicemia/diagnóstico , Veias Mesentéricas/fisiopatologia , Veia Porta/fisiopatologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/sangue , Desoxiglucose/farmacologia , Técnica Clamp de Glucose , Hiperinsulinismo/fisiopatologia , Hipoglicemia/metabolismo , Masculino , Ratos , Ratos Wistar
13.
Am J Physiol Endocrinol Metab ; 293(3): E857-64, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17638706

RESUMO

Antecedent hypoglycemia leads to impaired counterregulation and hypoglycemic unawareness. To ascertain whether antecedent portal vein hypoglycemia impairs portal vein glucose sensing, thereby inducing counterregulatory failure, we compared the effects of antecedent hypoglycemia, with and without normalization of portal vein glycemia, upon the counterregulatory response to subsequent hypoglycemia. Male Wistar rats were chronically cannulated in the carotid artery (sampling), jugular vein (glucose and insulin infusion), and mesenteric vein (glucose infusion). On day 1, the following three distinct antecedent protocols were employed: 1) HYPO-HYPO: systemic hypoglycemia (2.52 +/- 0.11 mM); 2) HYPO-EUG: systemic hypoglycemia (2.70 +/- 0.03 mM) with normalization of portal vein glycemia (portal vein glucose = 5.86 +/- 0.10 mM); and 3) EUG-EUG: systemic euglycemia (6.33 +/- 0.31 mM). On day 2, all groups underwent a hyperinsulinemic-hypoglycemic clamp in which the fall in glycemia was controlled so as to reach the nadir (2.34 +/- 0.04 mM) by minute 75. Counterregulatory hormone responses were measured at basal (-30 and 0) and during hypoglycemia (60-105 min). Compared with EUG-EUG, antecedent hypoglycemia (HYPO-HYPO) significantly blunted the peak epinephrine (10.44 +/- 1.35 vs. 15.75 +/- 1.33 nM: P = 0.01) and glucagon (341 +/- 16 vs. 597 +/- 82 pg/ml: P = 0.03) responses to next-day hypoglycemia. Normalization of portal glycemia during systemic hypoglycemia on day 1 (HYPO-EUG) prevented blunting of the peak epinephrine (15.59 +/- 1.43 vs. 15.75 +/- 1.33 nM: P = 0.94) and glucagon (523 +/- 169 vs. 597 +/- 82 pg/ml: P = 0.66) responses to day 2 hypoglycemia. Consistent with hormonal responses, the glucose infusion rate during day 2 hypoglycemia was substantially elevated in HYPO-HYPO (74 +/- 12 vs. 49 +/- 4 micromol x kg(-1) x min(-1); P = 0.03) but not HYPO-EUG (39 +/- 7 vs. 49 +/- 4 micromol x kg(-1) x min(-1): P = 0.36). Antecedent hypoglycemia local to the portal vein is required for the full induction of hypoglycemia-associated counterregulatory failure with slow-onset hypoglycemia.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Glucose/metabolismo , Hipoglicemia/fisiopatologia , Veia Porta/inervação , Veia Porta/fisiopatologia , Animais , Masculino , Ratos , Ratos Wistar
14.
Metabolism ; 51(7): 859-63, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12077731

RESUMO

The aim of this investigation was to determine if a high-fat diet impaired and subsequent leptin administration improved non-insulin-stimulated (basal) glucose transport in rodents. Twenty-four male Sprague Dawley rats were divided into 1 of 2 groups: (1) normal diet (control [CON], n = 8) or (2) high-fat diet (n = 16) and received standard rat chow or a high-fat diet, respectively, for 12 weeks. The high-fat diet animals were then further subdivided into high fat (HF) (n = 8) or high-fat-leptin (HF-LEP) (n = 8) groups. The HF-LEP animals were injected with leptin (10 mg leptin/kg/d), while the CON and HF animals received vehicle over a 12-day treatment period. Following the 12-day treatment period, all animals were subjected to hind limb perfusion to assess rates of basal skeletal muscle 3-O-methyl-D-glucose (3-MG) transport. Compared with the CON group, rates of 3-MG transport were reduced in the soleus (sol) and plantaris (plant) of the HF, but not the HF-LEP animals. Differences in skeletal muscle 3-MG transport could not be accounted for by an altered GLUT1 protein concentration. In contrast, a high-fat diet reduced and chronic leptin treatment normalized the skeletal muscle GLUT4 protein concentration. The results indicate that a high-fat diet reduces and subsequent leptin treatment improves basal skeletal muscle glucose transport in a fiber-type-specific manner, but these changes do not appear to be due to alterations in the GLUT1 protein concentration.


Assuntos
Gorduras na Dieta/farmacologia , Glucose/metabolismo , Leptina/administração & dosagem , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , 3-O-Metilglucose/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 4 , Membro Posterior , Masculino , Proteínas de Transporte de Monossacarídeos/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Perfusão , Ratos , Ratos Sprague-Dawley , Tempo , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...