Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 291(16): 8816-24, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26896795

RESUMO

Evidence continues to emerge detailing the myriad of ways the gut microbiota influences host energy homeostasis. Among the potential mechanisms, short chain fatty acids (SCFAs), the byproducts of microbial fermentation of dietary fibers, exhibit correlative beneficial metabolic effects in humans and rodents, including improvements in glucose homeostasis. The underlying mechanisms, however, remain elusive. We here report that one of the main bacterially produced SCFAs, propionate, activates ileal mucosal free fatty acid receptor 2 to trigger a negative feedback pathway to lower hepatic glucose production in healthy rats in vivo We further demonstrate that an ileal glucagon-like peptide-1 receptor-dependent neuronal network is necessary for ileal propionate and long chain fatty acid sensing to regulate glucose homeostasis. These findings highlight the potential to manipulate fatty acid sensing machinery in the ileum to regulate glucose homeostasis.


Assuntos
Ácidos Graxos/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/metabolismo , Íleo/metabolismo , Animais , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
2.
J Biol Chem ; 289(17): 11642-11649, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24577102

RESUMO

The gut is anatomically positioned to play a critical role in the regulation of metabolic homeostasis, providing negative feedback via nutrient sensing and local hormonal signaling. Gut hormones, such as cholecystokinin (CCK) and glucagon-like peptide-1 (GLP-1), are released following a meal and act on local receptors to regulate glycemia via a neuronal gut-brain axis. Additionally, jejunal nutrient sensing and leptin action are demonstrated to suppress glucose production, and both are required for the rapid antidiabetic effect of duodenal jejunal bypass surgery. Strategies aimed at targeting local gut hormonal signaling pathways may prove to be efficacious therapeutic options to improve glucose control in diabetes.


Assuntos
Hormônios Gastrointestinais/metabolismo , Transdução de Sinais , Mucosa Gástrica/metabolismo , Humanos
3.
Gastroenterology ; 142(4): 834-843.e3, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22245844

RESUMO

BACKGROUND & AIMS: The duodenum senses nutrients to maintain energy and glucose homeostasis, but little is known about the signaling and neuronal mechanisms involved. We tested whether duodenal activation of adenosine 3',5'-cyclic monophosphate (cAMP)-dependent protein kinase A (PKA) is sufficient and necessary for cholecystokinin (CCK) signaling to trigger vagal afferent firing and regulate glucose production. METHODS: In rats, we selectively activated duodenal PKA and evaluated changes in glucose kinetics during the pancreatic (basal insulin) pancreatic clamps and vagal afferent firing. The requirement of duodenal PKA signaling in glucose regulation was evaluated by inhibiting duodenal activation of PKA in the presence of infusion of the intraduodenal PKA agonist (Sp-cAMPS) or CCK1 receptor agonist (CCK-8). We also assessed the involvement of a neuronal network and the metabolic impact of duodenal PKA activation in rats placed on high-fat diets. RESULTS: Intraduodenal infusion of Sp-cAMPS activated duodenal PKA and lowered glucose production, in association with increased vagal afferent firing in control rats. The metabolic and neuronal effects of duodenal Sp-cAMPS were negated by coinfusion with either the PKA inhibitor H89 or Rp-CAMPS. The metabolic effect was also negated by coinfusion with tetracaine, molecular and pharmacologic inhibition of NR1-containing N-methyl-d-aspartate (NMDA) receptors within the dorsal vagal complex, or hepatic vagotomy in rats. Inhibition of duodenal PKA blocked the ability of duodenal CCK-8 to reduce glucose production in control rats, whereas duodenal Sp-cAMPS bypassed duodenal CCK resistance and activated duodenal PKA and lowered glucose production in rats on high-fat diets. CONCLUSIONS: We identified a neural glucoregulatory function of duodenal PKA signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Duodeno/enzimologia , Duodeno/inervação , Glucose/metabolismo , Fígado/inervação , Fígado/metabolismo , Nervo Vago/fisiologia , Animais , Colecistocinina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Dieta Hiperlipídica , Duodeno/efeitos dos fármacos , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Técnica Clamp de Glucose , Homeostase , Antagonistas de Hormônios/farmacologia , Masculino , Pâncreas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/antagonistas & inibidores , Receptor de Colecistocinina B/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Vagotomia , Nervo Vago/efeitos dos fármacos
4.
Nat Commun ; 9(1): 1118, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29549253

RESUMO

High protein feeding improves glucose homeostasis in rodents and humans with diabetes, but the mechanisms that underlie this improvement remain elusive. Here we show that acute administration of casein hydrolysate directly into the upper small intestine increases glucose tolerance and inhibits glucose production in rats, independently of changes in plasma amino acids, insulin levels, and food intake. Inhibition of upper small intestinal peptide transporter 1 (PepT1), the primary oligopeptide transporter in the small intestine, reverses the preabsorptive ability of upper small intestinal casein infusion to increase glucose tolerance and suppress glucose production. The glucoregulatory role of PepT1 in the upper small intestine of healthy rats is further demonstrated by glucose homeostasis disruption following high protein feeding when PepT1 is inhibited. PepT1-mediated protein-sensing mechanisms also improve glucose homeostasis in models of early-onset insulin resistance and obesity. We demonstrate that preabsorptive upper small intestinal protein-sensing mechanisms mediated by PepT1 have beneficial effects on whole-body glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Dieta Rica em Proteínas , Glucose/metabolismo , Absorção Intestinal/fisiologia , Intestino Delgado/metabolismo , Transportador 1 de Peptídeos/metabolismo , Aminoácidos/sangue , Animais , Caseínas/administração & dosagem , Hiperglicemia/patologia , Insulina/sangue , Intestino Delgado/enzimologia , Masculino , Transportador 1 de Peptídeos/antagonistas & inibidores , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
5.
Cell Metab ; 27(3): 572-587.e6, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514066

RESUMO

Long-chain acyl-CoA synthetase (ACSL)-dependent upper small intestinal lipid metabolism activates pre-absorptive pathways to regulate metabolic homeostasis, but whether changes in the upper small intestinal microbiota alter specific fatty acid-dependent pathways to impact glucose homeostasis remains unknown. We here first find that upper small intestinal infusion of Intralipid, oleic acid, or linoleic acid pre-absorptively increases glucose tolerance and lowers glucose production in rodents. High-fat feeding impairs pre-absorptive fatty acid sensing and reduces upper small intestinal Lactobacillus gasseri levels and ACSL3 expression. Transplantation of healthy upper small intestinal microbiota to high-fat-fed rodents restores L. gasseri levels and fatty acid sensing via increased ACSL3 expression, while L. gasseri probiotic administration to non-transplanted high-fat-fed rodents is sufficient to restore upper small intestinal ACSL3 expression and fatty acid sensing. In summary, we unveil a glucoregulatory role of upper small intestinal L. gasseri that impacts an ACSL3-dependent glucoregulatory fatty acid-sensing pathway.


Assuntos
Coenzima A Ligases/metabolismo , Ácidos Graxos/metabolismo , Microbioma Gastrointestinal , Glucose/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Lactobacillus gasseri/metabolismo , Animais , Dieta Hiperlipídica/métodos , Emulsões/metabolismo , Transplante de Microbiota Fecal/métodos , Homeostase , Ácido Linoleico/metabolismo , Camundongos Endogâmicos C57BL , Ácido Oleico/metabolismo , Fosfolipídeos/metabolismo , Ratos Sprague-Dawley , Óleo de Soja/metabolismo
6.
Cell Metab ; 27(1): 101-117.e5, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29056513

RESUMO

The gut microbiota alters energy homeostasis. In parallel, metformin regulates upper small intestinal sodium glucose cotransporter-1 (SGLT1), but whether changes of the microbiota or SGLT1-dependent pathways in the upper small intestine mediate metformin action is unknown. Here we report that upper small intestinal glucose sensing triggers an SGLT1-dependent pathway to lower glucose production in rodents. High-fat diet (HFD) feeding reduces glucose sensing and SGLT1 expression in the upper small intestine. Upper small intestinal metformin treatment restores SGLT1 expression and glucose sensing while shifting the upper small intestinal microbiota partly by increasing the abundance of Lactobacillus. Transplantation of upper small intestinal microbiota from metformin-treated HFD rats to the upper small intestine of untreated HFD rats also increases the upper small intestinal abundance of Lactobacillus and glucose sensing via an upregulation of SGLT1 expression. Thus, we demonstrate that metformin alters upper small intestinal microbiota and impacts a glucose-SGLT1-sensing glucoregulatory pathway.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/metabolismo , Metformina/farmacologia , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Dieta Hiperlipídica , Comportamento Alimentar , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Análise de Componente Principal , Ratos
7.
Nat Med ; 21(5): 506-11, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25849133

RESUMO

Metformin is a first-line therapeutic option for the treatment of type 2 diabetes, even though its underlying mechanisms of action are relatively unclear. Metformin lowers blood glucose levels by inhibiting hepatic glucose production (HGP), an effect originally postulated to be due to a hepatic AMP-activated protein kinase (AMPK)-dependent mechanism. However, studies have questioned the contribution of hepatic AMPK to the effects of metformin on lowering hyperglycemia, and a gut-brain-liver axis that mediates intestinal nutrient- and hormone-induced lowering of HGP has been identified. Thus, it is possible that metformin affects HGP through this inter-organ crosstalk. Here we show that intraduodenal infusion of metformin for 50 min activated duodenal mucosal Ampk and lowered HGP in a rat 3 d high fat diet (HFD)-induced model of insulin resistance. Inhibition of duodenal Ampk negated the HGP-lowering effect of intraduodenal metformin, and both duodenal glucagon-like peptide-1 receptor (Glp-1r)-protein kinase A (Pka) signaling and a neuronal-mediated gut-brain-liver pathway were required for metformin to lower HGP. Preabsorptive metformin also lowered HGP in rat models of 28 d HFD-induced obesity and insulin resistance and nicotinamide (NA)-streptozotocin (STZ)-HFD-induced type 2 diabetes. In an unclamped setting, inhibition of duodenal Ampk reduced the glucose-lowering effects of a bolus metformin treatment in rat models of diabetes. These findings show that, in rat models of both obesity and diabetes, metformin activates a previously unappreciated duodenal Ampk-dependent pathway to lower HGP and plasma glucose levels.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Duodeno/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Fígado/enzimologia , Metformina/química , Animais , Glicemia/química , Diabetes Mellitus Tipo 2/sangue , Receptor do Peptídeo Semelhante ao Glucagon 1 , Técnica Clamp de Glucose , Células HEK293 , Humanos , Insulina , Resistência à Insulina , Masculino , Metformina/administração & dosagem , Niacinamida/química , Obesidade/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucagon/metabolismo , Transdução de Sinais
8.
Nat Med ; 21(5): 498-505, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25849131

RESUMO

Resveratrol improves insulin sensitivity and lowers hepatic glucose production (HGP) in rat models of obesity and diabetes, but the underlying mechanisms for these antidiabetic effects remain elusive. One process that is considered a key feature of resveratrol action is the activation of the nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase sirtuin 1 (SIRT1) in various tissues. However, the low bioavailability of resveratrol raises questions about whether the antidiabetic effects of oral resveratrol can act directly on these tissues. We show here that acute intraduodenal infusion of resveratrol reversed a 3 d high fat diet (HFD)-induced reduction in duodenal-mucosal Sirt1 protein levels while also enhancing insulin sensitivity and lowering HGP. Further, we found that duodenum-specific knockdown of Sirt1 expression for 14 d was sufficient to induce hepatic insulin resistance in rats fed normal chow. We also found that the glucoregulatory role of duodenally acting resveratrol required activation of Sirt1 and AMP-activated protein kinase (Ampk) in this tissue to initiate a gut-brain-liver neuronal axis that improved hypothalamic insulin sensitivity and in turn, reduced HGP. In addition to the effects of duodenally acting resveratrol in an acute 3 d HFD-fed model of insulin resistance, we also found that short-term infusion of resveratrol into the duodenum lowered HGP in two other rat models of insulin resistance--a 28 d HFD-induced model of obesity and a nicotinamide (NA)-streptozotocin (STZ)-HFD-induced model of mild type 2 diabetes. Together, these studies highlight the therapeutic relevance of targeting duodenal SIRT1 to reverse insulin resistance and improve glucose homeostasis in obesity and diabetes.


Assuntos
Resistência à Insulina , Rede Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Sirtuína 1/metabolismo , Estilbenos/uso terapêutico , Animais , Antioxidantes/uso terapêutico , Glicemia/química , Diabetes Mellitus/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células HEK293 , Homeostase , Humanos , Insulina/sangue , Masculino , Niacinamida/química , Obesidade/sangue , Obesidade/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Resveratrol , Estreptozocina
9.
Cell Metab ; 21(4): 527-42, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25863246

RESUMO

Obesity has reached epidemic proportions, but little is known about its influence on the intestinal immune system. Here we show that the gut immune system is altered during high-fat diet (HFD) feeding and is a functional regulator of obesity-related insulin resistance (IR) that can be exploited therapeutically. Obesity induces a chronic phenotypic pro-inflammatory shift in bowel lamina propria immune cell populations. Reduction of the gut immune system, using beta7 integrin-deficient mice (Beta7(null)), decreases HFD-induced IR. Treatment of wild-type HFD C57BL/6 mice with the local gut anti-inflammatory, 5-aminosalicyclic acid (5-ASA), reverses bowel inflammation and improves metabolic parameters. These beneficial effects are dependent on adaptive and gut immunity and are associated with reduced gut permeability and endotoxemia, decreased visceral adipose tissue inflammation, and improved antigen-specific tolerance to luminal antigens. Thus, the mucosal immune system affects multiple pathways associated with systemic IR and represents a novel therapeutic target in this disease.


Assuntos
Anti-Inflamatórios/farmacologia , Trato Gastrointestinal/imunologia , Imunidade nas Mucosas/imunologia , Resistência à Insulina/imunologia , Obesidade/imunologia , Animais , Western Blotting , Citocinas/sangue , Dieta Hiperlipídica/efeitos adversos , Citometria de Fluxo , Trato Gastrointestinal/efeitos dos fármacos , Técnicas Histológicas , Imuno-Histoquímica , Cadeias beta de Integrinas/metabolismo , Mesalamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/citologia , Mucosa/imunologia
10.
Cell Metab ; 19(1): 155-61, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24361011

RESUMO

The fat-derived hormone leptin binds to its hypothalamic receptors to regulate glucose homeostasis. Leptin is also synthesized in the stomach and subsequently binds to its receptors expressed in the intestine, although the functional relevance of such activation remains largely unknown. We report here that intrajejunal leptin administration activates jejunal leptin receptors and signals through a phosphatidylinositol 3-kinase (PI3K)-dependent and signal transducer and activator of transcription 3 (STAT3)-independent signaling pathway to lower glucose production in healthy rodents. Jejunal leptin action is sufficient to lower glucose production in uncontrolled diabetic and high-fat-fed rodents and contributes to the early antidiabetic effect of duodenal-jejunal bypass surgery. These data unveil a glucoregulatory site of leptin action and suggest that enhancing leptin-PI3K signaling in the jejunum lowers plasma glucose concentrations in diabetes.


Assuntos
Glucose/biossíntese , Jejuno/enzimologia , Leptina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Animais , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica , Procedimentos Cirúrgicos do Sistema Digestório , Hipoglicemiantes/farmacologia , Jejuno/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Diabetes ; 62(9): 3005-13, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23970519

RESUMO

The small intestine is traditionally viewed as an organ that mediates nutrient digestion and absorption. This view has recently been revised owing to the ability of the duodenum to sense nutrient influx and trigger negative feedback loops to inhibit glucose production and food intake to maintain metabolic homeostasis. Further, duodenal nutrient-sensing defects are acquired in diabetes and obesity, leading to increased glucose production. In contrast, jejunal nutrient sensing inhibits glucose production and mediates the early antidiabetic effect of bariatric surgery, and gut microbiota composition may alter intestinal nutrient-sensing mechanisms to regain better control of glucose homeostasis in diabetes and obesity in the long term. This perspective highlights nutrient-sensing mechanisms in the gut that regulate glucose homeostasis and the potential of targeting gut nutrient-sensing mechanisms as a therapeutic strategy to lower blood glucose concentrations in diabetes.


Assuntos
Trato Gastrointestinal/metabolismo , Glucose/metabolismo , Glicemia/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia
12.
Trends Endocrinol Metab ; 23(2): 49-55, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22169756

RESUMO

Elevation of lipid levels affects energy and glucose homeostasis. Organs such as the gut, brain and liver detect a rise in lipids and orchestrate a biochemical, molecular, neuronal and physiological network of responses that alters appetite and the rate of hepatic glucose production. The factors involved in these responses are unclear but the formation of esterified lipids (long-chain fatty acyl-CoAs) and subsequent activation of protein kinase Cδ remain a common sensing mechanism in all three organs. In this paper, we discuss the mechanisms underlying lipid sensing within the gut, brain and liver and their physiological impact on the regulation of glucose and energy homeostasis.


Assuntos
Encéfalo/metabolismo , Trato Gastrointestinal/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Acil Coenzima A/metabolismo , Animais , Colecistocinina/fisiologia , Glucose/biossíntese , Glucose/metabolismo , Homeostase/fisiologia , Humanos , Fragmentos de Peptídeos/fisiologia , Proteína Quinase C-delta/metabolismo , Receptor de Colecistocinina A/fisiologia , Receptor de Colecistocinina B/fisiologia , Transdução de Sinais/fisiologia
13.
Nat Med ; 18(6): 950-5, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22610279

RESUMO

Gastrointestinal bypass surgeries restore metabolic homeostasis in patients with type 2 diabetes and obesity(1), but the underlying mechanisms remain elusive. Duodenal-jejunal bypass surgery (DJB), an experimental surgical technique that excludes the duodenum and proximal jejunum from nutrient transit(1,2), lowers glucose concentrations in nonobese type 2 diabetic rats(2­5). Given that DJB redirects and enhances nutrient flow into the jejunum and that jejunal nutrient sensing affects feeding(6,7), the repositioned jejunum after DJB represents a junction at which nutrients could regulate glucose homeostasis. Here we found that intrajejunal nutrient administration lowered endogenous glucose production in normal rats through a gut-brain-liver network in the presence of basal plasma insulin concentrations. Inhibition of jejunal glucose uptake or formation of long chain fatty acyl-coA negated the metabolic effects of glucose or lipid, respectively, in normal rats, and altered the rapid (2 d) glucose-lowering effect induced by DJB in streptozotocin (STZ)-induced uncontrolled diabetic rats during refeeding. Lastly, in insulin-deficient autoimmune type 1 diabetic rats and STZ-induced diabetic rats, DJB lowered glucose concentrations in 2 d independently of changes in plasma insulin concentrations, food intake and body weight. These data unveil a glucoregulatory role of jejunal nutrient sensing and its relevance in the early improvement of glycemic control after DJB in rat models of uncontrolled diabetes.


Assuntos
Glicemia/análise , Diabetes Mellitus Experimental/cirurgia , Duodeno/cirurgia , Jejuno/cirurgia , Animais , Diabetes Mellitus Experimental/sangue , Derivação Gástrica , Masculino , Ratos , Ratos Sprague-Dawley , Estreptozocina
14.
Diabetes ; 60(12): 3148-53, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21984583

RESUMO

OBJECTIVE: Metabolism of long-chain fatty acids within the duodenum leads to the activation of duodenal mucosal protein kinase C (PKC)-δ and the cholecystokinin (CCK)-A receptor to lower glucose production through a neuronal network. However, the interfunctional relationship between duodenal PKC-δ and CCK remains elusive. Although long-chain fatty acids activate PKC to stimulate the release of CCK in CCK-secreting cells, CCK has also been found to activate PKC-δ in pancreatic acinar cells. We here evaluate whether activation of duodenal mucosal PKC-δ lies upstream (and/or downstream) of CCK signaling to lower glucose production. RESEARCH DESIGN AND METHODS: We first determined with immunofluorescence whether PKC-δ and CCK were colocalized within the duodenal mucosa. We then performed gain- and loss-of-function experiments targeting duodenal PKC-δ and the CCK-A receptor and evaluated the impact on changes in glucose kinetics during pancreatic (basal insulin) clamps in rats in vivo. RESULTS: Immunostaining of PKC-δ was found to colocalize with CCK in the duodenal mucosa. Intraduodenal coinfusion of either the CCK-A receptor antagonist MK-329 or CR-1409 with the PKC activator negated the ability of duodenal mucosal PKC-δ activation to lower glucose production during the pancreatic clamps in normal rats. Conversely, molecular and pharmacological inhibition of duodenal PKC-δ did not negate the ability of the duodenal CCK-A receptor agonist CCK-8 to lower glucose production, indicating that activation of duodenal PKC-δ lies upstream (and not downstream) of CCK signaling. Finally, intraduodenal PKC activator infusion failed to lower glucose production in rats with high-fat diet-induced duodenal CCK resistance. CONCLUSIONS: In summary, activation of duodenal PKC-δ leads to the stimulation of CCK release and activation of the CCK-A receptor signaling axis to lower glucose production in normal rats, but fails to bypass duodenal CCK-resistance in high fat-fed rats.


Assuntos
Colecistocinina/metabolismo , Duodeno/metabolismo , Glucose/metabolismo , Proteína Quinase C-delta/metabolismo , Transdução de Sinais/fisiologia , Animais , Duodeno/enzimologia , Imunofluorescência , Masculino , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina A/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa