Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nutrients ; 15(9)2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37432375

RESUMEN

The gut microbiome plays an essential role in regulating lipid metabolism. However, little is known about how gut microbiome modulates sex differences in lipid metabolism. The present study aims to determine whether gut microbiota modulates sexual dimorphism of lipid metabolism in mice fed a high-fat diet (HFD). Conventional and germ-free male and female mice were fed an HFD for four weeks, and lipid absorption, plasma lipid profiles, and apolipoprotein levels were then evaluated. The gut microbiota was analyzed by 16S rRNA gene sequencing. After 4-week HFD consumption, the females exhibited less body weight gain and body fat composition and significantly lower triglyceride levels in very-low-density lipoprotein (VLDL) and cholesterol levels in high-density lipoprotein (HDL) compared to male mice. The fecal microbiota analysis revealed that the male mice were associated with reduced gut microbial diversity. The female mice had considerably different microbiota composition compared to males, e.g., enriched growth of beneficial microbes (e.g., Akkermansia) and depleted growth of Adlercreutzia and Enterococcus. Correlation analyses suggested that the different compositions of the gut microbiota were associated with sexual dimorphism in body weight, fat mass, and lipid metabolism in mice fed an HFD. Our findings demonstrated significant sex differences in lipid metabolism and the microbiota composition at baseline (during LFD), along with sex-dependent responses to HFD. A comprehensive understanding of sexual dimorphism in lipid metabolism modulated by microbiota will help to develop more sex-specific effective treatment options for dyslipidemia and metabolic disorders in females.


Asunto(s)
Microbioma Gastrointestinal , Femenino , Masculino , Animales , Ratones , Caracteres Sexuales , Dieta Alta en Grasa/efectos adversos , Metabolismo de los Lípidos , ARN Ribosómico 16S/genética , Peso Corporal , Lipoproteínas HDL
2.
Nutrients ; 15(11)2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37299447

RESUMEN

Long-chain fatty acids induce apolipoprotein A4 (APOA4) production in the small intestine and activate brown adipose tissue (BAT) thermogenesis. The increase in BAT thermogenesis enhances triglyceride clearance and insulin sensitivity. Acute administration of recombinant APOA4 protein elevates BAT thermogenesis in chow-fed mice. However, the physiological role of continuous infusion of recombinant APOA4 protein in regulating sympathetic activity, thermogenesis, and lipid and glucose metabolism in low-fat-diet (LFD)-fed mice remained elusive. The hypothesis of this study was that continuous infusion of mouse APOA4 protein would increase sympathetic activity and thermogenesis in BAT and subcutaneous inguinal white adipose tissue (IWAT), attenuate plasma lipid levels, and improve glucose tolerance. To test this hypothesis, sympathetic activity, BAT temperature, energy expenditure, body weight, fat mass, caloric intake, glucose tolerance, and levels of BAT and IWAT thermogenic and lipolytic proteins, plasma lipids, and markers of fatty acid oxidation in the liver in mice with APOA4 or saline treatment were measured. Plasma APOA4 levels were elevated, BAT temperature and thermogenesis were upregulated, and plasma triglyceride (TG) levels were reduced, while body weight, fat mass, caloric intake, energy expenditure, and plasma cholesterol and leptin levels were comparable between APOA4- and saline-treated mice. Additionally, APOA4 infusion stimulated sympathetic activity in BAT and liver but not in IWAT. APOA4-treated mice had greater fatty acid oxidation but less TG content in the liver than saline-treated mice had. Plasma insulin in APOA4-treated mice was lower than that in saline-treated mice after a glucose challenge. In conclusion, continuous infusion of mouse APOA4 protein stimulated sympathetic activity in BAT and the liver, elevated BAT thermogenesis and hepatic fatty acid oxidation, and consequently attenuated levels of plasma and hepatic TG and plasma insulin without altering caloric intake, body weight gain and fat mass.


Asunto(s)
Dieta Alta en Grasa , Insulinas , Masculino , Animales , Ratones , Peso Corporal , Tejido Adiposo Pardo/metabolismo , Apolipoproteínas A , Triglicéridos/metabolismo , Metabolismo Energético , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Termogénesis , Insulinas/metabolismo , Ratones Endogámicos C57BL
3.
Int J Mol Sci ; 24(4)2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36835642

RESUMEN

Dietary lipids induce apolipoprotein A4 (APOA4) production and brown adipose tissue (BAT) thermogenesis. Administration of exogenous APOA4 elevates BAT thermogenesis in chow-fed mice, but not high-fat diet (HFD)-fed mice. Chronic feeding of HFD attenuates plasma APOA4 production and BAT thermogenesis in wildtype (WT) mice. In light of these observations, we sought to determine whether steady production of APOA4 could keep BAT thermogenesis elevated, even in the presence of HFD consumption, with an aim toward eventual reduction of body weight, fat mass and plasma lipid levels. Transgenic mice with overexpression of mouse APOA4 in the small intestine (APOA4-Tg mice) produce greater plasma APOA4 than their WT controls, even when fed an atherogenic diet. Thus, we used these mice to investigate the correlation of levels of APOA4 and BAT thermogenesis during HFD consumption. The hypothesis of this study was that overexpression of mouse APOA4 in the small intestine and increased plasma APOA4 production would increase BAT thermogenesis and consequently reduce fat mass and plasma lipids of HFD-fed obese mice. To test this hypothesis, BAT thermogenic proteins, body weight, fat mass, caloric intake, and plasma lipids in male APOA4-Tg mice and WT mice fed either a chow diet or a HFD were measured. When fed a chow diet, APOA4 levels were elevated, plasma triglyceride (TG) levels were reduced, and BAT levels of UCP1 trended upward, while body weight, fat mass, caloric intake, and plasma lipids were comparable between APOA4-Tg and WT mice. After a four-week feeding of HFD, APOA4-Tg mice maintained elevated plasma APOA4 and reduced plasma TG, but UCP1 levels in BAT were significantly elevated in comparison to WT controls; body weight, fat mass and caloric intake were still comparable. After 10-week consumption of HFD, however, while APOA4-Tg mice still exhibited increased plasma APOA4, UCP1 levels and reduced TG levels, a reduction in body weight, fat mass and levels of plasma lipids and leptin were finally observed in comparison to their WT controls and independent of caloric intake. Additionally, APOA4-Tg mice exhibited increased energy expenditure at several time points when measured during the 10-week HFD feeding. Thus, overexpression of APOA4 in the small intestine and maintenance of elevated levels of plasma APOA4 appear to correlate with elevation of UCP1-dependent BAT thermogenesis and subsequent protection against HFD-induced obesity in mice.


Asunto(s)
Tejido Adiposo Pardo , Obesidad , Ratones , Masculino , Animales , Tejido Adiposo Pardo/metabolismo , Ratones Transgénicos , Obesidad/metabolismo , Grasas de la Dieta/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Termogénesis , Ratones Endogámicos C57BL , Proteína Desacopladora 1/metabolismo
4.
Int J Mol Sci ; 22(3)2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33513710

RESUMEN

Stimulation of thermogenesis in brown adipose tissue (BAT) could have far-reaching health benefits in combatting obesity and obesity-related complications. Apolipoprotein A-IV (ApoA-IV), produced by the gut and the brain in the presence of dietary lipids, is a well-known short-term satiating protein. While our previous studies have demonstrated reduced diet-induced thermogenesis in ApoA-IV-deficient mice, it is unclear whether this reduction is due to a loss of peripheral or central effects of ApoA-IV. We hypothesized that central administration of ApoA-IV stimulates BAT thermogenesis and that sympathetic and sensory innervation is necessary for this action. To test this hypothesis, mice with unilateral denervation of interscapular BAT received central injections of recombinant ApoA-IV protein or artificial cerebrospinal fluid (CSF). The effects of central ApoA-IV on BAT temperature and thermogenesis in mice with unilateral denervation of the intrascapular BAT were monitored using transponder probe implantation, qPCR, and immunoblots. Relative to CSF, central administration of ApoA-IV significantly increased temperature and UCP expression in BAT. However, all of these effects were significantly attenuated or prevented in mice with unilateral denervation. Together, these results clearly demonstrate that ApoA-IV regulates BAT thermogenesis centrally, and this effect is mediated through sympathetic and sensory nerves.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Apolipoproteínas A/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Termogénesis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Pardo/metabolismo , Animales , Apolipoproteínas A/deficiencia , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Regulación de la Expresión Génica/genética , Lipasa/genética , Lipasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes , Tercer Ventrículo/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
5.
Front Physiol ; 11: 796, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733283

RESUMEN

Fatty meals induce intestinal secretion of chylomicrons (CMs) containing apolipoprotein (Apo) B48. These CMs travel via the lymphatic system before entering the circulation. ApoB48 is produced after post-transcriptional RNA modification by Apobec-1 editing enzyme, exclusively in the small intestine of humans and most other mammals. In contrast, in the liver where Apobec-1 editing enzyme is not expressed (except in rats and mice), the unedited transcript encodes a larger protein, ApoB100, which is used in the formation of very low-density lipoproteins (VLDL) to transport liver-synthesized fat to peripheral tissues. Apobec-1 knockout (KO) mice lack the ability to perform ApoB RNA editing, and thus, express ApoB100 in the intestine. These mice, maintained on either a chow diet or high fat diet, have body weight gain and food intake comparable to their wildtype (WT) counterparts on the respective diet; however, they secrete larger triglyceride (TG)-rich lipoprotein particles and at a slower rate than the WT mice. Using a lymph fistula model, we demonstrated that Apobec-1 KO mice also produced fewer CMs and exhibited reduced lymphatic transport of TG in response to duodenal infusion of TG at a moderate dose; in contrast, the Apobec-1 KO and WT mice had similar lymphatic transport of TG when they received a high dose of TG. Thus, the smaller, energy-saving ApoB48 appears to play a superior role in comparison with ApoB100 in the control of intestinal lipid transport in response to dietary lipid intake, at least at low to moderate lipid levels.

6.
Endocrinology ; 161(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32157301

RESUMEN

Apolipoprotein A-IV (ApoA-IV) synthesized by the gut regulates lipid metabolism. Sympathetic innervation of adipose tissues also controls lipid metabolism. We hypothesized that ApoA-IV required sympathetic innervation to increase fatty acid (FA) uptake by adipose tissues and brown adipose tissue (BAT) thermogenesis. After 3 weeks feeding of either a standard chow diet or a high-fat diet (HFD), mice with unilateral denervation of adipose tissues received intraperitoneal administration of recombinant ApoA-IV protein and intravenous infusion of lipid mixture with radioactive triolein. In chow-fed mice, ApoA-IV administration increased FA uptake by intact BAT but not the contralateral denervated BAT or intact white adipose tissue (WAT). Immunoblots showed that, in chow-fed mice, ApoA-IV increased expression of lipoprotein lipase and tyrosine hydroxylase in both intact BAT and inguinal WAT (IWAT), while ApoA-IV enhanced protein levels of ß3 adrenergic receptor, adipose triglyceride lipase, and uncoupling protein 1 in the intact BAT only. In HFD-fed mice, ApoA-IV elevated FA uptake by intact epididymal WAT (EWAT) but not intact BAT or IWAT. ApoA-IV increased sympathetic activity assessed by norepinephrine turnover (NETO) rate in BAT and EWAT of chow-fed mice, whereas it elevated NETO only in EWAT of HFD-fed mice. These observations suggest that, in chow-fed mice, ApoA-IV activates sympathetic activity of BAT and increases FA uptake by BAT via innervation, while in HFD-fed mice, ApoA-IV stimulates sympathetic activity of EWAT to shunt FAs into the EWAT.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Apolipoproteínas A/farmacología , Ácidos Grasos/metabolismo , Sistema Nervioso Simpático/metabolismo , Termogénesis/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Dieta Alta en Grasa , Masculino , Ratones , Norepinefrina/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos
7.
Biomolecules ; 9(9)2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31492038

RESUMEN

Centrally administered brain-derived neurotrophic factor (BDNF) decreases body adiposity beyond what can be accounted for by decreased food intake, implying enhanced lipid metabolism by BDNF. Consistent with this notion, intracerebroventricular (icv) injection of BDNF in rats increased the expression of lipolytic enzymes in white adipose tissues (WAT) and increased circulating concentrations of lipolytic products without changing the levels of adrenal gland hormones. This suggests that central BDNF-induced lipid mobilization is likely due to sympathetic neural activation, rather than activation of the adrenocortical or adrenomedullary system. We hypothesized that BDNF activated sympathetic innervation of adipose tissues to regulate lipolysis. Rats with unilateral denervation of interscapular brown adipose tissue (BAT) and different WAT depots received icv injections of saline or BDNF. Both intact and denervated adipose tissues were exposed to the same circulating factors, but denervated adipose tissues did not receive neural signals. Norepinephrine (NE) turnover (NETO) of BAT and WAT was assessed as a measure of sympathetic activity. Findings revealed that central BDNF treatment induced a change in NETO in some but not all the adipose tissues tested. Specifically, greater NETO rates were found in BAT and gonadal epididymal WAT (EWAT), but not in inguinal WAT (IWAT) or retroperitoneal WAT (RWAT), of BDNF-treated rats compared to saline-treated rats. Furthermore, intact innervation was necessary for BDNF-induced NETO in BAT and EWAT. In addition, BDNF increased the expression of lipolytic enzymes in both intact and denervated EWAT and IWAT, suggesting that BDNF-induced WAT lipolysis was independent of intact innervation. To summarize, centrally administered BDNF selectively provoked sympathetic drives to BAT and EWAT that was dependent on intact innervation, while BDNF also increased lipolysis in a manner independent of intact innervation.


Asunto(s)
Tejido Adiposo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Lipólisis , Masculino , Ratas , Ratas Long-Evans
8.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261740

RESUMEN

In the presence of dietary lipids, both apolipoprotein A-IV (ApoA-IV) production and brown adipose tissue (BAT) thermogenesis are increased. The effect of dietary lipid-induced AproA-IV on BAT thermogenesis and energy expenditure remains unknown. In the present study, we hypothesized that ApoA-IV knockout (ApoA-IV-KO) mice exhibited decreased BAT thermogenesis to affect energy homeostasis. To test this hypothesis, BAT thermogenesis in wildtype (WT) and ApoA-IV-KO mice fed either a standard low-fat chow diet or a high-fat diet (HFD) was investigated. When fed a chow diet, energy expenditure and food intake were comparable between WT and ApoA-IV-KO mice. After 1 week of HFD consumption, ApoA-IV-KO mice had comparable energy intake but produced lower energy expenditure relative to their WT controls in the dark phase. After an acute feeding of dietary lipids or 1-week HFD feeding, ApoA-IV-KO mice produced lower levels of uncoupling protein 1 (UCP1) and exhibited reduced expression of thermogenic genes in the BAT compared with WT controls. In response to cold exposure, however, ApoA-IV-KO mice had comparable energy expenditure and BAT temperature relative to WT mice. Thus, ApoA-IV-KO mice exhibited reduced diet-induced BAT thermogenesis and energy expenditure.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Apolipoproteínas A/genética , Dieta Alta en Grasa , Termogénesis , Tejido Adiposo Pardo/fisiología , Animales , Apolipoproteínas A/deficiencia , Grasas de la Dieta/metabolismo , Metabolismo Energético , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Desacopladora 1/metabolismo
9.
Physiol Behav ; 188: 11-17, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378187

RESUMEN

Cholecystokinin (CCK) and apolipoprotein A-IV (ApoA-IV) are gastrointestinal peptides that play an important role in controlling energy homeostasis. Lymphatic ApoA-IV and plasma CCK secretion are mediated via a chylomicron formation-dependent pathway during a dietary lipid infusion. Given their similar roles as satiating proteins, the present study examines how the two peptides interact in their function. Specifically, this study sought to understand how ApoA-IV regulates CCK secretion. For this purpose, Cck gene expression in the small intestines of ApoA-IV knockout (ApoA-IV-KO) and wild-type (WT) mice were compared under an array of feeding conditions. When fed with a chow or high-fat diet (HFD), basal levels of Cck transcripts were significantly reduced in the duodenum of ApoA-IV-KO mice compared to WT mice. Furthermore, after an oral gavage of a lipid mixture, Cck gene expression in the duodenum was significantly reduced in ApoA-IV-KO mice relative to the change seen in WT mice. To determine the mechanism by which ApoA-IV modulates Cck gene expression, STC-1 cells were transfected with predesigned mouse lysophosphatidic acid receptor 5 (LPAR5) small interfering RNA (siRNA) to knockdown Lpar5 gene expression. In this in-vitro study, mouse recombinant ApoA-IV protein increased Cck gene expression in enteroendocrine STC-1 cells and stimulated CCK release from the STC-1 cells. However, the levels of CCK protein and Cck expression were attenuated when Lpar5 was knocked down in the STC-1 cells. Together these observations suggest that dietary lipid-induced ApoA-IV is associated with Cck synthesis in the duodenum and that ApoA-IV protein directly enhances CCK release through the activation of a LPAR5-dependent pathway.


Asunto(s)
Antioxidantes/farmacología , Apolipoproteínas A/farmacología , Colecistoquinina/metabolismo , Duodeno/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Apolipoproteínas A/deficiencia , Apolipoproteínas A/genética , Línea Celular Transformada , Colecistoquinina/genética , Grasas de la Dieta/administración & dosificación , Relación Dosis-Respuesta a Droga , Duodeno/metabolismo , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Factores de Tiempo , Triglicéridos/sangre
10.
Biochem Biophys Res Commun ; 494(1-2): 152-157, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29037812

RESUMEN

Apolipoprotein A-IV (apoA-IV) is a satiation factor that acts in the hypothalamus, however, the intracellular mechanisms responsible for this action are still largely unknown. Here we report that apoA-IV treatment elicited a rapid activation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway in cultured primary hypothalamic neurons, and this effect was significantly attenuated by pretreatment with LY294002, an inhibitor of the PI3K pathway. To determine if the activation of PI3K is required for apoA-IV's inhibitory effect on food intake, apoA-IV was administered intracerebroventricularly. We found that apoA-IV significantly reduced food intake and activated PI3K signaling in the hypothalamus, and these effects were abolished by icv pre-treatment with LY294002. To identify the distinct brain sites where apoA-IV exerts its anorectic action, apoA-IV was administered into the ventromedial hypothalamus (VMH) through implanted bilateral cannula. At a low dose (0.5 µg), apoA-IV significantly inhibited food intake and activated PI3K signaling pathway in the VMH of lean rats, but not in high-fat diet-induced obese (DIO) rats. These results collectively demonstrate a critical role of the PI3K/Akt pathway in apoA-IV's anorectic action in lean rats and suggest a defective PI3K pathway in the VMH is responsible for the impaired apoA-IV's anorectic action in the DIO animals.


Asunto(s)
Apolipoproteínas A/metabolismo , Depresores del Apetito/metabolismo , Hipotálamo/metabolismo , Animales , Apolipoproteínas A/administración & dosificación , Depresores del Apetito/administración & dosificación , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos
11.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R535-R548, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768657

RESUMEN

Apolipoprotein AIV (ApoAIV) and cholecystokinin (CCK) are well-known satiating signals that are stimulated by fat consumption. Peripheral ApoAIV and CCK interact to prolong satiating signals. In the present study, we hypothesized that ApoAIV and CCK control energy homeostasis in response to high-fat diet feeding. To test this hypothesis, energy homeostasis in ApoAIV and CCK double knockout (ApoAIV/CCK-KO), ApoAIV knockout (ApoAIV-KO), and CCK knockout (CCK-KO) mice were monitored. When animals were maintained on a low-fat diet, ApoAIV/CCK-KO, ApoAIV-KO, and CCK-KO mice had comparable energy intake and expenditure, body weight, fat mass, fat absorption, and plasma parameters relative to the controls. In contrast, these KO mice exhibited impaired lipid transport to epididymal fat pads in response to intraduodenal infusion of dietary lipids. Furthermore, ApoAIV-KO mice had upregulated levels of CCK receptor 2 (CCK2R) in the small intestine while ApoAIV/CCK-KO mice had upregulated levels of CCK2R in the brown adipose tissue. After 20 wk of a high-fat diet, ApoAIV-KO and CCK-KO mice had comparable body weight and fat mass, as well as lower energy expenditure at some time points. However, ApoAIV/CCK-KO mice exhibited reduced body weight and adiposity relative to wild-type mice, despite having normal food intake. Furthermore, ApoAIV/CCK-KO mice displayed normal fat absorption and locomotor activity, as well as enhanced energy expenditure. These observations suggest that mice lacking ApoAIV and CCK have reduced body weight and adiposity, possibly due to impaired lipid transport and elevated energy expenditure.


Asunto(s)
Apolipoproteínas A/metabolismo , Colecistoquinina/metabolismo , Grasas de la Dieta/metabolismo , Homeostasis/fisiología , Adiposidad/genética , Adiposidad/fisiología , Animales , Apolipoproteínas A/deficiencia , Peso Corporal/fisiología , Colecistoquinina/deficiencia , Dieta con Restricción de Grasas/métodos , Ingestión de Alimentos/fisiología , Ingestión de Energía/fisiología , Metabolismo Energético/genética , Ratones Noqueados
12.
Physiol Rep ; 3(9)2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26359241

RESUMEN

Although ginseng has been reported to ameliorate hyperglycemia in animal models and clinical studies, the molecular mechanisms are largely unknown. We previously reported that chronic treatment with ginsenoside Rb1 (Rb1), a major component of ginseng, significantly reduced fasting glucose and improved glucose tolerance in high-fat diet (HFD)-induced obese rats. These effects were greater than those observed in pair-fed rats, suggesting a direct effect of Rb1 on glucose homeostasis, and this possibility was confirmed in the present study. In lean rats fed standard rodent chow, 5-day treatment with Rb1 significantly improved glucose tolerance and enhanced insulin sensitivity. Notably, those effects were not accompanied by reduced food intake or changed body weight. To elucidate the underlying molecular mechanisms, rats fed a HFD for 4 weeks were treated with Rb1 for 5 days. Subsequently, euglycemic-hyperinsulinemic clamp studies found that compared to vehicle, Rb1, while not changing food intake or body weight, significantly increased glucose infusion rate required to maintain euglycemia. Consistent with this, insulin-induced inhibition of hepatic gluconeogenesis was significantly enhanced and hepatic phosphoenolpyruvate carboxykinase and glucose-6-phosphatase gene expression was suppressed. Additionally, glucose uptake was significantly increased in skeletal muscle. While proximal insulin signaling was not changed after Rb1 treatment, increased phosphorylation of TBC1D4, a downstream target of AMPK signaling, appears to be a key part of the mechanism for Rb1-stimulated glucose uptake in skeletal muscle. These findings indicate that Rb1 has multiple effects on glucose homeostasis, and provide strong rationale for further evaluation of its potential therapeutic role.

13.
Physiol Behav ; 152(Pt A): 62-7, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26384952

RESUMEN

Ginsenoside Rb1 (Rb1) reduces food intake in both lean and high-fat diet induced-obese rats; however, the sites and/or mediation of the eating-suppressive effect of Rb1 have not previously been identified. We hypothesized that intraperitoneally (ip) administered Rb1 exerts its anorectic action by enhancing sensitivity to satiation signals, such as cholecystokinin (CCK), and/or that it acts through vagal afferent nerves that relay the satiating signaling to the hindbrain. To test these hypotheses, we gave ip bolus doses of Rb1 (2.5-10.0mg/kg) and CCK-8 (0.125-4.0µg/kg) alone or in combination and assessed food intake in rats. Low doses of Rb1 (2.5mg/kg) or CCK-8 (0.125µg/kg) alone had no effect on food intake whereas higher doses did. When these subthreshold doses of Rb1 and CCK-8 were co-administered, the combination significantly reduced food intake relative to saline controls, and this effect was attenuated by lorglumide, a selective CCK1-receptor antagonist. Interestingly, lorglumide blocked food intake induced by an effective dose of CCK-8 alone, but not by Rb1 alone, suggesting that Rb1's anorectic effect is independent of the CCK1 receptor. To determine whether peripherally administered Rb1 suppresses feeding via abdominal vagal nerves, we evaluated the effect of ip Rb1 injection in subdiaphragmatic vagal deafferentation (SDA) and control rats. Rb1's effect on food intake was significantly attenuated in SDA rats, compared with that in SHAM controls. These data indicate that the vagal afferent system is the major pathway conveying peripherally administered Rb1's satiation signal.


Asunto(s)
Depresores del Apetito/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Ginsenósidos/administración & dosificación , Neuronas Aferentes/efectos de los fármacos , Sincalida/administración & dosificación , Nervio Vago/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/fisiología , Glucosa , Antagonistas de Hormonas/farmacología , Inyecciones Intraperitoneales , Masculino , Neuronas Aferentes/citología , Neuronas Aferentes/fisiología , Proglumida/análogos & derivados , Proglumida/farmacología , Ratas Long-Evans , Receptor de Colecistoquinina A/antagonistas & inhibidores , Receptor de Colecistoquinina A/metabolismo , Saciedad/efectos de los fármacos , Saciedad/fisiología , Nervio Vago/citología , Nervio Vago/fisiología
14.
Physiol Behav ; 151: 198-206, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26171590

RESUMEN

Cholecystokinin (CCK) is released in response to lipid feeding and regulates pancreatic digestive enzymes vital to the absorption of nutrients. Our previous reports demonstrated that cholecystokinin knockout (CCK-KO) mice fed for 10 weeks of HFD had reduced body fat mass, but comparable glucose uptake by white adipose tissues and skeletal muscles. We hypothesized that CCK is involved in energy homeostasis and lipid transport from the small intestine to tissues in response to acute treatment with dietary lipids. CCK-KO mice with comparable fat absorption had increased energy expenditure and were resistant to HFD-induced obesity. Using intraduodenal infusion of butter fat and intravenous infusion using Liposyn III, we determined the mechanism of lipid transport from the small intestine to deposition in lymph and adipocytes in CCK-KO mice. CCK-KO mice had delayed secretion of Apo B48-chylomicrons, lipid transport to the lymphatic system, and triglyceride (TG)-derived fatty acid uptake by epididymal fat in response to acute treatment of intraduodenal lipids. In contrast, CCK-KO mice had comparable TG clearance and lipid uptake by white adipocytes in response to TGs in chylomicron-like emulsion. Thus, we concluded that CCK is important for lipid transport and energy expenditure to control body weight in response to dietary lipid feeding.


Asunto(s)
Colecistoquinina/metabolismo , Grasas de la Dieta/metabolismo , Metabolismo de los Lípidos/fisiología , Adipocitos/metabolismo , Tejido Adiposo/fisiología , Animales , Apolipoproteína B-48/metabolismo , Peso Corporal/fisiología , Colecistoquinina/genética , Quilomicrones/metabolismo , Dieta Alta en Grasa , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Intestino Delgado/metabolismo , Hígado/anatomía & histología , Hígado/fisiología , Sistema Linfático/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos
15.
Endocrinology ; 155(5): 1700-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24564397

RESUMEN

Apolipoprotein AIV (Apo AIV) and cholecystokinin (CCK) are secreted in response to fat consumption, and both cause satiation via CCK 1 receptor (CCK-1R)-containing vagal afferent nerves to the nucleus of the solitary tract (NTS), where Apo AIV is also synthesized. Fasted male Long-Evans rats received ip CCK-8 or fourth-ventricular (i4vt) Apo AIV alone or in combination. Food intake and c-Fos proteins (a product of the c-Fos immediate-early gene) were assessed. i4vt Apo AIV and/or ip CCK at effective doses reduced food intake and activated c-Fos proteins in the NTS and hypothalamic arcuate nucleus and paraventricular nucleus. Blockade of the CCK-1R by i4vt lorglumide adjacent to the NTS attenuated the satiating and c-Fos-stimulating effects of CCK and Apo AIV, alone or in combination. Maintenance on a high-fat diet (HFD) for 10 weeks resulted in weight gain and attenuation of both the behavioral and c-Fos responses to a greater extent than occurred in low-fat diet-fed and pair-fed HFD animals. These observations suggest that NTS Apo AIV or/and peripheral CCK requires vagal CCK-1R signaling to elicit satiation and that maintenance on a HFD reduces the satiating capacity of these 2 signals.


Asunto(s)
Apolipoproteínas A/metabolismo , Regulación del Apetito , Colecistoquinina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas Aferentes/metabolismo , Receptor de Colecistoquinina A/metabolismo , Núcleo Solitario/metabolismo , Animales , Apolipoproteínas A/administración & dosificación , Apolipoproteínas A/genética , Apolipoproteínas A/farmacología , Depresores del Apetito/administración & dosificación , Depresores del Apetito/farmacología , Depresores del Apetito/uso terapéutico , Regulación del Apetito/efectos de los fármacos , Estimulantes del Apetito/administración & dosificación , Estimulantes del Apetito/farmacología , Conducta Apetitiva/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Colecistoquinina/análogos & derivados , Colecistoquinina/antagonistas & inhibidores , Dieta Alta en Grasa/efectos adversos , Antagonistas de Hormonas/administración & dosificación , Antagonistas de Hormonas/farmacología , Infusiones Intraventriculares , Inyecciones Intraperitoneales , Masculino , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas Aferentes/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/metabolismo , Ratas , Ratas Long-Evans , Receptor de Colecistoquinina A/agonistas , Receptor de Colecistoquinina A/antagonistas & inhibidores , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Sincalida/administración & dosificación , Sincalida/análogos & derivados , Sincalida/farmacología , Núcleo Solitario/efectos de los fármacos
16.
Endocrinology ; 153(12): 5857-65, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23027805

RESUMEN

Apolipoprotein AIV (apo AIV) and cholecystokinin (CCK) are gastrointestinal satiation signals that are stimulated by fat consumption. Previous studies have demonstrated that peripheral apo AIV cannot cross the blood-brain barrier. In the present study, we hypothesized that peripheral apo AIV uses a CCK-dependent system and intact vagal nerves to relay its satiation signal to the hindbrain. To test this hypothesis, CCK-knockout (CCK-KO) mice and Long-Evan rats that had undergone subdiaphragmatic vagal deafferentation (SDA) were used. Intraperitoneal administration of apo AIV at 100 or 200 µg/kg suppressed food intake of wild-type (WT) mice at 30, 60, and 90 min. In contrast, the same dose did not reduce food intake in the CCK-KO mice. Blockade of the CCK 1 receptor by lorglumide, a CCK 1 receptor antagonist, attenuated apo AIV-induced satiation. Apo AIV at 100 µg/kg reduced food intake in SHAM rats but not in SDA rats. Furthermore, apo AIV elicited an increase in c-Fos-positive cells in the nucleus of the solitary tract (NTS), area postrema, dorsal motor nucleus of the vagus, and adjacent areas of WT mice but elicited only an attenuated increase in these same regions in CCK-KO mice. Apo AIV-induced c-Fos positive cells in the NTS and area postrema of WT mice were reduced by lorglumide. Lastly, apo AIV increased c-Fos positive cells in the NTS of SHAM rats but not in SDA rats. These observations imply that peripheral apo AIV requires an intact CCK system and vagal afferents to activate neurons in the hindbrain to reduce food intake.


Asunto(s)
Apolipoproteínas A/metabolismo , Colecistoquinina/metabolismo , Rombencéfalo/metabolismo , Nervio Vago/metabolismo , Animales , Ingestión de Alimentos , Conducta Alimentaria , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Long-Evans , Factores de Tiempo
17.
Am J Physiol Gastrointest Liver Physiol ; 302(11): G1336-42, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22461023

RESUMEN

Apolipoprotein AIV (apo AIV) and cholecystokinin (CCK) are satiation factors secreted by the small intestine in response to lipid meals. Apo AIV and CCK-8 has an additive effect to suppress food intake relative to apo AIV or CCK-8 alone. In this study, we determined whether CCK-8 (1, 3, or 5 µg/kg ip) reduces food intake in fasted apo AIV knockout (KO) mice as effectively as in fasted wild-type (WT) mice. Food intake was monitored by the DietMax food system. Apo AIV KO mice had significantly reduced 30-min food intake following all doses of CCK-8, whereas WT mice had reduced food intake only at doses of 3 µg/kg and above. Post hoc analysis revealed that the reduction of 10-min and 30-min food intake elicited by each dose of CCK-8 was significantly larger in the apo AIV KO mice than in the WT mice. Peripheral CCK 1 receptor (CCK1R) gene expression (mRNA) in the duodenum and gallbladder of the fasted apo AIV KO mice was comparable to that in WT mice. In contrast, CCK1R mRNA in nodose ganglia of the apo AIV KO mice was upregulated relative to WT animals. Similarly, upregulated CCK1R gene expression was found in the brain stem of apo AIV KO mice by in situ hybridization. Although it is possible that the increased satiating potency of CCK in apo AIV KO mice is mediated by upregulation of CCK 1R in the nodose ganglia and nucleus tractus solitarius, additional experiments are required to confirm such a mechanism.


Asunto(s)
Apolipoproteínas A/metabolismo , Colecistoquinina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Saciedad/efectos de los fármacos , Animales , Apolipoproteínas A/genética , Duodeno/metabolismo , Ingestión de Alimentos/genética , Conducta Alimentaria/fisiología , Vesícula Biliar/metabolismo , Masculino , Ratones , Ratones Noqueados , Ganglio Nudoso/metabolismo , Receptores de Colecistoquinina/genética , Receptores de Colecistoquinina/metabolismo , Saciedad/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...