Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 326(5): E626-E639, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38536037

RESUMEN

Loss of ovarian function imparts increased susceptibility to obesity and metabolic disease. These effects are largely attributed to decreased estradiol (E2), but the role of increased follicle-stimulating hormone (FSH) in modulating energy balance has not been fully investigated. Previous work that blocked FSH binding to its receptor in mice suggested this hormone may play a part in modulating body weight and energy expenditure after ovariectomy (OVX). We used an alternate approach to isolate the individual and combined contributions of FSH and E2 in mediating energy imbalance and changes in tissue-level metabolic health. Female Wistar rats were ovariectomized and given the gonadotropin releasing hormone (GnRH) antagonist degarelix to suppress FSH production. E2 and FSH were then added back individually and in combination for a period of 3 wk. Energy balance, body mass composition, and transcriptomic profiles of individual tissues were obtained. In contrast to previous studies, suppression and replacement of FSH in our paradigm had no effect on body weight, body composition, food intake, or energy expenditure. We did, however, observe organ-specific effects of FSH that produced unique transcriptomic signatures of FSH in retroperitoneal white adipose tissue. These included reductions in biological processes related to lipogenesis and carbohydrate transport. In addition, rats administered FSH had reduced liver triglyceride concentration (P < 0.001), which correlated with FSH-induced changes at the transcriptomic level. Although not appearing to modulate energy balance after loss of ovarian function in rats, FSH may still impart tissue-specific effects in the liver and white adipose tissue that might affect the metabolic health of those organs.NEW & NOTEWORTHY We find no effect of follicle-stimulating hormone (FSH) on energy balance using a novel model in which rats are ovariectomized, subjected to gonadotropin-releasing hormone antagonism, and systematically given back FSH by osmotic pump. However, tissue-specific effects of FSH on adipose tissue and liver were observed in this study. These include unique transcriptomic signatures induced by the hormone and a stark reduction in hepatic triglyceride accumulation.


Asunto(s)
Metabolismo Energético , Estradiol , Hormona Folículo Estimulante , Ovariectomía , Ratas Wistar , Animales , Femenino , Metabolismo Energético/efectos de los fármacos , Ratas , Hormona Folículo Estimulante/metabolismo , Estradiol/farmacología , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos , Transcriptoma/efectos de los fármacos
2.
Pediatr Res ; 95(3): 647-659, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37935884

RESUMEN

BACKGROUND: Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS: FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS: Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION: FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT: Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.


Asunto(s)
Diabetes Mellitus Tipo 2 , Retardo del Crecimiento Fetal , Humanos , Femenino , Adulto , Masculino , Lactante , Retardo del Crecimiento Fetal/metabolismo , Dieta Alta en Grasa , Aumento de Peso , Glucosa , Desarrollo Fetal
3.
J Nutr ; 153(4): 988-998, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37061344

RESUMEN

BACKGROUND: Sestrins (SESN1-3) act as proximal sensors in leucine-induced activation of the protein kinase mechanistic target of rapamycin (mTOR) in complex 1 (mTORC1), a key regulator of cell growth and metabolism. OBJECTIVE: In the present study, the hypothesis that SESNs also mediate glucose-induced activation of mTORC1 was tested. METHODS: Rats underwent overnight fasting, and in the morning, either saline or a glucose solution (4 g⋅kg-1 BW/10 mL⋅kg-1) was administered by oral gavage; mTORC1 activation in the tibialis anterior muscle was assessed. To further assess the mechanism through which glucose promotes mTORC1 activation, wild-type (WT) HEK293T and HEK293T cells lacking either all 3 SESNs (SESNTKO) or hexokinase 2 (HK2KO) were deprived of glucose, followed by glucose addback, and mTORC1 activation was assessed. In addition, glucose-induced changes in the association of the SESNs with components of the GAP activity toward the Rags (GATOR2) complex and with hexokinase 2 (HK2) were assessed by co-immunoprecipitation. One- and two-way ANOVA with Tukey post hoc comparisons were used. RESULTS: Glucose administration to fasted rats promoted mTORC1 activation. Similarly, glucose readdition (GluAB) to the medium of glucose-deprived WT cells also promoted mTORC1 activation. By contrast, SESNTKO cells demonstrated attenuated mTORC1 activation following GluAB compared with WT cells. Interestingly, HK2 associated with all 3 SESNs in a glucose-dependent manner, i.e., HK2 abundance in SESN immunoprecipitates was high in cells deprived of glucose and decreased in response to GluAB. Moreover, similar to SESNTKO cells, the sensitivity of mTORC1 to GluAB was attenuated in HK2KO cells compared with WT cells. CONCLUSIONS: The results of this study demonstrate that the SESNs and HK2 play important roles in glucose-induced mTORC1 activation in HEK293T cells. However, unlike leucine-induced mTORC1 activation, the effect was independent of the changes in SESN-GATOR2 interaction, and instead, it was associated with alterations in the association of SESNs with HK2.


Asunto(s)
Transducción de Señal , Serina-Treonina Quinasas TOR , Ratas , Animales , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células HEK293 , Serina-Treonina Quinasas TOR/metabolismo , Leucina/farmacología , Sestrinas/metabolismo , Hexoquinasa/metabolismo , Hexoquinasa/farmacología , Glucosa/farmacología
4.
Breast Cancer Res ; 24(1): 42, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35725493

RESUMEN

BACKGROUND: Obesity and adult weight gain are linked to increased breast cancer risk and poorer clinical outcomes in postmenopausal women, particularly for hormone-dependent tumors. Menopause is a time when significant weight gain occurs in many women, and clinical and preclinical studies have identified menopause (or ovariectomy) as a period of vulnerability for breast cancer development and promotion. METHODS: We hypothesized that preventing weight gain after ovariectomy (OVX) may be sufficient to prevent the formation of new tumors and decrease growth of existing mammary tumors. We tested this hypothesis in a rat model of obesity and carcinogen-induced postmenopausal mammary cancer and validated our findings in a murine xenograft model with implanted human tumors. RESULTS: In both models, preventing weight gain after OVX significantly decreased obesity-associated tumor development and growth. Importantly, we did not induce weight loss in these animals, but simply prevented weight gain. In both lean and obese rats, preventing weight gain reduced visceral fat accumulation and associated insulin resistance. Similarly, the intervention decreased circulating tumor-promoting growth factors and inflammatory cytokines (i.e., BDNF, TNFα, FGF-2), with greater effects in obese compared to lean rats. In obese rats, preventing weight gain decreased adipocyte size, adipose tissue macrophage infiltration, reduced expression of the tumor-promoting growth factor FGF-1 in mammary adipose, and reduced phosphorylated FGFR indicating reduced FGF signaling in tumors. CONCLUSIONS: Together, these findings suggest that the underlying mechanisms associated with the anti-tumor effects of weight maintenance are multi-factorial, and that weight maintenance during the peri-/postmenopausal period may be a viable strategy for reducing obesity-associated breast cancer risk and progression in women.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/prevención & control , Femenino , Humanos , Ratones , Obesidad/complicaciones , Obesidad/metabolismo , Ovariectomía , Posmenopausia , Ratas , Roedores , Carga Tumoral , Aumento de Peso
5.
Proc Natl Acad Sci U S A ; 115(12): 3138-3143, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507217

RESUMEN

Dietary guidelines for obesity typically focus on three food groups (carbohydrates, fat, and protein) and caloric restriction. Intake of noncaloric nutrients, such as salt, are rarely discussed. However, recently high salt intake has been reported to predict the development of obesity and insulin resistance. The mechanism for this effect is unknown. Here we show that high intake of salt activates the aldose reductase-fructokinase pathway in the liver and hypothalamus, leading to endogenous fructose production with the development of leptin resistance and hyperphagia that cause obesity, insulin resistance, and fatty liver. A high-salt diet was also found to predict the development of diabetes and nonalcoholic fatty liver disease in a healthy population. These studies provide insights into the pathogenesis of obesity and diabetes and raise the potential for reduction in salt intake as an additional interventional approach for reducing the risk for developing obesity and metabolic syndrome.


Asunto(s)
Fructosa/metabolismo , Leptina/sangre , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Obesidad/inducido químicamente , Cloruro de Sodio Dietético/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Diabetes Mellitus/inducido químicamente , Fructoquinasas/genética , Humanos , Leptina/genética , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/metabolismo , Sacarosa/efectos adversos , Sacarosa/análogos & derivados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
J Mammary Gland Biol Neoplasia ; 25(4): 367-387, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33216249

RESUMEN

Cells in human milk are an untapped source, as potential "liquid breast biopsies", of material for investigating lactation physiology in a non-invasive manner. We used single cell RNA sequencing (scRNA-seq) to identify milk-derived mammary epithelial cells (MECs) and their transcriptional signatures in women with diet-controlled gestational diabetes (GDM) with normal lactation. Methodology is described for coordinating milk collections with single cell capture and library preparation via cryopreservation, in addition to scRNA-seq data processing and analyses of MEC transcriptional signatures. We comprehensively characterized 3740 cells from milk samples from two mothers at two weeks postpartum. Most cells (>90%) were luminal MECs (luMECs) expressing lactalbumin alpha and casein beta and positive for keratin 8 and keratin 18. Few cells were keratin 14+ basal MECs and a small immune cell population was present (<10%). Analysis of differential gene expression among clusters identified six potentially distinct luMEC subpopulation signatures, suggesting the potential for subtle functional differences among luMECs, and included one cluster that was positive for both progenitor markers and mature milk transcripts. No expression of pluripotency markers POU class 5 homeobox 1 (POU5F1, encoding OCT4) SRY-box transcription factor 2 (SOX2) or nanog homeobox (NANOG), was observed. These observations were supported by flow cytometric analysis of MECs from mature milk samples from three women with diet-controlled GDM (2-8 mo postpartum), indicating a negligible basal/stem cell population (epithelial cell adhesion molecule (EPCAM)-/integrin subunit alpha 6 (CD49f)+, 0.07%) and a small progenitor population (EPCAM+/CD49f+, 1.1%). We provide a computational framework for others and future studies, as well as report the first milk-derived cells to be analyzed by scRNA-seq. We discuss the clinical potential and current limitations of using milk-derived cells as material for characterizing human mammary physiology.


Asunto(s)
Biología Computacional/métodos , Diabetes Gestacional/metabolismo , Lactancia/fisiología , Glándulas Mamarias Humanas/metabolismo , Leche Humana/citología , Adulto , Diabetes Gestacional/dietoterapia , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Humanos , Glándulas Mamarias Humanas/citología , Periodo Posparto/metabolismo , Embarazo , RNA-Seq/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto , Análisis de la Célula Individual , Células Madre/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 319(2): R171-R183, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32551825

RESUMEN

Exercise is often used as a strategy for weight loss maintenance. In preclinical models, we have shown that exercise may be beneficial because it counters the biological drive to regain weight. However, our studies have demonstrated sex differences in the response to exercise in this context. In the present study, we sought to better understand why females and males exhibit different compensatory food eating behaviors in response to regular exercise. Using a forced treadmill exercise paradigm, we measured weight gain, energy expenditure, food intake in real time, and the anorectic effects of leptin. The 4-wk exercise training resulted in reduced weight gain in males and sustained weight gain in females. In male rats, exercise decreased intake, whereas it increased food intake in females. Our results suggest that the anorectic effects of leptin were not responsible for these sex differences in appetite in response to exercise. If these results translate to the human condition, they may reveal important information for the use and application of regular exercise programs.


Asunto(s)
Apetito/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Condicionamiento Físico Animal/fisiología , Animales , Ingestión de Energía/fisiología , Femenino , Masculino , Ratas
8.
Int J Mol Sci ; 21(5)2020 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-32151061

RESUMEN

BACKGROUND: Glaucoma is an optic neuropathy and involves the progressive degeneration of retinal ganglion cells (RGCs), which leads to blindness in patients. We investigated the role of the neuroprotective kynurenic acid (KYNA) in RGC death against retinal ischemia/reperfusion (I/R) injury. METHODS: We injected KYNA intravenously or intravitreally to mice. We generated a knockout mouse strain of kynurenine 3-monooxygenase (KMO), an enzyme in the kynurenine pathway that produces neurotoxic 3-hydroxykynurenine. To test the effect of mild hyperglycemia on RGC protection, we used streptozotocin (STZ) induced diabetic mice. Retinal I/R injury was induced by increasing intraocular pressure for 60 min followed by reperfusion and RGC numbers were counted in the retinal flat mounts. RESULTS: Intravenous or intravitreal administration of KYNA protected RGCs against I/R injury. The I/R injury caused a greater loss of RGCs in wild type than in KMO knockout mice. KMO knockout mice had mildly higher levels of fasting blood glucose than wild type mice. Diabetic mice showed significantly lower loss of RGCs when compared with non-diabetic mice subjected to I/R injury. CONCLUSION: Together, our study suggests that the absence of KMO protects RGCs against I/R injury, through mechanisms that likely involve higher levels of KYNA and glucose.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Modelos Animales de Enfermedad , Glaucoma/prevención & control , Ácido Quinurénico/farmacología , Quinurenina 3-Monooxigenasa/fisiología , Daño por Reperfusión/complicaciones , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Glaucoma/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
9.
Am J Physiol Endocrinol Metab ; 316(6): E1136-E1145, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30964702

RESUMEN

Liver X receptors (LXRs) are ligand-dependent transcription factors activated by cholesterol metabolites. These receptors induce a suite of target genes required for de novo synthesis of triglycerides and cholesterol transport in many tissues. Two different isoforms, LXRα and LXRß, have been well characterized in liver, adipocytes, macrophages, and intestinal epithelium among others, but their contribution to cholesterol and fatty acid efflux in the lactating mammary epithelium is poorly understood. We hypothesize that LXR regulates lipogenesis during milk fat production in lactation. Global mRNA analysis of mouse mammary epithelial cells (MECs) revealed multiple LXR/RXR targets upregulated sharply early in lactation compared with midpregnancy. LXRα is the primary isoform, and its protein levels increase throughout lactation in MECs. The LXR agonist GW3965 markedly induced several genes involved in cholesterol transport and lipogenesis and enhanced cytoplasmic lipid droplet accumulation in the HC11 MEC cell line. Importantly, in vivo pharmacological activation of LXR increased the milk cholesterol percentage and induced sterol regulatory element-binding protein 1c (Srebp1c) and ATP-binding cassette transporter a7 (Abca7) expression in MECs. Cumulatively, our findings identify LXRα as an important regulator of cholesterol incorporation into the milk through key nodes of de novo lipogenesis, suggesting a potential therapeutic target in women with difficulty initiating lactation.


Asunto(s)
Colesterol/metabolismo , Epitelio/metabolismo , Lactancia/genética , Receptores X del Hígado/genética , Glándulas Mamarias Animales/metabolismo , Leche/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Línea Celular , Femenino , Regulación de la Expresión Génica , Lactancia/metabolismo , Lipogénesis/genética , Receptores X del Hígado/metabolismo , Ratones , ARN Mensajero/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
10.
Am J Physiol Endocrinol Metab ; 316(5): E977-E986, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30912962

RESUMEN

Prevalence of obesity is exacerbated by low rates of successful long-term weight loss maintenance (WLM). In part, relapse from WLM to obesity is due to a reduction in energy expenditure (EE) that persists throughout WLM and relapse. Thus, interventions that increase EE might facilitate WLM. In obese mice that were calorically restricted to reduce body weight by ~20%, we manipulated EE throughout WLM and early relapse using intermittent cold exposure (ICE; 4°C, 90 min/day, 5 days/wk, within the last 3 h of the light cycle). EE, energy intake, and spontaneous physical activity were measured during the obese, WLM, and relapse phases. During WLM and relapse, the ICE group expended more energy during the light cycle because of cold exposure but expended less energy in the dark cycle, which led to no overall difference in total daily EE. The compensation in EE appeared to be mediated by activity, whereby the ICE group was more active during the light cycle because of cold exposure but less active during the dark cycle, which led to no overall effect on total daily activity during WLM and relapse. In brown adipose tissue of relapsing mice, the ICE group had greater mRNA expression of Dio2 and protein expression of UCP1 but lower mRNA expression of Prdm16. In summary, these findings indicate that despite robust increases in EE during cold exposures, ICE is unable to alter total daily EE during WLM or early relapse, likely due to compensatory behaviors in activity.


Asunto(s)
Mantenimiento del Peso Corporal/fisiología , Frío , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Actividad Motora/fisiología , Termogénesis/fisiología , Aumento de Peso/fisiología , Pérdida de Peso/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Ratones , Obesidad , Fotoperiodo , ARN Mensajero/metabolismo , Recurrencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Yodotironina Deyodinasa Tipo II
11.
Am J Physiol Regul Integr Comp Physiol ; 317(5): R684-R695, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31553623

RESUMEN

Exercise is a potent facilitator of long-term weight loss maintenance (WLM), whereby it decreases appetite and increases energy expenditure beyond the cost of the exercise bout. We have previously shown that exercise may amplify energy expenditure through energetically expensive nutrient deposition. Therefore, we investigated the effect of exercise on hepatic de novo lipogenesis (DNL) during WLM and relapse to obesity. Obese rats were calorically restricted with (EX) or without (SED) treadmill exercise (1 h/day, 6 days/wk, 15 m/min) to induce and maintain weight loss. After 6 wk of WLM, subsets of WLM-SED and WLM-EX rats were allowed ad libitum access to food for 1 day to promote relapse (REL). An energy gap-matched group of sedentary, relapsing rats (REL-GM) were provided a diet matched to the positive energy imbalance of the REL-EX rats. During relapse, exercise increased enrichment of hepatic DN-derived lipids and induced hepatic molecular adaptations favoring DNL compared with the gap-matched controls. In the liver, compared with both REL-SED and REL-GM rats, REL-EX rats had lower hepatic expression of genes required for cholesterol biosynthesis; greater hepatic expression of genes that mediate very low-density lipoprotein synthesis and secretion; and greater mRNA expression of Cyp27a1, which encodes an enzyme involved in the biosynthesis of bile acids. Altogether, these data provide compelling evidence that the liver has an active role in exercise-mediated potentiation of energy expenditure during early relapse.


Asunto(s)
Colesterol/biosíntesis , Metabolismo Energético , Lipogénesis , Hígado/metabolismo , Obesidad/terapia , Condicionamiento Físico Animal , Aumento de Peso , Pérdida de Peso , Animales , Ácidos y Sales Biliares/biosíntesis , Restricción Calórica , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Regulación Enzimológica de la Expresión Génica , Insulina/sangre , Lipogénesis/genética , Masculino , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Recurrencia , Carrera , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Breast Cancer Res ; 20(1): 50, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29898754

RESUMEN

BACKGROUND: Obesity and type II diabetes are linked to increased breast cancer risk in postmenopausal women. Patients treated with the antidiabetic drug metformin for diabetes or metabolic syndrome have reduced breast cancer risk, a greater pathologic complete response to neoadjuvant therapy, and improved breast cancer survival. We hypothesized that metformin may be especially effective when targeted to the menopausal transition, as this is a lifecycle window when weight gain and metabolic syndrome increase, and is also when the risk for obesity-related breast cancer increases. METHODS: Here, we used an 1-methyl-1-nitrosourea (MNU)-induced mammary tumor rat model of estrogen receptor (ER)-positive postmenopausal breast cancer to evaluate the long-term effects of metformin administration on metabolic and tumor endpoints. In this model, ovariectomy (OVX) induces rapid weight gain, and an impaired whole-body response to excess calories contributes to increased tumor glucose uptake and increased tumor proliferation. Metformin treatment was initiated in tumor-bearing animals immediately prior to OVX and maintained for the duration of the study. RESULTS: Metformin decreased the size of existing mammary tumors and inhibited new tumor formation without changing body weight or adiposity. Decreased lipid accumulation in the livers of metformin-treated animals supports the ability of metformin to improve overall metabolic health. We also found a decrease in the number of aromatase-positive, CD68-positive macrophages within the tumor microenvironment, suggesting that metformin targets the immune microenvironment in addition to improving whole-body metabolism. CONCLUSIONS: These findings suggest that peri-menopause/menopause represents a unique window of time during which metformin may be highly effective in women with established, or at high risk for developing, breast cancer.


Asunto(s)
Aromatasa/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Mamarias Animales/tratamiento farmacológico , Metformina/administración & dosificación , Animales , Mama/efectos de los fármacos , Mama/inmunología , Mama/patología , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Metilnitrosourea/toxicidad , Ovariectomía , Posmenopausia/efectos de los fármacos , Posmenopausia/genética , Posmenopausia/inmunología , Ratas , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
13.
J Neurophysiol ; 117(2): 646-654, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852737

RESUMEN

Fructose stimulates vasopressin in humans and can be generated endogenously by activation of the polyol pathway with hyperosmolarity. We hypothesized that fructose metabolism in the hypothalamus might partly control vasopressin responses after acute dehydration. Wild-type and fructokinase-knockout mice were deprived of water for 24 h. The supraoptic nucleus was evaluated for vasopressin and markers of the aldose reductase-fructokinase pathway. The posterior pituitary vasopressin and serum copeptin levels were examined. Hypothalamic explants were evaluated for vasopressin secretion in response to exogenous fructose. Water restriction increased serum and urine osmolality and serum copeptin in both groups of mice, although the increase in copeptin in wild-type mice was larger than that in fructokinase-knockout mice. Water-restricted, wild-type mice showed an increase in vasopressin and aldose reductase mRNA, sorbitol, fructose and uric acid in the supraoptic nucleus. In contrast, fructokinase-knockout mice showed no change in vasopressin or aldose reductase mRNA, and no changes in sorbitol or uric acid, although fructose levels increased. With water restriction, vasopressin in the pituitary of wild-type mice was significantly less than that of fructokinase-knockout mice, indicating that fructokinase-driven vasopressin secretion overrode synthesis. Fructose increased vasopressin release in hypothalamic explants that was not observed in fructokinase-knockout mice. In situ hybridization documented fructokinase mRNA in the supraoptic nucleus, paraventricular nucleus and suprachiasmatic nucleus. Acute dehydration activates the aldose reductase-fructokinase pathway in the hypothalamus and partly drives the vasopressin response. Exogenous fructose increases vasopressin release in hypothalamic explants dependent on fructokinase. Nevertheless, circulating vasopressin is maintained and urinary concentrating is not impaired. NEW & NOTEWORTHY: This study increases our understanding of the mechanisms leading to vasopressin release under conditions of water restriction (acute dehydration). Specifically, these studies suggest that the aldose reductase-fructokinase pathways may be involved in vasopressin synthesis in the hypothalamus and secretion by the pituitary in response to acute dehydration. Nevertheless, mice undergoing water restriction remain capable of maintaining sufficient vasopressin (copeptin) levels to allow normal urinary concentration. Further studies of the aldose reductase-fructokinase system in vasopressin regulation appear indicated.


Asunto(s)
Deshidratación/fisiopatología , Fructoquinasas/deficiencia , Fructosa/farmacología , Regulación de la Expresión Génica , Hipotálamo , Vasopresinas/metabolismo , Análisis de Varianza , Animales , Ensayo de Inmunoadsorción Enzimática , Fructoquinasas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Calor/efectos adversos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Factores de Tiempo , Vasopresinas/genética , Privación de Agua
14.
J Mammary Gland Biol Neoplasia ; 21(3-4): 131-138, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27796616

RESUMEN

Accurate assessment of the long chain polyunsaturated fatty acid (LC-PUFA) content of human milk (HM) provides a powerful means to evaluate the FA nutrient status of breastfed infants. The conventional standard for FA composition analysis of HM is liquid extraction, trans-methylation, and analyte detection resolved by gas chromatography. This standard approach requires fresh or frozen samples, storage in deep freeze, organic solvents, and specialized equipment in processing and analysis. Further, HM collection is often impractical for many studies in the free living environment, particularly for studies in developing countries. In the present study, we compare a novel and more practical approach to sample collection and processing that involves the spotting and drying ~50 µL of HM on a specialized paper stored and transported at ambient temperatures until analysis. Deming regression indicated the two methods aligned very well for all LC-PUFA and the abundant HM FA. Additionally, strong correlations (r > 0.85) were observed for DHA, ARA, EPA, linoleic (LA), and alpha-linolenic acids (ALA), which are of particular interest to the health of the developing infant. Taken together, our data suggest this more practical and inexpensive method of collection, storage, and transport of HM milk samples could dramatically facilitate studies of HM, as well as understanding its lipid composition influences on human health and development.


Asunto(s)
Ácidos Grasos Insaturados/química , Ácidos Grasos Insaturados/metabolismo , Leche Humana/química , Leche Humana/metabolismo , Cromatografía de Gases/métodos , Femenino , Humanos , Extracción Líquido-Líquido/métodos , Embarazo
15.
Am J Physiol Regul Integr Comp Physiol ; 310(4): R337-45, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26661099

RESUMEN

The oxytocin (OT) and vasopressin (VP) neurons of the supraoptic nucleus (SON) demonstrate characteristics of "metabolic sensors". They express insulin receptors and glucokinase (GK). They respond to an increase in glucose and insulin with an increase in intracellular [Ca(2+)] and increased OT and VP release that is GK dependent. Although this is consistent with the established role of OT as an anorectic agent, how these molecules function relative to the important role of OT during lactation and whether deficits in this metabolic sensor function contribute to obesity remain to be examined. Thus, we evaluated whether insulin and glucose-induced OT and VP secretion from perifused explants of the hypothalamo-neurohypophyseal system are altered during lactation and by diet-induced obesity (DIO). In explants from female day 8 lactating rats, increasing glucose (Glu, 5 mM) did not alter OT or VP release. However, insulin (Ins; 3 ng/ml) increased OT release, and increasing the glucose concentration in the presence of insulin (Ins+Glu) resulted in a sustained elevation in both OT and VP release that was not prevented by alloxan, a GK inhibitor. Explants from male DIO rats also responded to Ins+Glu with an increase in OT and VP regardless of whether obesity had been induced by feeding a high-fat diet (HFD). The HFD-DIO rats had elevated body weight, plasma Ins, Glu, leptin, and triglycerides. These findings suggest that the role of SON neurons as metabolic sensors is diminished during lactation, but not in this animal model of obesity.


Asunto(s)
Grasas de la Dieta/farmacología , Lactancia/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Oxitocina/metabolismo , Núcleo Supraóptico/metabolismo , Vasopresinas/metabolismo , Aloxano/farmacología , Animales , Deshidratación/metabolismo , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Femenino , Glucoquinasa/antagonistas & inhibidores , Glucoquinasa/metabolismo , Glucosa/farmacología , Técnicas In Vitro , Insulina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Insulina/biosíntesis
16.
Proc Natl Acad Sci U S A ; 109(11): 4320-5, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371574

RESUMEN

Fructose intake from added sugars correlates with the epidemic rise in obesity, metabolic syndrome, and nonalcoholic fatty liver disease. Fructose intake also causes features of metabolic syndrome in laboratory animals and humans. The first enzyme in fructose metabolism is fructokinase, which exists as two isoforms, A and C. Here we show that fructose-induced metabolic syndrome is prevented in mice lacking both isoforms but is exacerbated in mice lacking fructokinase A. Fructokinase C is expressed primarily in liver, intestine, and kidney and has high affinity for fructose, resulting in rapid metabolism and marked ATP depletion. In contrast, fructokinase A is widely distributed, has low affinity for fructose, and has less dramatic effects on ATP levels. By reducing the amount of fructose for metabolism in the liver, fructokinase A protects against fructokinase C-mediated metabolic syndrome. These studies provide insights into the mechanisms by which fructose causes obesity and metabolic syndrome.


Asunto(s)
Fructoquinasas/metabolismo , Síndrome Metabólico/enzimología , Animales , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Fructosa/administración & dosificación , Fructosa/metabolismo , Fructosa/farmacología , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Am J Physiol Endocrinol Metab ; 307(4): E355-64, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24961240

RESUMEN

Aerobic capacity/fitness significantly impacts susceptibility for fatty liver and diabetes, but the mechanisms remain unknown. Herein, we utilized rats selectively bred for high (HCR) and low (LCR) intrinsic aerobic capacity to examine the mechanisms by which aerobic capacity impacts metabolic vulnerability for fatty liver following a 3-day high-fat diet (HFD). Indirect calorimetry assessment of energy metabolism combined with radiolabeled dietary food was employed to examine systemic metabolism in combination with ex vivo measurements of hepatic lipid oxidation. The LCR, but not HCR, displayed increased hepatic lipid accumulation in response to the HFD despite both groups increasing energy intake. However, LCR rats had a greater increase in energy intake and demonstrated greater daily weight gain and percent body fat due to HFD compared with HCR. Additionally, total energy expenditure was higher in the larger LCR. However, controlling for the difference in body weight, the LCR has lower resting energy expenditure compared with HCR. Importantly, respiratory quotient was significantly higher during the HFD in the LCR compared with HCR, suggesting reduced whole body lipid utilization in the LCR. This was confirmed by the observed lower whole body dietary fatty acid oxidation in LCR compared with HCR. Furthermore, LCR liver homogenate and isolated mitochondria showed lower complete fatty acid oxidation compared with HCR. We conclude that rats bred for low intrinsic aerobic capacity show greater susceptibility for dietary-induced hepatic steatosis, which is associated with a lower energy expenditure and reduced whole body and hepatic mitochondrial lipid oxidation.


Asunto(s)
Dieta Alta en Grasa , Tolerancia al Ejercicio/fisiología , Hígado Graso/etiología , Aptitud Física/fisiología , Animales , Células Cultivadas , Grasas de la Dieta/metabolismo , Susceptibilidad a Enfermedades , Metabolismo Energético , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Masculino , Condicionamiento Físico Animal , Ratas , Ratas Endogámicas
18.
Hepatology ; 58(5): 1632-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23813872

RESUMEN

UNLABELLED: Fructose intake from added sugars has been implicated as a cause of nonalcoholic fatty liver disease. Here we tested the hypothesis that fructose may interact with a high-fat diet to induce fatty liver, and to determine if this was dependent on a key enzyme in fructose metabolism, fructokinase. Wild-type or fructokinase knockout mice were fed a low-fat (11%), high-fat (36%), or high-fat (36%) and high-sucrose (30%) diet for 15 weeks. Both wild-type and fructokinase knockout mice developed obesity with mild hepatic steatosis and no evidence of hepatic inflammation on a high-fat diet compared to a low-fat diet. In contrast, wild-type mice fed a high-fat and high-sucrose diet developed more severe hepatic steatosis with low-grade inflammation and fibrosis, as noted by increased CD68, tumor necrosis factor alpha, monocyte chemoattractant protein-1, alpha-smooth muscle actin, and collagen I and TIMP1 expression. These changes were prevented in the fructokinase knockout mice. CONCLUSION: An additive effect of high-fat and high-sucrose diet on the development of hepatic steatosis exists. Further, the combination of sucrose with high-fat diet may induce steatohepatitis. The protection in fructokinase knockout mice suggests a key role for fructose (from sucrose) in this development of steatohepatitis. These studies emphasize the important role of fructose in the development of fatty liver and nonalcoholic steatohepatitis.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/etiología , Fructoquinasas/fisiología , Sacarosa/administración & dosificación , Animales , Ingestión de Energía , Fructosa/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Aumento de Peso
19.
J Lipid Res ; 54(5): 1346-59, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23402988

RESUMEN

The cytoplasmic lipid droplet (CLD) protein perilipin-2 (Plin2) is expressed in multiple nonadipose tissues, where it is thought to play a role in regulating their lipid storage properties. However, the extent to which Plin2 functions in nutrient utilization and metabolism, or how it influences the consequences of over-feeding, remains unclear. In this study, we demonstrate that the absence of Plin2 prevents high-fat diet(HFD)-induced obesity in male and female mice. This response is associated with increased formation of subcutaneous beige adipocyte cells with uncoupling protein 1 expression, and amelioration of inflammatory foci formation in white adipose tissue and steatosis in the liver. Experiments demonstrate that Plin2 loss results in reduced energy intake and increased physical activity in response to HFD feeding. Our study provides the first evidence that Plin2 contributes to HFD-induced obesity by modulating food intake, and that its absence prevents obesity-associated adipose tissue inflammatory foci and liver steatosis.


Asunto(s)
Hígado Graso/metabolismo , Proteínas de la Membrana/metabolismo , Obesidad/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Hígado Graso/genética , Hígado Graso/patología , Femenino , Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/patología , Perilipina-2
20.
Am J Physiol Gastrointest Liver Physiol ; 305(11): G868-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24091599

RESUMEN

Changes in substrate utilization and reduced mitochondrial respiratory capacity following exposure to energy-dense, high-fat diets (HFD) are putatively key components in the development of obesity-related metabolic disease. We examined the effect of a 3-day HFD on isolated liver mitochondrial respiration and whole body energy utilization in obesity-prone (OP) rats. We also examined if hepatic overexpression of peroxisomal proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial respiratory capacity and biogenesis, would modify liver and whole body responses to the HFD. Acute, 3-day HFD (45% kcal) in OP rats resulted in increased daily energy intake, energy balance, weight gain, and adiposity, without an increase in liver triglyceride (triacylglycerol) accumulation. HFD-fed OP rats also displayed decreased whole body substrate switching from the dark to the light cycle, which was paired with reductions in hepatic mitochondrial respiration of multiple substrates in multiple respiratory states. Hepatic PGC-1α overexpression was observed to protect whole body substrate switching, as well as maintain mitochondrial respiration, following the acute HFD. Additionally, liver PGC-1α overexpression did not alter whole body dietary fatty acid oxidation but resulted in greater storage of dietary free fatty acids in liver lipid, primarily as triacylglycerol. Together, these data demonstrate that a short-term HFD can result in a decrease in metabolic flexibility and hepatic mitochondrial respiratory capacity in OP rats that is completely prevented by hepatic overexpression of PGC-1α.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Mitocondrias Hepáticas/metabolismo , Fosforilación Oxidativa , Factores de Transcripción/metabolismo , Adiposidad , Animales , Respiración de la Célula , Ingestión de Energía , Hígado/metabolismo , Masculino , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Ratas , Ratas Wistar , Factores de Transcripción/genética , Transcripción Genética , Triglicéridos/metabolismo , Aumento de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA