Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Front Cardiovasc Med ; 9: 948332, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061565

RESUMEN

CD36 mediates the uptake of long-chain fatty acids (FAs), a major energy substrate for the myocardium. Under excessive FA supply, CD36 can cause cardiac lipid accumulation and inflammation while its deletion reduces heart FA uptake and lipid content and increases glucose utilization. As a result, CD36 was proposed as a therapeutic target for obesity-associated heart disease. However, more recent reports have shown that CD36 deficiency suppresses myocardial flexibility in fuel preference between glucose and FAs, impairing tissue energy balance, while CD36 absence in tissue macrophages reduces efferocytosis and myocardial repair after injury. In line with the latter homeostatic functions, we had previously reported that CD36-/- mice have chronic subclinical inflammation. Lipids are important for the maintenance of tissue homeostasis and there is limited information on heart lipid metabolism in CD36 deficiency. Here, we document in the hearts of unchallenged CD36-/- mice abnormalities in the metabolism of triglycerides, plasmalogens, cardiolipins, acylcarnitines, and arachidonic acid, and the altered remodeling of these lipids in response to an overnight fast. The hearts were examined for evidence of inflammation by monitoring the presence of neutrophils and pro-inflammatory monocytes/macrophages using the respective positron emission tomography (PET) tracers, 64Cu-AMD3100 and 68Ga-DOTA-ECL1i. We detected significant immune cell infiltration in unchallenged CD36-/- hearts as compared with controls and immune infiltration was also observed in hearts of mice with cardiomyocyte-specific CD36 deficiency. Together, the data show that the CD36-/- heart is in a non-homeostatic state that could compromise its stress response. Non-invasive immune cell monitoring in humans with partial or total CD36 deficiency could help evaluate the risk of impaired heart remodeling and disease.

3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1867(11): 159207, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35882297

RESUMEN

The vascular and lymphatic systems in the gut regulate lipid transport while restricting transfer of commensal gut microbiota and directing immune cell trafficking. Increased permeability of the endothelial systems in the intestine associates with passage of antigens and microbiota from the gut into the bloodstream leading to tissue inflammation, the release of pro-inflammatory mediators and ultimately to abnormalities of systemic metabolism. Recent studies show that lipid metabolism maintains homeostasis and function of intestinal blood and lymphatic endothelial cells, BECs and LECs, respectively. This review highlights recent progress in this area, and information related to the contribution of the lipid transporter CD36, abundant in BECs and LECs, to gastrointestinal barrier integrity, inflammation, and to gut regulation of whole body metabolism. The potential role of endothelial lipid delivery in epithelial tissue renewal after injury and consequently in the risk of gastric and intestinal diseases is also discussed.


Asunto(s)
Células Endoteliales , Microbioma Gastrointestinal , Células Endoteliales/metabolismo , Humanos , Inflamación/metabolismo , Lípidos , Sistema Linfático/metabolismo
4.
Nat Commun ; 12(1): 3350, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099721

RESUMEN

Disruption of lymphatic lipid transport is linked to obesity and type 2 diabetes (T2D), but regulation of lymphatic vessel function and its link to disease remain unclear. Here we show that intestinal lymphatic endothelial cells (LECs) have an increasing CD36 expression from lymphatic capillaries (lacteals) to collecting vessels, and that LEC CD36 regulates lymphatic integrity and optimizes lipid transport. Inducible deletion of CD36 in LECs in adult mice (Cd36ΔLEC) increases discontinuity of LEC VE-cadherin junctions in lacteals and collecting vessels. Cd36ΔLEC mice display slower transport of absorbed lipid, more permeable mesenteric lymphatics, accumulation of inflamed visceral fat and impaired glucose disposal. CD36 silencing in cultured LECs suppresses cell respiration, reduces VEGF-C-mediated VEGFR2/AKT phosphorylation and destabilizes VE-cadherin junctions. Thus, LEC CD36 optimizes lymphatic junctions and integrity of lymphatic lipid transport, and its loss in mice causes lymph leakage, visceral adiposity and glucose intolerance, phenotypes that increase risk of T2D.


Asunto(s)
Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Endoteliales/metabolismo , Resistencia a la Insulina/fisiología , Obesidad Abdominal/metabolismo , Animales , Antígenos CD , Cadherinas , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucosa/metabolismo , Inflamación , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Transcriptoma , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Atherosclerosis ; 329: 1-8, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34130222

RESUMEN

Lipids released from circulating lipoproteins by intravascular action of lipoprotein lipase (LpL) reach parenchymal cells in tissues with a non-fenestrated endothelium by transfer through or around endothelial cells. The actions of LpL are controlled at multiple sites, its synthesis and release by myocytes and adipocytes, its transit and association with the endothelial cell luminal surface, and finally its activation and inhibition by a number of proteins and by its product non-esterified fatty acids. Multiple pathways mediate endothelial transit of lipids into muscle and adipose tissues. These include movement of fatty acids via the endothelial cell fatty acid transporter CD36 and movement of whole or partially LpL-hydrolyzed lipoproteins via other apical endothelial cell receptors such as SR-B1and Alk1. Lipids also likely change the barrier function of the endothelium and operation of the paracellular pathway around endothelial cells. This review summarizes in vitro and in vivo support for the key role of endothelial cells in delivery of lipids and highlights incompletely understood processes that are the focus of active investigation.


Asunto(s)
Células Endoteliales , Ácidos Grasos no Esterificados , Endotelio , Ácidos Grasos , Humanos , Lipoproteína Lipasa , Lipoproteínas , Triglicéridos
6.
Gastroenterology ; 161(3): 968-981.e12, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34004161

RESUMEN

BACKGROUND AND AIMS: Insulin resistance is a key factor in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We evaluated the importance of subcutaneous abdominal adipose tissue (SAAT) inflammation and both plasma and SAAT-derived exosomes in regulating insulin sensitivity in people with obesity and NAFLD. METHODS: Adipose tissue inflammation (macrophage and T-cell content and expression of proinflammatory cytokines), liver and whole-body insulin sensitivity (assessed using a hyperinsulinemic-euglycemic clamp and glucose tracer infusion), and 24-hour serial plasma cytokine concentrations were evaluated in 3 groups stratified by adiposity and intrahepatic triglyceride (IHTG) content: (1) lean with normal IHTG content (LEAN; N = 14); (2) obese with normal IHTG content (OB-NL; N = 28); and (3) obese with NAFLD (OB-NAFLD; N = 28). The effect of plasma and SAAT-derived exosomes on insulin-stimulated Akt phosphorylation in human skeletal muscle myotubes and mouse primary hepatocytes was assessed in a subset of participants. RESULTS: Proinflammatory macrophages, proinflammatory CD4 and CD8 T-cell populations, and gene expression of several cytokines in SAAT were greater in the OB-NAFLD than the OB-NL and LEAN groups. However, with the exception of PAI-1, which was greater in the OB-NAFLD than the LEAN and OB-NL groups, 24-hour plasma cytokine concentration areas-under-the-curve were not different between groups. The percentage of proinflammatory macrophages and plasma PAI-1 concentration areas-under-the-curve were inversely correlated with both hepatic and whole-body insulin sensitivity. Compared with exosomes from OB-NL participants, plasma and SAAT-derived exosomes from the OB-NAFLD group decreased insulin signaling in myotubes and hepatocytes. CONCLUSIONS: Systemic insulin resistance in people with obesity and NAFLD is associated with increased plasma PAI-1 concentrations and both plasma and SAAT-derived exosomes. ClinicalTrials.gov number: NCT02706262 (https://clinicaltrials.gov/ct2/show/NCT02706262).


Asunto(s)
Citocinas/sangre , Exosomas/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Células T de Memoria/metabolismo , Enfermedad del Hígado Graso no Alcohólico/sangre , Obesidad/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Grasa Subcutánea Abdominal/metabolismo , Adulto , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Células Cultivadas , Exosomas/inmunología , Femenino , Hepatocitos/metabolismo , Humanos , Insulina/sangre , Hígado/metabolismo , Macrófagos/inmunología , Masculino , Células T de Memoria/inmunología , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Obesidad/diagnóstico , Obesidad/inmunología , Obesidad/fisiopatología , Grasa Subcutánea Abdominal/inmunología , Técnicas de Cultivo de Tejidos
8.
J Clin Invest ; 130(12): 6688-6699, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33164985

RESUMEN

BACKGROUNDData from studies conducted in rodent models have shown that decreased adipose tissue (AT) oxygenation is involved in the pathogenesis of obesity-induced insulin resistance. Here, we evaluated the potential influence of AT oxygenation on AT biology and insulin sensitivity in people.METHODSWe evaluated subcutaneous AT oxygen partial pressure (pO2); liver and whole-body insulin sensitivity; AT expression of genes and pathways involved in inflammation, fibrosis, and branched-chain amino acid (BCAA) catabolism; systemic markers of inflammation; and plasma BCAA concentrations, in 3 groups of participants that were rigorously stratified by adiposity and insulin sensitivity: metabolically healthy lean (MHL; n = 11), metabolically healthy obese (MHO; n = 15), and metabolically unhealthy obese (MUO; n = 20).RESULTSAT pO2 progressively declined from the MHL to the MHO to the MUO group, and was positively associated with hepatic and whole-body insulin sensitivity. AT pO2 was positively associated with the expression of genes involved in BCAA catabolism, in conjunction with an inverse relationship between AT pO2 and plasma BCAA concentrations. AT pO2 was negatively associated with AT gene expression of markers of inflammation and fibrosis. Plasma PAI-1 increased from the MHL to the MHO to the MUO group and was negatively correlated with AT pO2, whereas the plasma concentrations of other cytokines and chemokines were not different among the MHL and MUO groups.CONCLUSIONThese results support the notion that reduced AT oxygenation in individuals with obesity contributes to insulin resistance by increasing plasma PAI-1 concentrations and decreasing AT BCAA catabolism and thereby increasing plasma BCAA concentrations.TRIAL REGISTRATIONClinicalTrials.gov NCT02706262.FUNDINGThis study was supported by NIH grants K01DK109119, T32HL130357, K01DK116917, R01ES027595, P42ES010337, DK56341 (Nutrition Obesity Research Center), DK20579 (Diabetes Research Center), DK052574 (Digestive Disease Research Center), and UL1TR002345 (Clinical and Translational Science Award); NIH Shared Instrumentation Grants S10RR0227552, S10OD020025, and S10OD026929; and the Foundation for Barnes-Jewish Hospital.


Asunto(s)
Resistencia a la Insulina , Obesidad/metabolismo , Oxígeno/metabolismo , Grasa Subcutánea/metabolismo , Adulto , Aminoácidos de Cadena Ramificada/metabolismo , Biomarcadores/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Obesidad/patología , Grasa Subcutánea/patología
9.
Cell Mol Gastroenterol Hepatol ; 7(3): 503-513, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30557701

RESUMEN

The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.


Asunto(s)
Intestinos/fisiología , Sistema Linfático/metabolismo , Animales , Colesterol/metabolismo , Grasas de la Dieta/metabolismo , Humanos , Absorción Intestinal , Factores de Crecimiento Endotelial Vascular/metabolismo
10.
J Lipid Res ; 59(7): 1294-1300, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29764924

RESUMEN

Interest in measuring tissue lipids has increased as the link between fat-laden tissues and metabolic disease has become obvious; however, linking disease to a specific cell type within a tissue has been hampered by methodological limitations. Flow cytometry (FC) has been used to assess relative lipid levels in cells. Unfortunately, its usefulness is limited because comparisons between samples generated over several hours is problematic. We show that: 1) in lipophilic fluorophore stained cells, fluorescence intensity measured by FC reflects lipid levels; 2) this technique can be used to assess lipid levels in a mixed cell population; 3) normalizing to a control condition can decrease experiment-to-experiment variation; and 4) fluorescence intensity increases linearly with lipid levels. This allows triacylglycerol (TG) mass to be estimated in mixed cell populations comparing cells with known fluorescence and TG levels. We exploited this strategy to estimate lipid levels in monocytes within a mixed population of cells isolated from human blood. Using this strategy, we also confirmed that perilipin (PLIN)1 increases TG accumulation by ectopically expressing fluorescently tagged PLIN1 in Huh7 cells. In both examples, biochemically assaying for TG in specific cell populations is problematic due to limited cell numbers and isolation challenges. Other advantages are discussed.


Asunto(s)
Citometría de Flujo/métodos , Metabolismo de los Lípidos , Animales , Línea Celular , Citometría de Flujo/normas , Humanos , Ratones , Estándares de Referencia
11.
Compr Physiol ; 8(2): 493-507, 2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29687890

RESUMEN

Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.


Asunto(s)
Antígenos CD36/fisiología , Absorción Intestinal/fisiología , Metabolismo de los Lípidos/fisiología , Animales , Transporte Biológico/fisiología , Quilomicrones/biosíntesis , Sistema Digestivo/metabolismo , Enterocitos/metabolismo , Ácidos Grasos/metabolismo , Homeostasis/fisiología , Humanos , Fosfolípidos/metabolismo
12.
Front Physiol ; 9: 1783, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30618798

RESUMEN

The lymphatic system transports dietary lipids absorbed and packaged as chylomicrons by enterocytes, for delivery to the bloodstream. Once considered a passive drainage, chylomicron entry into intestinal lymphatic vessels, or lacteals, is now emerging to be an active process controlled by a dynamic and complex regulation. Vascular endothelial growth factor (VEGF)-C, a major lymphangiogenic factor, regulates lacteal maintenance and function. Little is known about the role of its cognate tyrosine kinase VEGF receptor 3 (VEGFR-3) during lipid absorption. Here we investigated role of VEGFR-3 signaling in triglyceride (TG) absorption and distribution into tissues using the Chy mouse model, which bears an inactivating mutation in the tyrosine kinase domain of VEGFR-3 (heterozygous A3157T mutation resulting in I1053F substitution). Our data show that inactivation of VEGFR-3 tyrosine kinase motif leads to retention of TGs in the enterocytes of the small intestine, decreased postprandial levels of TGs in the plasma and increased excretion of free fatty acids (FFAs) and TGs into their stools. We further show that levels of nitric oxide (NO), required for chylomicron mobilization into the bloodstream, are significantly reduced in the Chy intestine after a fat bolus suggesting a critical role for VEGFR-3 signaling in the generation of NO during lipid absorption. Our data support the hypothesis that VEGFR-3 signaling plays an important role in chylomicron-TG entry into lacteals, possibly affecting TG trafficking to peripheral tissues.

13.
J Lipid Res ; 58(8): 1692-1701, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28634191

RESUMEN

The scavenger receptor and multiligand transporter CD36 functions to promote cellular free fatty acid uptake and regulates aspects of both hepatic and intestinal cholesterol metabolism. However, the role of CD36 in regulating canalicular and biliary cholesterol transport and secretion is unknown. Here, we show that germline Cd36 knockout (KO) mice are protected against lithogenic diet (LD)-induced gallstones compared with congenic (C57BL6/J) controls. Cd36 KO mice crossed into congenic L-Fabp KO mice (DKO mice) demonstrated protection against LD-induced gallstones, reversing the susceptibility phenotype observed in L-Fabp KO mice. DKO mice demonstrated reduced biliary cholesterol secretion and a shift into more hydrophophilic bile acid species, without changes in either BA pool size or fecal excretion. In addition, we found that the mean and maximum force of gallbladder contraction was increased in germline Cd36 KO mice, and gallbladder lipid content was reduced compared with wild-type controls. Finally, whereas germline Cd36 KO mice were protected against LD-induced gallstones, neither liver- nor intestine-specific Cd36 KO mice were protected. Taken together, our findings show that CD36 plays an important role in modifying gallstone susceptibility in mice, at least in part by altering biliary lipid composition, but also by promoting gallbladder contractility.


Asunto(s)
Antígenos CD36/deficiencia , Antígenos CD36/genética , Dieta/efectos adversos , Cálculos Biliares/genética , Animales , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Vesícula Biliar/metabolismo , Vesícula Biliar/fisiopatología , Cálculos Biliares/etiología , Cálculos Biliares/metabolismo , Cálculos Biliares/fisiopatología , Técnicas de Inactivación de Genes , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/genética
14.
J Lipid Res ; 58(6): 1132-1142, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28404638

RESUMEN

Lipid accumulation is a pathological feature of every type of kidney injury. Despite this striking histological feature, physiological accumulation of lipids in the kidney is poorly understood. We studied whether the accumulation of lipids in the fasted kidney are derived from lipoproteins or NEFAs. With overnight fasting, kidneys accumulated triglyceride, but had reduced levels of ceramide and glycosphingolipid species. Fasting led to a nearly 5-fold increase in kidney uptake of plasma [14C]oleic acid. Increasing circulating NEFAs using a ß adrenergic receptor agonist caused a 15-fold greater accumulation of lipid in the kidney, while mice with reduced NEFAs due to adipose tissue deficiency of adipose triglyceride lipase had reduced triglycerides. Cluster of differentiation (Cd)36 mRNA increased 2-fold, and angiopoietin-like 4 (Angptl4), an LPL inhibitor, increased 10-fold. Fasting-induced kidney lipid accumulation was not affected by inhibition of LPL with poloxamer 407 or by use of mice with induced genetic LPL deletion. Despite the increase in CD36 expression with fasting, genetic loss of CD36 did not alter fatty acid uptake or triglyceride accumulation. Our data demonstrate that fasting-induced triglyceride accumulation in the kidney correlates with the plasma concentrations of NEFAs, but is not due to uptake of lipoprotein lipids and does not involve the fatty acid transporter, CD36.


Asunto(s)
Ayuno/sangre , Ayuno/metabolismo , Ácidos Grasos no Esterificados/sangre , Riñón/metabolismo , Triglicéridos/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
15.
Cell Mol Gastroenterol Hepatol ; 3(1): 82-98, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28066800

RESUMEN

BACKGROUND & AIMS: CD36 has immuno-metabolic actions and is abundant in the small intestine on epithelial, endothelial and immune cells. We examined the role of CD36 in gut homeostasis using mice null for CD36 (CD36KO) and with CD36 deletion specific to enterocytes (Ent-CD36KO) or endothelial cells (EC-CD36KO). METHODS: Intestinal morphology was evaluated using immunohistochemistry and electron microscopy (EM). Intestinal inflammation was determined from neutrophil infiltration and expression of cytokines, toll-like receptors and COX-2. Barrier integrity was assessed from circulating lipopolysaccharide (LPS) and dextran administered intragastrically. Epithelial permeability to luminal dextran was visualized using two photon microscopy. RESULTS: The small intestines of CD36KO mice fed a chow diet showed several abnormalities including extracellular matrix (ECM) accumulation with increased expression of ECM proteins, evidence of neutrophil infiltration, inflammation and compromised barrier function. EM showed shortened desmosomes with decreased desmocollin 2 expression. Systemically, leukocytosis and neutrophilia were present together with 80% reduction of anti-inflammatory Ly6Clow monocytes. Bone marrow transplants supported the primary contribution of non-hematopoietic cells to the inflammatory phenotype. Specific deletion of endothelial but not of enterocyte CD36 reproduced many of the gut phenotypes of germline CD36KO mice including fibronectin deposition, increased interleukin 6, neutrophil infiltration, desmosome shortening and impaired epithelial barrier function. CONCLUSIONS: CD36 loss results in chronic neutrophil infiltration of the gut, impairs barrier integrity and systemically causes subclinical inflammation. Endothelial cell CD36 deletion reproduces the major intestinal phenotypes. The findings suggest an important role of the endothelium in etiology of gut inflammation and loss of epithelial barrier integrity.

16.
J Lipid Res ; 57(4): 663-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26912395

RESUMEN

Obesity induces accumulation of adipose tissue macrophages (ATMs), which contribute to both local and systemic inflammation and modulate insulin sensitivity. Adipocyte lipolysis during fasting and weight loss also leads to ATM accumulation, but without proinflammatory activation suggesting distinct mechanisms of ATM recruitment. We examined the possibility that specific lipid mediators with anti-inflammatory properties are released from adipocytes undergoing lipolysis to induce macrophage migration. In the present study, we showed that conditioned medium (CM) from adipocytes treated with forskolin to stimulate lipolysis can induce migration of RAW 264.7 macrophages. In addition to FFAs, lipolytic stimulation increased release of prostaglandin E2(PGE2) and prostaglandin D2(PGD2), reflecting cytosolic phospholipase A2α activation and enhanced cyclooxygenase (COX) 2 expression. Reconstituted medium with the anti-inflammatory PGE2potently induced macrophage migration while different FFAs and PGD2had modest effects. The ability of CM to induce macrophage migration was abolished by treating adipocytes with the COX2 inhibitor sc236 or by treating macrophages with the prostaglandin E receptor 4 antagonist AH23848. In fasted mice, macrophage accumulation in adipose tissue coincided with increases of PGE2levels and COX1 expression. Collectively, our data show that adipocyte-originated PGE2with inflammation suppressive properties plays a significant role in mediating ATM accumulation during lipolysis.


Asunto(s)
Adipocitos/metabolismo , Quimiotaxis , Dinoprostona/metabolismo , Lipólisis , Macrófagos/citología , Células 3T3-L1 , Animales , Ácido Araquidónico/metabolismo , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Activación Enzimática , Ayuno , Regulación Enzimológica de la Expresión Génica , Fosfolipasas A2 Grupo IV/metabolismo , Ratones , Células RAW 264.7
17.
Diabetes ; 64(5): 1632-42, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25576058

RESUMEN

Metformin treatment is associated with a decreased risk and better prognosis of pancreatic cancer (PC) in patients with type 2 diabetes, but the mechanism of metformin's PC growth inhibition in the context of a prediabetic state is unknown. We used a Panc02 pancreatic tumor cell transplant model in diet-induced obese (DIO) C57BL/6 mice to compare the effects of metformin and the direct mammalian target of rapamycin (mTOR) inhibitor rapamycin on PC growth, glucose regulation, mTOR pathway signaling, and candidate microRNA (miR) expression. In DIO/prediabetic mice, metformin and rapamycin significantly reduced pancreatic tumor growth and mTOR-related signaling. The rapamycin effects centered on decreased mTOR-regulated growth and survival signaling, including increased expression of let-7b and cell cycle-regulating miRs. Metformin (but not rapamycin) reduced glucose and insulin levels and expression of miR-34a and its direct targets Notch, Slug, and Snail. Metformin also reduced the number and size of Panc02 tumor spheres in vitro and inhibited the expression of Notch in spheroids. Our results suggest that metformin and rapamycin can both inhibit pancreatic tumor growth in obese, prediabetic mice through shared and distinct mechanisms. Metformin and direct mTOR inhibitors, alone or possibly in combination, represent promising intervention strategies for breaking the diabetes-PC link.


Asunto(s)
Metformina/uso terapéutico , MicroARNs/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Estado Prediabético/tratamiento farmacológico , Sirolimus/uso terapéutico , Animales , Peso Corporal , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Dieta para Diabéticos , Ingestión de Energía , Intolerancia a la Glucosa , Hipoglucemiantes/uso terapéutico , Inmunosupresores/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , MicroARNs/genética , Neoplasias Experimentales/tratamiento farmacológico , Distribución Aleatoria , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vimentina/genética , Vimentina/metabolismo
18.
Mol Endocrinol ; 26(7): 1213-24, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22570335

RESUMEN

Chronic hyperglycemia exerts a deleterious effect on endothelium, contributing to endothelial dysfunction and microvascular complications in poorly controlled diabetes. To understand the underlying mechanism, we studied the effect of endothelin-1 (ET-1) on endothelial production of Forkhead box O1 (FOXO1), a forkhead transcription factor that plays an important role in cell survival. ET-1 is a 21-amino acid peptide that is secreted primarily from endothelium. Using adenovirus-mediated gene transfer approach, we delivered FOXO1 cDNA into cultured human aorta endothelial cells. FOXO1 was shown to stimulate B cell leukemia/lymphoma 2-associated death promoter (BAD) production and promote cellular apoptosis. This effect was counteracted by ET-1. In response to ET-1, FOXO1 was phosphorylated and translocated from the nucleus to cytoplasm, resulting in inhibition of BAD production and mitigation of FOXO1-mediated apoptosis. Hyperglycemia stimulated FOXO1 O-glycosylation and promoted its nuclear localization in human aorta endothelial cells. This effect accounted for unbridled FOXO1 activity in the nucleus, contributing to augmented BAD production and endothelial apoptosis under hyperglycemic conditions. FOXO1 expression became deregulated in the aorta of both streptozotocin-induced diabetic mice and diabetic db/db mice. This hyperglycemia-elicited FOXO1 deregulation and its ensuing effect on endothelial cell survival was corrected by ET-1. Likewise, FoxO1 deregulation in the aorta of diabetic mice was reversible after the reduction of hyperglycemia by insulin therapy. These data reveal a mechanism by which FOXO1 mediated the autocrine effect of ET-1 on endothelial cell survival. FOXO1 deregulation, resulting from an impaired ability of ET-1 to control FOXO1 activity in endothelium, may contribute to hyperglycemia-induced endothelial lesion in diabetes.


Asunto(s)
Supervivencia Celular , Células Endoteliales/fisiología , Endotelina-1/metabolismo , Factores de Transcripción Forkhead/metabolismo , Animales , Aorta , Apoptosis , Línea Celular , Diabetes Mellitus/metabolismo , Células Endoteliales/citología , Femenino , Proteína Forkhead Box O1 , Técnicas de Transferencia de Gen , Humanos , Hiperglucemia/metabolismo , Insulina/metabolismo , Insulina/farmacología , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Fosforilación , Distribución Aleatoria , Proteína Letal Asociada a bcl/biosíntesis
19.
Inflamm Res ; 61(1): 27-35, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21947415

RESUMEN

OBJECTIVE: We investigated C-peptide effects on inflammatory cytokine release and adhesion of monocytes exposed to high glucose and lipopolysaccharide (LPS) in vitro. MATERIALS AND METHODS: Monocytic cells (U-937) were cultured in the presence of 30 mmol/L glucose and stimulated with 0.5 ng/µL LPS in the presence or absence of C-peptide (1 µmol/L) for 24 h to induce inflammatory cytokine secretion. Adhesion of U-937 monocytes to human aortic endothelial cells (HAEC) was also studied in the presence or absence of C-peptide. Concentrations of IL-6, IL-8, macrophage inflammatory protein(MIP)-1α, and MIP-1ß in supernatants from LPS-stimulated U-937 monocytes were assessed by Luminex. To gain insights into potential intracellular signaling pathways affected by C-peptide, we investigated nuclear translocation of nuclear factor(NF)-κB p65/p50 subunits by western blot in LPS-treated U-937 cells. The effect of C-peptide on LPS-induced phosphorylation of the cytoplasmic protein IκB-α was also investigated by immunoblotting. RESULTS: Addition of C-peptide significantly reduced cytokine secretion from LPS-stimulated U-937 monocytes. Adhesion of U-937 cells to HAEC was also significantly reduced by C-peptide. These effects were accompanied by reduced NF-κB p65/p50 nuclear translocation and decreased phosphorylation of IκB-α. CONCLUSIONS: We conclude that, in conditions of hyperglycemia, C-peptide reduces monocytes activation via inhibition of the NF-κB pathway.


Asunto(s)
Péptido C/farmacología , Hiperglucemia/metabolismo , Lipopolisacáridos/metabolismo , Aorta/citología , Péptido C/química , Quimiocina CCL3/metabolismo , Quimiocina CCL4/metabolismo , Células Endoteliales/citología , Humanos , Inflamación , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Monocitos/citología , Fosforilación , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...