Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 165(4): 882-95, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27133169

RESUMEN

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Asunto(s)
Encéfalo/metabolismo , Dieta Alta en Grasa , Glucosa/metabolismo , Obesidad/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Cognición , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Ratones , Células Mieloides/metabolismo
2.
Nat Immunol ; 15(5): 423-30, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24681566

RESUMEN

Obesity and resistance to insulin are closely associated with the development of low-grade inflammation. Interleukin 6 (IL-6) is linked to obesity-associated inflammation; however, its role in this context remains controversial. Here we found that mice with an inactivated gene encoding the IL-6Rα chain of the receptor for IL-6 in myeloid cells (Il6ra(Δmyel) mice) developed exaggerated deterioration of glucose homeostasis during diet-induced obesity, due to enhanced resistance to insulin. Tissues targeted by insulin showed increased inflammation and a shift in macrophage polarization. IL-6 induced expression of the receptor for IL-4 and augmented the response to IL-4 in macrophages in a cell-autonomous manner. Il6ra(Δmyel) mice were resistant to IL-4-mediated alternative polarization of macrophages and exhibited enhanced susceptibility to lipopolysaccharide (LPS)-induced endotoxemia. Our results identify signaling via IL-6 as an important determinant of the alternative activation of macrophages and assign an unexpected homeostatic role to IL-6 in limiting inflammation.


Asunto(s)
Endotoxemia/inmunología , Resistencia a la Insulina , Interleucina-6/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Obesidad/inmunología , Animales , Células Cultivadas , Humanos , Resistencia a la Insulina/genética , Resistencia a la Insulina/inmunología , Interleucina-4/inmunología , Interleucina-6/genética , Lipopolisacáridos/inmunología , Activación de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Receptores de Interleucina-6/genética , Transducción de Señal/genética
4.
Annu Rev Physiol ; 83: 303-330, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33158378

RESUMEN

The global prevalence of metabolic diseases such as type 2 diabetes mellitus, steatohepatitis, myocardial infarction, and stroke has increased dramatically over the past two decades. These obesity-fueled disorders result, in part, from the aberrant accumulation of harmful lipid metabolites in tissues not suited for lipid storage (e.g., the liver, vasculature, heart, and pancreatic beta-cells). Among the numerous lipid subtypes that accumulate, sphingolipids such as ceramides are particularly impactful, as they elicit the selective insulin resistance, dyslipidemia, and ultimately cell death that underlie nearly all metabolic disorders. This review summarizes recent findings on the regulatory pathways controlling ceramide production, the molecular mechanisms linking the lipids to these discrete pathogenic events, and exciting attempts to develop therapeutics to reduce ceramide levels to combat metabolic disease.


Asunto(s)
Ceramidas/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Humanos , Resistencia a la Insulina/fisiología , Enfermedades Metabólicas/metabolismo , Esfingolípidos/metabolismo
5.
Gastroenterology ; 165(5): 1136-1150, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37541526

RESUMEN

BACKGROUND & AIMS: Cancers of the alimentary tract, including esophageal adenocarcinomas, colorectal cancers, and cancers of the gastric cardia, are common comorbidities of obesity. Prolonged, excessive delivery of macronutrients to the cells lining the gut can increase one's risk for these cancers by inducing imbalances in the rate of intestinal stem cell proliferation vs differentiation, which can produce polyps and other aberrant growths. We investigated whether ceramides, which are sphingolipids that serve as a signal of nutritional excess, alter stem cell behaviors to influence cancer risk. METHODS: We profiled sphingolipids and sphingolipid-synthesizing enzymes in human adenomas and tumors. Thereafter, we manipulated expression of sphingolipid-producing enzymes, including serine palmitoyltransferase (SPT), in intestinal progenitors of mice, cultured organoids, and Drosophila to discern whether sphingolipids altered stem cell proliferation and metabolism. RESULTS: SPT, which diverts dietary fatty acids and amino acids into the biosynthetic pathway that produces ceramides and other sphingolipids, is a critical modulator of intestinal stem cell homeostasis. SPT and other enzymes in the sphingolipid biosynthesis pathway are up-regulated in human intestinal adenomas. They produce ceramides, which serve as prostemness signals that stimulate peroxisome-proliferator activated receptor-α and induce fatty acid binding protein-1. These actions lead to increased lipid utilization and enhanced proliferation of intestinal progenitors. CONCLUSIONS: Ceramides serve as critical links between dietary macronutrients, epithelial regeneration, and cancer risk.


Asunto(s)
Adenoma , Ceramidas , Humanos , Animales , Ratones , Ceramidas/metabolismo , Ácidos Grasos , Esfingolípidos/metabolismo , Serina C-Palmitoiltransferasa/metabolismo
6.
J Lipid Res ; 63(7): 100197, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35300982

RESUMEN

Plasma lipid levels are altered in chronic conditions such as type 2 diabetes and cardiovascular disease as well as during acute stresses such as fasting and cold exposure. Advances in MS-based lipidomics have uncovered a complex plasma lipidome of more than 500 lipids that serve functional roles, including as energy substrates and signaling molecules. This plasma lipid pool is maintained through regulation of tissue production, secretion, and uptake. A major challenge in understanding the lipidome complexity is establishing the tissues of origin and uptake for various plasma lipids, which is valuable for determining lipid functions. Using cold exposure as an acute stress, we performed global lipidomics on plasma and in nine tissues that may contribute to the circulating lipid pool. We found that numerous species of plasma acylcarnitines (ACars) and ceramides (Cers) were significantly altered upon cold exposure. Through computational assessment, we identified the liver and brown adipose tissue as major contributors and consumers of circulating ACars, in agreement with our previous work. We further identified the kidney and intestine as novel contributors to the circulating ACar pool and validated these findings with gene expression analysis. Regression analysis also identified that the brown adipose tissue and kidney are interactors with the plasma Cer pool. Taken together, these studies provide an adaptable computational tool to assess tissue contribution to the plasma lipid pool. Our findings have further implications in understanding the function of plasma ACars and Cers, which are elevated in metabolic diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Tejido Adiposo Pardo/metabolismo , Frío , Diabetes Mellitus Tipo 2/metabolismo , Ayuno , Humanos , Lipidómica , Lípidos , Termogénesis
7.
FASEB J ; 33(4): 5782-5792, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30645148

RESUMEN

Cone photoreceptors are essential for vision under moderate to high illuminance and allow color discrimination. Their fast dark adaptation rate and resistance to saturation are believed to depend in part on an intraretinal visual cycle that supplies 11- cis-retinaldehyde to cone opsins. Candidate enzymes of this pathway have been reported, but their physiologic contribution to cone photoresponses remains unknown. Here, we evaluate the role of a candidate retinol isomerase of this pathway, sphingolipid δ4 desaturase 1 (Des1). Single-cell RNA sequencing analysis revealed Des1 expression not only in Müller glia but also throughout the retina and in the retinal pigment epithelium. We assessed cone functional dependence on Müller cell-expressed Des1 through a conditional knockout approach. Floxed Des1 mice, on a guanine nucleotide-binding protein subunit α transducin 1 knockout ( Gnat1-/-) background to allow isolated recording of cone-driven photoresponses, were bred with platelet-derived growth factor receptor α (Pdgfrα)-Cre mice to delete Des1 in Müller cells. Conditional knockout of Des1 expression, as shown by tissue-selective Des1 gene recombination and reduced Des1 catalytic activity, caused no gross changes in the retinal structure and had no effect on cone sensitivity or dark adaptation but did slightly accelerate the rate of cone phototransduction termination. These results indicate that Des1 expression in Müller cells is not required for cone visual pigment regeneration in the mouse.-Kiser, P. D., Kolesnikov, A.V., Kiser, J. Z., Dong, Z., Chaurasia, B., Wang, L., Summers, S. A., Hoang, T., Blackshaw, S., Peachey, N. S., Kefalov, V. J., Palczewski, K. Conditional deletion of Des1 in the mouse retina does not impair the visual cycle in cones.


Asunto(s)
Proteínas de la Membrana/metabolismo , Oxidorreductasas/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Visión Ocular/fisiología , Animales , Células Ependimogliales/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinaldehído/metabolismo , Transducina/metabolismo
8.
J Biol Chem ; 290(25): 15371-15379, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25947377

RESUMEN

Sphingolipid synthesis involves a highly conserved biosynthetic pathway that produces fundamental precursors of complex sphingolipids. The final reaction involves the insertion of a double bond into dihydroceramides to generate the more abundant ceramides, which are converted to sphingomyelins and glucosylceramides/gangliosides by the addition of polar head groups. Although ceramides have long been known to mediate cellular stress responses, the dihydroceramides that are transiently produced during de novo sphingolipid synthesis were deemed inert. Evidence published in the last few years suggests that these dihydroceramides accumulate to a far greater extent in tissues than previously thought. Moreover, they have biological functions that are distinct and non-overlapping with those of the more prevalent ceramides. Roles are being uncovered in autophagy, hypoxia, and cellular proliferation, and the lipids are now implicated in the etiology, treatment, and/or diagnosis of diabetes, cancer, ischemia/reperfusion injury, and neurodegenerative diseases. This minireview summarizes recent findings on this emerging class of bioactive lipids.


Asunto(s)
Ceramidas/metabolismo , Diabetes Mellitus/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Daño por Reperfusión/metabolismo , Animales , Autofagia , Proliferación Celular , Ceramidas/genética , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/patología , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Daño por Reperfusión/diagnóstico , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Esfingomielinas/genética , Esfingomielinas/metabolismo
9.
J Immunol ; 188(9): 4141-4, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467660

RESUMEN

Circulating IL-6 levels correlate with the severity of blood-stage malaria in humans and mouse models, but the impact of IL-6 classic signaling through membrane IL-6Rα, as well as IL-6 trans-signaling through soluble IL-6Rα, on the outcome of malaria has remained unknown. In this study, we created IL-6Rα-deficient mice that exhibit a 50% survival of otherwise lethal blood-stage malaria of the genus Plasmodium chabaudi. Inducing IL-6 trans-signaling by injection of mouse recombinant soluble IL-6Rα in IL-6Rα-deficient mice restores the lethal outcome to malaria infection. In contrast, inhibition of IL-6 trans-signaling via injection of recombinant sGP130Fc protein in control mice results in a 40% survival rate. Our data demonstrate that IL-6 trans-signaling, rather than classic IL-6 signaling, contributes to malaria-induced lethality in mice, preceded by an increased inflammatory response. Therefore, inhibition of IL-6 trans-signaling may serve as a novel promising therapeutic basis to combat malaria.


Asunto(s)
Interleucina-6/inmunología , Malaria/inmunología , Plasmodium chabaudi/inmunología , Transducción de Señal/inmunología , Animales , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/inmunología , Receptor gp130 de Citocinas/farmacología , Interleucina-6/genética , Subunidad alfa del Receptor de Interleucina-6/genética , Subunidad alfa del Receptor de Interleucina-6/inmunología , Malaria/genética , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
10.
PLoS Genet ; 6(5): e1000938, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20463885

RESUMEN

A major component of obesity-related insulin resistance is the establishment of a chronic inflammatory state with invasion of white adipose tissue by mononuclear cells. This results in the release of pro-inflammatory cytokines, which in turn leads to insulin resistance in target tissues such as skeletal muscle and liver. To determine the role of insulin action in macrophages and monocytes in obesity-associated insulin resistance, we conditionally inactivated the insulin receptor (IR) gene in myeloid lineage cells in mice (IR(Deltamyel)-mice). While these animals exhibit unaltered glucose metabolism on a normal diet, they are protected from the development of obesity-associated insulin resistance upon high fat feeding. Euglycemic, hyperinsulinemic clamp studies demonstrate that this results from decreased basal hepatic glucose production and from increased insulin-stimulated glucose disposal in skeletal muscle. Furthermore, IR(Deltamyel)-mice exhibit decreased concentrations of circulating tumor necrosis factor (TNF) alpha and thus reduced c-Jun N-terminal kinase (JNK) activity in skeletal muscle upon high fat feeding, reflecting a dramatic reduction of the chronic and systemic low-grade inflammatory state associated with obesity. This is paralleled by a reduced accumulation of macrophages in white adipose tissue due to a pronounced impairment of matrix metalloproteinase (MMP) 9 expression and activity in these cells. These data indicate that insulin action in myeloid cells plays an unexpected, critical role in the regulation of macrophage invasion into white adipose tissue and in the development of obesity-associated insulin resistance.


Asunto(s)
Resistencia a la Insulina , Macrófagos/inmunología , Monocitos/inmunología , Obesidad/inmunología , Receptor de Insulina/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Hígado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inmunología , Obesidad/genética , Obesidad/metabolismo , Receptor de Insulina/genética
11.
Sci Rep ; 12(1): 7273, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35508667

RESUMEN

Ectopic ceramide accumulation in insulin-responsive tissues contributes to the development of obesity and impairs insulin sensitivity. Moreover, pharmacological inhibition of serine palmitoyl transferase (SPT), the first enzyme essential for ceramide biosynthesis using myriocin in rodents reduces body weight and improves insulin sensitivity and associated metabolic indices. Myriocin was originally extracted from fruiting bodies of the fungus Isaria sinclairii and has been found abundant in a number of closely related fungal species such as the Cordyceps. Myriocin is not approved for human use but extracts from Cordyceps are routinely consumed as part of traditional Chinese medication for the treatment of numerous diseases including diabetes. Herein, we screened commercially available extracts of Cordyceps currently being consumed by humans, to identify Cordyceps containing myriocin and test the efficacy of Cordyceps extract containing myriocin in obese mice to improve energy and glucose homeostasis. We demonstrate that commercially available Cordyceps contain variable amounts of myriocin and treatment of mice with a human equivalent dose of Cordyceps extract containing myriocin, reduces ceramide accrual, increases energy expenditure, prevents diet-induced obesity, improves glucose homeostasis and resolves hepatic steatosis. Mechanistically, these beneficial effects were due to increased adipose tissue browning/beiging, improved brown adipose tissue function and hepatic insulin sensitivity as well as alterations in the abundance of gut microbes such as Clostridium and Bilophila. Collectively, our data provide proof-of-principle that myriocin containing Cordyceps extract inhibit ceramide biosynthesis and attenuate metabolic impairments associated with obesity. Moreover, these studies identify commercially available Cordyceps as a readily available supplement to treat obesity and associated metabolic diseases.


Asunto(s)
Cordyceps , Hígado Graso , Resistencia a la Insulina , Animales , Ceramidas/metabolismo , Cordyceps/metabolismo , Hígado Graso/tratamiento farmacológico , Glucosa , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Extractos Vegetales
12.
Mol Metab ; 45: 101145, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33352310

RESUMEN

OBJECTIVE: Aging and weight gain lead to a decline in brown and beige adipocyte functionality that exacerbates obesity and insulin resistance. We sought to determine whether sphingolipids, such as ceramides, a class of lipid metabolites that accumulate in aging and overnutrition, are sufficient or necessary for the metabolic impairment of these thermogenic adipocytes. METHODS: We generated new mouse models allowing for the conditional ablation of genes required for ceramide synthesis (i.e., serine palmitoyltransferase subunit 2, Sptlc2) or degradation (i.e., acid ceramidase 1, Asah1) from mature, thermogenic adipocytes (i.e., from cells expressing uncoupling protein-1). Mice underwent a comprehensive suite of phenotyping protocols to assess energy expenditure and glucose and lipid homeostasis. Complementary studies were conducted in primary brown adipocytes to dissect the mechanisms controlling ceramide synthesis or action. RESULTS: Depletion of Sptlc2 increased energy expenditure, improved glucose homeostasis, and prevented diet-induced obesity. Conversely, depletion of Asah1 led to ceramide accumulation, diminution of energy expenditure, and exacerbation of insulin resistance and obesity. Mechanistically, ceramides slowed lipolysis, inhibited glucose uptake, and decreased mitochondrial respiration. Moreover, ß-adrenergic receptor agonists, which activate thermogenesis in brown adipocytes, decreased transcription of enzymes required for ceramide synthesis. CONCLUSIONS: These studies support our hypothesis that ceramides are necessary and sufficient for the impairment in thermogenic adipocyte function that accompanies obesity. Moreover, they suggest that implementation of therapeutic strategies to block ceramide synthesis in thermogenic adipocytes may serve as a means of improving adipose health and combating obesity and cardiometabolic disease.


Asunto(s)
Adipocitos/metabolismo , Ceramidas/metabolismo , Dieta Alta en Grasa/efectos adversos , Termogénesis , Ceramidasa Ácida/genética , Ceramidasa Ácida/metabolismo , Adipocitos/patología , Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Ceramidas/genética , Metabolismo Energético , Hígado Graso/metabolismo , Hígado Graso/patología , Resistencia a la Insulina , Lipidómica , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Serina C-Palmitoiltransferasa/genética , Serina C-Palmitoiltransferasa/metabolismo , Esfingolípidos/metabolismo , Termogénesis/genética , Transcriptoma , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
13.
Front Immunol ; 11: 576347, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072120

RESUMEN

Adipose depots are heterogeneous tissues that store and sense fuel levels. Through the secretion of lipids, cytokines, and protein hormones (adipokines), they communicate with other organ systems, informing them of the organism's nutritional status. The adipose tissues include diverse types of adipocytes (white, beige, and brown) distinguished by the number/size of lipid droplets, mitochondrial density, and thermogenic capacity. Moreover, they include a spectrum of immune cells that modulate metabolic activity and tissue remodeling. The unique characteristics and interplay of these cells control the production of ceramides, a class of nutrient signals derived from fat and protein metabolism that modulate adipocyte function to regulate glucose and lipid metabolism. The excessive accumulation of ceramides contributes to the adipose tissue inflammation and dysfunction that underlies cardiometabolic disease. Herein we review findings on this important class of lipid species and discuss their role at the convergence point that links overnutrition/inflammation to key features of the metabolic syndrome.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Ceramidas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Adipocitos/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/fisiopatología , Animales , Metabolismo Energético , Ácidos Grasos no Esterificados/metabolismo , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Resistencia a la Insulina , Síndrome Metabólico/inmunología , Síndrome Metabólico/fisiopatología , Obesidad/inmunología , Obesidad/fisiopatología , Transducción de Señal
14.
Artículo en Inglés | MEDLINE | ID: mdl-32636806

RESUMEN

Adipose tissue is a key nutrient-sensing depot that regulates excess energy storage and consumption. Adipocytes, the key components of the adipose tissue, have unique ability to store excess energy in the form of triglycerides, sense systemic energy demands, and secrete factors (lipids, peptides, cytokines, and adipokines) to regulate other metabolic tissues. The presence of various types of adipocytes (white, brown, and beige) characterized by the number/size of lipid droplets, mitochondrial density, and thermogenic capacity, further highlights how intricate is the communication of these cell-types with other metabolic tissues to sense nutrients. In obesity the inherent capacity of adipose tissue to store and sense nutrients is compromised, causing spillover of the intermediate lipid metabolites into circulation and resulting in their ectopic deposition in tissues not suitable for lipid storage, a phenomenon known as lipotoxicity. This results in a spectrum of cellular dysfunction, that underlies various metabolic diseases. Of the numerous lipid classes implicated in eliciting lipotoxicity, sphingolipid: ceramides are among the most deleterious as they modulate signaling pathways involved in regulating glucose metabolism, triglyceride synthesis, apoptosis, and fibrosis. Notably, recent experimental studies have strongly implicated ceramides in the development of numerous metabolic diseases such as insulin resistance, diabetes, cardiomyopathy, hepatic-steatosis, and atherosclerosis. Herein we discuss and summarizes recent findings that implicate ceramides as a key contributor to adipocyte dysfunction underlying metabolic diseases and how depletion of ceramides can be exploited to improve metabolic health.


Asunto(s)
Tejido Adiposo/patología , Ceramidas/metabolismo , Metabolismo de los Lípidos , Enfermedades Metabólicas/patología , Termogénesis , Tejido Adiposo/metabolismo , Animales , Humanos , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo
15.
Diabetes ; 69(8): 1650-1661, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32444367

RESUMEN

An adverse maternal in utero and lactation environment can program offspring for increased risk for metabolic disease. The aim of this study was to determine whether N-acetylcysteine (NAC), an anti-inflammatory antioxidant, attenuates programmed susceptibility to obesity and insulin resistance in offspring of mothers on a high-fat diet (HFD) during pregnancy. CD1 female mice were acutely fed a standard breeding chow or HFD. NAC was added to the drinking water (1 g/kg) of the treatment cohorts from embryonic day 0.5 until the end of lactation. NAC treatment normalized HFD-induced maternal weight gain and oxidative stress, improved the maternal lipidome, and prevented maternal leptin resistance. These favorable changes in the in utero environment normalized postnatal growth, decreased white adipose tissue (WAT) and hepatic fat, improved glucose and insulin tolerance and antioxidant capacity, reduced leptin and insulin, and increased adiponectin in HFD offspring. The lifelong metabolic improvements in the offspring were accompanied by reductions in proinflammatory gene expression in liver and WAT and increased thermogenic gene expression in brown adipose tissue. These results, for the first time, provide a mechanistic rationale for how NAC can prevent the onset of metabolic disease in the offspring of mothers who consume a typical Western HFD.


Asunto(s)
Acetilcisteína/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adiposidad/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Temperatura Corporal , Calorimetría Indirecta , Femenino , Prueba de Tolerancia a la Glucosa , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inyecciones Intraperitoneales , Resistencia a la Insulina , Masculino , Ratones , Aumento de Peso/efectos de los fármacos
16.
Nat Metab ; 1(11): 1051-1058, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-32694860

RESUMEN

Ceramides are products of metabolism that accumulate in individuals with obesity or dyslipidaemia and alter cellular processes in response to fuel surplus. Their actions, when prolonged, elicit the tissue dysfunction that underlies diabetes and heart disease. Here, we review the history of research on these enigmatic molecules, exploring their discovery and mechanisms of action, the evolutionary pressures that have given them their unique attributes and the potential of ceramide-reduction therapies as treatments for cardiometabolic disease.


Asunto(s)
Ceramidas/metabolismo , Dislipidemias/metabolismo , Animales , Ceramidas/sangre , Humanos , Resistencia a la Insulina , Obesidad/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal
17.
Physiol Rep ; 7(18): e14238, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31552709

RESUMEN

The FOXN3 gene locus is associated with fasting blood glucose levels in non-diabetic human population genetic studies. The blood glucose-modifying variation within this gene regulates the abundance of both FOXN3 protein and transcript in primary human hepatocytes, with the hyperglycemia risk allele causing increases in both FOXN3 protein and transcript. Using transgenic and knock-out zebrafish models, we showed previously that FOXN3 is a transcriptional repressor that regulates fasting blood glucose by altering liver gene expression of MYC, a  master transcriptional regulator of glucose utilization, and by modulating pancreatic α cell mass and function through an unknown mechanism. Since homozygous Foxn3 null mice die perinatally, and heterozygous carries of the null allele are smaller than wild-type siblings, we examine the metabolic effects of decreasing mouse liver Foxn3 expression in adult life, performing dynamic endocrine tests not feasible in adult zebrafish. Fasting glucose, glucagon, and insulin; and dynamic responses to glucose, insulin, pyruvate, glutamine, and glucagon were measured. Gluconeogenic and amino acid catabolic gene expression was examined in livers, as well. Knocking down liver Foxn3 expression via transduction with adeno-associated virus serotype 8 particles encoding a short hairpin RNA targeting Fonx3 decreases fasting glucose and increases Myc expression, without altering fasting glucagon or fasting insulin. Liver Foxn3 knock-down confers increases glucose tolerance, has no effect on insulin tolerance or response to glucagon challenge, blunts pyruvate and glutamine tolerance, and modulates expression of amino acid transporters and catabolic enzymes. We conclude that liver Foxn3 regulates substrate selection for gluconeogenesis.


Asunto(s)
Glucemia/metabolismo , Proteínas de Ciclo Celular/fisiología , Factores de Transcripción Forkhead/fisiología , Gluconeogénesis/fisiología , Hígado/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Ayuno/sangre , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes myc , Glucagón/sangre , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética
18.
Cell Rep ; 29(2): 270-282.e5, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31597091

RESUMEN

Accumulation of visceral (VIS) is a predictor of metabolic disorders and insulin resistance. This is due in part to the limited capacity of VIS fat to buffer lipids allowing them to deposit in insulin-sensitive tissues. Mechanisms underlying selective hypertrophic growth and tissue remodeling properties of VIS fat are not well understood. We identified subsets of adipose progenitors (APs) unique to VIS fat with differential Cd34 expression and adipogenic capacity. VIS low (Cd34 low) APs are adipogenic, whereas VIS high (Cd34 high) APs are not. Furthermore, VIS high APs inhibit adipogenic differentiation of SUB and VIS low APs in vitro through the secretion of soluble inhibitory factor(s). The number of VIS high APs increased with adipose tissue expansion, and their abundance in vivo caused hypertrophic growth, fibrosis, inflammation, and metabolic dysfunction. This study unveils the presence of APs unique to VIS fat involved in the paracrine regulation of adipogenesis and tissue remodeling.


Asunto(s)
Antígenos CD34/metabolismo , Grasa Intraabdominal/citología , Comunicación Paracrina , Transducción de Señal , Células Madre/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo Blanco/citología , Animales , Proteína Morfogenética Ósea 4/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Masculino , Ratones Endogámicos C57BL , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Comunicación Paracrina/efectos de los fármacos , Fenotipo , Rosiglitazona/farmacología , Transducción de Señal/efectos de los fármacos , Solubilidad , Células Madre/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
19.
Science ; 365(6451): 386-392, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31273070

RESUMEN

Ceramides contribute to the lipotoxicity that underlies diabetes, hepatic steatosis, and heart disease. By genetically engineering mice, we deleted the enzyme dihydroceramide desaturase 1 (DES1), which normally inserts a conserved double bond into the backbone of ceramides and other predominant sphingolipids. Ablation of DES1 from whole animals or tissue-specific deletion in the liver and/or adipose tissue resolved hepatic steatosis and insulin resistance in mice caused by leptin deficiency or obesogenic diets. Mechanistic studies revealed ceramide actions that promoted lipid uptake and storage and impaired glucose utilization, none of which could be recapitulated by (dihydro)ceramides that lacked the critical double bond. These studies suggest that inhibition of DES1 may provide a means of treating hepatic steatosis and metabolic disorders.


Asunto(s)
Ceramidas/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Resistencia a la Insulina/genética , Proteínas de la Membrana/genética , Oxidorreductasas/genética , Animales , Ceramidas/química , Ceramidas/genética , Dieta Alta en Grasa/efectos adversos , Eliminación de Gen , Leptina/deficiencia , Ratones , Ratones Mutantes , Esfingolípidos/química , Esfingolípidos/metabolismo
20.
Trends Endocrinol Metab ; 29(9): 597-599, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29685851

RESUMEN

Chaurasia and colleagues discuss the provocative new finding that some enzymes in the de novo sphingolipid synthesis pathway have dual roles as transcriptional regulators.


Asunto(s)
Oxidorreductasas/genética , Esfingolípidos , Expresión Génica , Lipogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA