Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 165(5): 1136-1150, 2023 11.
Article in English | MEDLINE | ID: mdl-37541526

ABSTRACT

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.


Subject(s)
Adenoma , Ceramides , Humans , Animals , Mice , Ceramides/metabolism , Fatty Acids , Sphingolipids/metabolism , Serine C-Palmitoyltransferase/metabolism
3.
Sci Rep ; 12(1): 7273, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508667

ABSTRACT

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.


Subject(s)
Cordyceps , Fatty Liver , Insulin Resistance , Animals , Ceramides/metabolism , Cordyceps/metabolism , Fatty Liver/drug therapy , Glucose , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Plant Extracts
4.
J Lipid Res ; 63(7): 100197, 2022 07.
Article in English | MEDLINE | ID: mdl-35300982

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Adipose Tissue, Brown/metabolism , Cold Temperature , Diabetes Mellitus, Type 2/metabolism , Fasting , Humans , Lipidomics , Lipids , Thermogenesis
5.
Mol Metab ; 45: 101145, 2021 03.
Article in English | MEDLINE | ID: mdl-33352310

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Ceramides/metabolism , Diet, High-Fat/adverse effects , Thermogenesis , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Adipocytes/pathology , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Ceramides/genetics , Energy Metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Insulin Resistance , Lipidomics , Male , Mice , Mice, Knockout , Obesity/metabolism , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingolipids/metabolism , Thermogenesis/genetics , Transcriptome , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
6.
Annu Rev Physiol ; 83: 303-330, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33158378

ABSTRACT

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.


Subject(s)
Ceramides/metabolism , Lipid Metabolism/physiology , Animals , Humans , Insulin Resistance/physiology , Metabolic Diseases/metabolism , Sphingolipids/metabolism
7.
Front Immunol ; 11: 576347, 2020.
Article in English | MEDLINE | ID: mdl-33072120

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Ceramides/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Metabolic Syndrome/metabolism , Obesity/metabolism , Adipocytes/immunology , Adipose Tissue/immunology , Adipose Tissue/physiopathology , Animals , Energy Metabolism , Fatty Acids, Nonesterified/metabolism , Humans , Inflammation/immunology , Inflammation/physiopathology , Insulin Resistance , Metabolic Syndrome/immunology , Metabolic Syndrome/physiopathology , Obesity/immunology , Obesity/physiopathology , Signal Transduction
8.
Article in English | MEDLINE | ID: mdl-32636806

ABSTRACT

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.


Subject(s)
Adipose Tissue/pathology , Ceramides/metabolism , Lipid Metabolism , Metabolic Diseases/pathology , Thermogenesis , Adipose Tissue/metabolism , Animals , Humans , Metabolic Diseases/etiology , Metabolic Diseases/metabolism
9.
Diabetes ; 69(8): 1650-1661, 2020 08.
Article in English | MEDLINE | ID: mdl-32444367

ABSTRACT

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.


Subject(s)
Acetylcysteine/therapeutic use , Diet, High-Fat/adverse effects , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Antioxidants/metabolism , Body Temperature , Calorimetry, Indirect , Female , Glucose Tolerance Test , Inflammation/drug therapy , Inflammation/metabolism , Injections, Intraperitoneal , Insulin Resistance , Male , Mice , Weight Gain/drug effects
10.
Cell Rep ; 29(2): 270-282.e5, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31597091

ABSTRACT

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.


Subject(s)
Antigens, CD34/metabolism , Intra-Abdominal Fat/cytology , Paracrine Communication , Signal Transduction , Stem Cells/metabolism , Adipogenesis/drug effects , Adipose Tissue, White/cytology , Animals , Bone Morphogenetic Protein 4/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Insulin Resistance , Intra-Abdominal Fat/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Paracrine Communication/drug effects , Phenotype , Rosiglitazone/pharmacology , Signal Transduction/drug effects , Solubility , Stem Cells/drug effects , Weight Gain/drug effects
11.
Physiol Rep ; 7(18): e14238, 2019 09.
Article in English | MEDLINE | ID: mdl-31552709

ABSTRACT

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.


Subject(s)
Blood Glucose/metabolism , Cell Cycle Proteins/physiology , Forkhead Transcription Factors/physiology , Gluconeogenesis/physiology , Liver/metabolism , Amino Acids/genetics , Amino Acids/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Fasting/blood , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Genes, myc , Glucagon/blood , Glucose Tolerance Test , Insulin/blood , Male , Mice, Inbred C57BL , RNA, Messenger/genetics
12.
Science ; 365(6451): 386-392, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31273070

ABSTRACT

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.


Subject(s)
Ceramides/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Insulin Resistance/genetics , Membrane Proteins/genetics , Oxidoreductases/genetics , Animals , Ceramides/chemistry , Ceramides/genetics , Diet, High-Fat/adverse effects , Gene Deletion , Leptin/deficiency , Mice , Mice, Mutant Strains , Sphingolipids/chemistry , Sphingolipids/metabolism
13.
FASEB J ; 33(4): 5782-5792, 2019 04.
Article in English | MEDLINE | ID: mdl-30645148

ABSTRACT

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.


Subject(s)
Membrane Proteins/metabolism , Oxidoreductases/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Vision, Ocular/physiology , Animals , Ependymoglial Cells/metabolism , Male , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinaldehyde/metabolism , Transducin/metabolism
14.
Nat Metab ; 1(11): 1051-1058, 2019 11.
Article in English | MEDLINE | ID: mdl-32694860

ABSTRACT

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.


Subject(s)
Ceramides/metabolism , Dyslipidemias/metabolism , Animals , Ceramides/blood , Humans , Insulin Resistance , Obesity/metabolism , Severity of Illness Index , Signal Transduction
15.
Cell Rep ; 25(7): 1708-1717.e5, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428342

ABSTRACT

Autophagy is a homeostatic cellular process involved in the degradation of long-lived or damaged cellular components. The role of autophagy in adipogenesis is well recognized, but its role in mature adipocyte function is largely unknown. We show that the autophagy proteins Atg3 and Atg16L1 are required for proper mitochondrial function in mature adipocytes. In contrast to previous studies, we found that post-developmental ablation of autophagy causes peripheral insulin resistance independently of diet or adiposity. Finally, lack of adipocyte autophagy reveals cross talk between fat and liver, mediated by lipid peroxide-induced Nrf2 signaling. Our data reveal a role for autophagy in preventing lipid peroxide formation and its transfer in insulin-sensitive peripheral tissues.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , Autophagy , Insulin Resistance , Lipid Peroxides/metabolism , Liver/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Autophagy-Related Proteins/metabolism , Body Composition , Body Weight , Humans , Inflammation/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Lipoproteins, HDL/metabolism , Mice, Knockout , Mitochondria/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
16.
Trends Endocrinol Metab ; 29(9): 597-599, 2018 09.
Article in English | MEDLINE | ID: mdl-29685851

ABSTRACT

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


Subject(s)
Oxidoreductases/genetics , Sphingolipids , Gene Expression , Lipogenesis
18.
Cell Metab ; 24(6): 820-834, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27818258

ABSTRACT

Adipocytes package incoming fatty acids into triglycerides and other glycerolipids, with only a fraction spilling into a parallel biosynthetic pathway that produces sphingolipids. Herein, we demonstrate that subcutaneous adipose tissue of type 2 diabetics contains considerably more sphingolipids than non-diabetic, BMI-matched counterparts. Whole-body and adipose tissue-specific inhibition/deletion of serine palmitoyltransferase (Sptlc), the first enzyme in the sphingolipid biosynthesis cascade, in mice markedly altered adipose morphology and metabolism, particularly in subcutaneous adipose tissue. The reduction in adipose sphingolipids increased brown and beige/brite adipocyte numbers, mitochondrial activity, and insulin sensitivity. The manipulation also increased numbers of anti-inflammatory M2 macrophages in the adipose bed and induced secretion of insulin-sensitizing adipokines. By comparison, deletion of serine palmitoyltransferase from macrophages had no discernible effects on metabolic homeostasis or adipose function. These data indicate that newly synthesized adipocyte sphingolipids are nutrient signals that drive changes in the adipose phenotype to influence whole-body energy expenditure and nutrient metabolism.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Ceramides/pharmacology , Inflammation/pathology , Subcutaneous Fat/pathology , Adipocytes/drug effects , Adipose Tissue, Brown/drug effects , Adrenergic beta-Agonists/pharmacology , Adult , Aged , Animals , Body Mass Index , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cold Temperature , Diabetes Mellitus/metabolism , Dioxoles/pharmacology , Energy Metabolism/drug effects , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation/drug effects , Glucose/metabolism , Humans , Inflammation/genetics , Mice , Middle Aged , Obesity/metabolism , Obesity/pathology , Organ Specificity/drug effects , Serine C-Palmitoyltransferase/metabolism , Sphingolipids/biosynthesis , Sphingolipids/metabolism , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Thermogenesis/drug effects , Thermogenesis/genetics , Young Adult
20.
Cell ; 165(4): 882-95, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133169

ABSTRACT

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.


Subject(s)
Brain/metabolism , Diet, High-Fat , Glucose/metabolism , Obesity/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/metabolism , Cognition , Endothelial Cells/metabolism , Fatty Acids/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Myeloid Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...