Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Diabetologia ; 61(6): 1459-1469, 2018 06.
Article in English | MEDLINE | ID: mdl-29550873

ABSTRACT

AIMS/HYPOTHESIS: Low-protein diets are well known to improve glucose tolerance and increase energy expenditure. Increases in circulating fibroblast growth factor 21 (FGF21) have been implicated as a potential underlying mechanism. METHODS: We aimed to test whether low-protein diets in the context of a high-carbohydrate or high-fat regimen would also protect against type 2 diabetes in New Zealand Obese (NZO) mice used as a model of polygenetic obesity and type 2 diabetes. Mice were placed on high-fat diets that provided protein at control (16 kJ%; CON) or low (4 kJ%; low-protein/high-carbohydrate [LP/HC] or low-protein/high-fat [LP/HF]) levels. RESULTS: Protein restriction prevented the onset of hyperglycaemia and beta cell loss despite increased food intake and fat mass. The effect was seen only under conditions of a lower carbohydrate/fat ratio (LP/HF). When the carbohydrate/fat ratio was high (LP/HC), mice developed type 2 diabetes despite the robustly elevated hepatic FGF21 secretion and increased energy expenditure. CONCLUSION/INTERPRETATION: Prevention of type 2 diabetes through protein restriction, without lowering food intake and body fat mass, is compromised by high dietary carbohydrates. Increased FGF21 levels and elevated energy expenditure do not protect against hyperglycaemia and type 2 diabetes per se.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, Protein-Restricted , Dietary Carbohydrates/metabolism , Adipose Tissue , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Type 2/genetics , Energy Metabolism , Fibroblast Growth Factors/genetics , Glucose Tolerance Test , Insulin/metabolism , Insulin Resistance , Male , Mice , Mice, Obese , Mice, Transgenic , Obesity/metabolism
2.
J Immunol ; 196(9): 3951-62, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27036914

ABSTRACT

Sphingolipids are major components of the plasma membrane. In particular, ceramide serves as an essential building hub for complex sphingolipids, but also as an organizer of membrane domains segregating receptors and signalosomes. Sphingomyelin breakdown as a result of sphingomyelinase activation after ligation of a variety of receptors is the predominant source of ceramides released at the plasma membrane. This especially applies to T lymphocytes where formation of ceramide-enriched membrane microdomains modulates TCR signaling. Because ceramide release and redistribution occur very rapidly in response to receptor ligation, novel tools to further study these processes in living T cells are urgently needed. To meet this demand, we synthesized nontoxic, azido-functionalized ceramides allowing for bio-orthogonal click-reactions to fluorescently label incorporated ceramides, and thus investigate formation of ceramide-enriched domains. Azido-functionalized C6-ceramides were incorporated into and localized within plasma membrane microdomains and proximal vesicles in T cells. They segregated into clusters after TCR, and especially CD28 ligation, indicating efficient sorting into plasma membrane domains associated with T cell activation; this was abolished upon sphingomyelinase inhibition. Importantly, T cell activation was not abrogated upon incorporation of the compound, which was efficiently excluded from the immune synapse center as has previously been seen in Ab-based studies using fixed cells. Therefore, the functionalized ceramides are novel, highly potent tools to study the subcellular redistribution of ceramides in the course of T cell activation. Moreover, they will certainly also be generally applicable to studies addressing rapid stimulation-mediated ceramide release in living cells.


Subject(s)
Azides/metabolism , Membrane Microdomains/metabolism , Microscopy, Fluorescence/methods , Receptors, Antigen, T-Cell/metabolism , Sphingolipids/metabolism , T-Lymphocytes/metabolism , Azides/chemistry , Cells, Cultured , Humans , Lymphocyte Activation , Protein Transport , Receptor Aggregation , Signal Transduction , Sphingolipids/chemistry , T-Lymphocytes/immunology
3.
J Immunol ; 197(8): 3130-3141, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27638864

ABSTRACT

CD4+ Foxp3+ regulatory T cells (Tregs) depend on CD28 signaling for their survival and function, a receptor that has been previously shown to activate the acid sphingomyelinase (Asm)/ceramide system. In this article, we show that the basal and CD28-induced Asm activity is higher in Tregs than in conventional CD4+ T cells (Tconvs) of wild-type (wt) mice. In Asm-deficient (Smpd1-/-; Asm-/-) mice, as compared with wt mice, the frequency of Tregs among CD4+ T cells, turnover of the effector molecule CTLA-4, and their suppressive activity in vitro were increased. The biological significance of these findings was confirmed in our Treg-sensitive mouse model of measles virus (MV) CNS infection, in which we observed more infected neurons and less MV-specific CD8+ T cells in brains of Asm-/- mice compared with wt mice. In addition to genetic deficiency, treatment of wt mice with the Asm inhibitor amitriptyline recapitulated the phenotype of Asm-deficient mice because it also increased the frequency of Tregs among CD4+ T cells. Reduced absolute cell numbers of Tconvs after inhibitor treatment in vivo and extensive in vitro experiments revealed that Tregs are more resistant toward Asm inhibitor-induced cell death than Tconvs. Mechanistically, IL-2 was capable of providing crucial survival signals to the Tregs upon inhibitor treatment in vitro, shifting the Treg/Tconv ratio to the Treg side. Thus, our data indicate that Asm-inhibiting drugs should be further evaluated for the therapy of inflammatory and autoimmune disorders.


Subject(s)
Brain/immunology , Measles/immunology , Morbillivirus/immunology , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Brain/virology , CD28 Antigens/metabolism , CD4 Antigens/metabolism , Cell Differentiation , Cell Survival , Cells, Cultured , Ceramides/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2/metabolism , Lymphocyte Activation , Measles/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/virology
4.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L625-L637, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28283474

ABSTRACT

Pulmonary complications from stored blood products are the leading cause of mortality related to transfusion. Transfusion-related acute lung injury is mediated by antibodies or bioactive mediators, yet underlying mechanisms are incompletely understood. Sphingolipids such as ceramide regulate lung injury, and their composition changes as a function of time in stored blood. Here, we tested the hypothesis that aged platelets may induce lung injury via a sphingolipid-mediated mechanism. To assess this hypothesis, a two-hit mouse model was devised. Recipient mice were treated with 2 mg/kg intraperitoneal lipopolysaccharide (priming) 2 h before transfusion of 10 ml/kg stored (1-5 days) platelets treated with or without addition of acid sphingomyelinase inhibitor ARC39 or platelets from acid sphingomyelinase-deficient mice, which both reduce ceramide formation. Transfused mice were examined for signs of pulmonary neutrophil accumulation, endothelial barrier dysfunction, and histological evidence of lung injury. Sphingolipid profiles in stored platelets were analyzed by mass spectrophotometry. Transfusion of aged platelets into primed mice induced characteristic features of lung injury, which increased in severity as a function of storage time. Ceramide accumulated in platelets during storage, but this was attenuated by ARC39 or in acid sphingomyelinase-deficient platelets. Compared with wild-type platelets, transfusion of ARC39-treated or acid sphingomyelinase-deficient aged platelets alleviated lung injury. Aged platelets elicit lung injury in primed recipient mice, which can be alleviated by pharmacological inhibition or genetic deletion of acid sphingomyelinase. Interventions targeting sphingolipid formation represent a promising strategy to increase the safety and longevity of stored blood products.


Subject(s)
Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Blood Platelets/metabolism , Cellular Senescence , Platelet Transfusion/adverse effects , Sphingomyelin Phosphodiesterase/metabolism , Acute Lung Injury/pathology , Animals , Blood Platelets/drug effects , Ceramides/metabolism , Enzyme Inhibitors/pharmacology , Gene Deletion , Humans , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Time Factors
5.
Ann Surg ; 265(1): 218-226, 2017 01.
Article in English | MEDLINE | ID: mdl-28009749

ABSTRACT

OBJECTIVE: We aimed to identify the role of the enzyme acid sphingomyelinase in the aging of stored units of packed red blood cells (pRBCs) and subsequent lung inflammation after transfusion. SUMMARY BACKGROUND DATA: Large volume pRBC transfusions are associated with multiple adverse clinical sequelae, including lung inflammation. Microparticles are formed in stored pRBCs over time and have been shown to contribute to lung inflammation after transfusion. METHODS: Human and murine pRBCs were stored with or without amitriptyline, a functional inhibitor of acid sphingomyelinase, or obtained from acid sphingomyelinase-deficient mice, and lung inflammation was studied in mice receiving transfusions of pRBCs and microparticles isolated from these units. RESULTS: Acid sphingomyelinase activity in pRBCs was associated with the formation of ceramide and the release of microparticles. Treatment of pRBCs with amitriptyline inhibited acid sphingomyelinase activity, ceramide accumulation, and microparticle production during pRBC storage. Transfusion of aged pRBCs or microparticles isolated from aged blood into mice caused lung inflammation. This was attenuated after transfusion of pRBCs treated with amitriptyline or from acid sphingomyelinase-deficient mice. CONCLUSIONS: Acid sphingomyelinase inhibition in stored pRBCs offers a novel mechanism for improving the quality of stored blood.


Subject(s)
Amitriptyline/pharmacology , Blood Preservation/methods , Enzyme Inhibitors/pharmacology , Erythrocyte Transfusion/adverse effects , Erythrocytes/drug effects , Pneumonia/etiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Animals , Biomarkers/metabolism , Blood Preservation/adverse effects , Cell-Derived Microparticles/metabolism , Erythrocytes/enzymology , Humans , Male , Mice , Mice, Inbred C57BL , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/prevention & control , Sphingomyelin Phosphodiesterase/deficiency
6.
Cell Physiol Biochem ; 43(2): 445-456, 2017.
Article in English | MEDLINE | ID: mdl-28922661

ABSTRACT

BACKGROUND/AIMS: Obesity is a main risk factor for the development of hepatic insulin resistance and it is accompanied by adipocyte hypertrophy and an elevated expression of different adipokines such as autotaxin (ATX). ATX converts lysophosphatidylcholine to lysophosphatidic acid (LPA) and acts as the main producer of extracellular LPA. This bioactive lipid regulates a broad range of physiological and pathological responses by activation of LPA receptors (LPA1-6). METHODS: The activation of phosphatidylinositide 3-kinases (PI3K) signaling (Akt and GSK-3ß) was analyzed via western blotting in primary rat hepatocytes. Incorporation of glucose into glycogen was measured by using radio labeled glucose. Real-time PCR analysis and pharmacological modulation of LPA receptors were performed. Human plasma LPA levels of obese (BMI > 30, n = 18) and normal weight individuals (BMI 18.5-25, n = 14) were analyzed by liquid chromatography tandem-mass spectrometry (LC-MS/MS). RESULTS: Pretreatment of primary hepatocytes with LPA resulted in an inhibition of insulin-mediated Gck expression, PI3K activation and glycogen synthesis. Pharmacological approaches revealed that the LPA3-receptor subtype is responsible for the inhibitory effect of LPA on insulin signaling. Moreover, human plasma LPA concentrations (16: 0 LPA) of obese participants (BMI > 30) are significantly elevated in comparison to normal weight individuals (BMI 18.5-25). CONCLUSION: LPA is able to interrupt insulin signaling in primary rat hepatocytes via the LPA3 receptor subtype. Moreover, the bioactive lipid LPA (16: 0) is increased in obesity.


Subject(s)
Hepatocytes/metabolism , Insulin/metabolism , Lysophospholipids/metabolism , Obesity/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Animals , Cells, Cultured , Glycogen/metabolism , Humans , Lysophospholipids/blood , Male , Obesity/blood , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar
7.
J Vasc Surg ; 65(4): 1171-1179.e1, 2017 04.
Article in English | MEDLINE | ID: mdl-26960947

ABSTRACT

BACKGROUND: Abdominal aortic aneurysm (AAA) is a deadly irreversible weakening and distension of the abdominal aortic wall. The pathogenesis of AAA remains poorly understood. Investigation into the physical and molecular characteristics of perivascular adipose tissue (PVAT) adjacent to AAA has not been done before and is the purpose of this study. METHODS AND RESULTS: Human aortae, periaortic PVAT, and fat surrounding peripheral arteries were collected from patients undergoing elective surgical repair of AAA. Control aortas were obtained from recently deceased healthy organ donors with no known arterial disease. Aorta and PVAT was found in AAA to larger extent compared with control aortas. Immunohistochemistry revealed neutrophils, macrophages, mast cells, and T-cells surrounding necrotic adipocytes. Gene expression analysis showed that neutrophils, mast cells, and T-cells were found to be increased in PVAT compared with AAA as well as cathepsin K and S. The concentration of ceramides in PVAT was determined using mass spectrometry and correlated with content of T-cells in the PVAT. CONCLUSIONS: Our results suggest a role for abnormal necrotic, inflamed, proteolytic adipose tissue to the adjacent aneurysmal aortic wall in ongoing vascular damage.


Subject(s)
Adipocytes/enzymology , Adipose Tissue/enzymology , Aortic Aneurysm, Abdominal/enzymology , Ceramides/analysis , Mast Cells/enzymology , Neutrophils/enzymology , Peptide Hydrolases/analysis , T-Lymphocytes/enzymology , Adipocytes/immunology , Adipocytes/pathology , Adipose Tissue/immunology , Adipose Tissue/pathology , Antigens, CD/analysis , Antigens, CD/genetics , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cytokines/analysis , Cytokines/genetics , GPI-Linked Proteins/analysis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Enzymologic , Humans , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Mast Cells/immunology , Mast Cells/pathology , Necrosis , Neutrophils/immunology , Neutrophils/pathology , Peptide Hydrolases/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology
8.
Biochim Biophys Acta ; 1851(2): 194-202, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25457224

ABSTRACT

The matricellular protein connective tissue growth factor (CTGF/CCN2) is recognized as key player in the onset of fibrosis in various tissues, including skeletal muscle. In many circumstances, CTGF has been shown to be induced by transforming growth factor beta (TGFß) and accounting, at least in part, for its biological action. In this study it was verified that in cultured myoblasts CTGF/CCN2 causes their transdifferentiation into myofibroblasts by up-regulating the expression of fibrosis marker proteins α-smooth muscle actin and transgelin. Interestingly, it was also found that the profibrotic effect exerted by CTGF/CCN2 was mediated by the sphingosine kinase (SK)-1/S1P3 signaling axis specifically induced by the treatment with the profibrotic cue. Following CTGF/CCN2-induced up-regulation, S1P3 became the S1P receptor subtype expressed at the highest degree, at least at mRNA level, and was thus capable of readdressing the sphingosine 1-phosphate signaling towards fibrosis rather than myogenic differentiation. Another interesting finding is that CTGF/CCN2 silencing prevented the TGFß-dependent up-regulation of SK1/S1P3 signaling axis and strongly reduced the profibrotic effect exerted by TGFß, pointing at a crucial role of endogenous CTGF/CCN2 generated following TGFß challenge in the transmission of at least part of its profibrotic effect. These results provide new insights into the molecular mechanism by which CTGF/CCN2 drives its biological action and strengthen the concept that SK1/S1P3 axis plays a critical role in the onset of fibrotic cell phenotype.


Subject(s)
Cell Transdifferentiation , Connective Tissue Growth Factor/metabolism , Myoblasts, Skeletal/enzymology , Myofibroblasts/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Cell Line , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/pharmacology , Dose-Response Relationship, Drug , Fibrosis , Mice , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Myofibroblasts/drug effects , Myofibroblasts/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Interference , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/drug effects , Receptors, Lysosphingolipid/genetics , Recombinant Proteins/pharmacology , Sphingosine-1-Phosphate Receptors , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology , Up-Regulation
9.
J Neurochem ; 137(4): 589-603, 2016 05.
Article in English | MEDLINE | ID: mdl-26788861

ABSTRACT

Reward-dependent instrumental behavior must continuously be re-adjusted according to environmental conditions. Failure to adapt to changes in reward contingencies may incur psychiatric disorders like anxiety and depression. When an expected reward is omitted, behavior undergoes extinction. While extinction involves active re-learning, it is also accompanied by emotional behaviors indicative of frustration, anxiety, and despair (extinction-induced depression). Here, we report evidence for a sphingolipid mechanism in the extinction of behavior. Rapid extinction, indicating efficient re-learning, coincided with a decrease in the activity of the enzyme acid sphingomyelinase (ASM), which catalyzes turnover of sphingomyelin to ceramide, in the dorsal hippocampus of rats. The stronger the decline in ASM activity, the more rapid was the extinction. Sphingolipid-focused lipidomic analysis showed that this results in a decline of local ceramide species in the dorsal hippocampus. Ceramides shape the fluidity of lipid rafts in synaptic membranes and by that way can control neural plasticity. We also found that aging modifies activity of enzymes and ceramide levels in selective brain regions. Aging also changed how the chronic treatment with corticosterone (stress) or intranasal dopamine modified regional enzyme activity and ceramide levels, coinciding with rate of extinction. These data provide first evidence for a functional ASM-ceramide pathway in the brain involved in the extinction of learned behavior. This finding extends the known cellular mechanisms underlying behavioral plasticity to a new class of membrane-located molecules, the sphingolipids, and their regulatory enzymes, and may offer new treatment targets for extinction- and learning-related psychopathological conditions. Sphingolipids are common lipids in the brain which form lipid domains at pre- and postsynaptic membrane compartments. Here we show a decline in dorsal hippocampus ceramide species together with a reduction of acid sphingomyelinase activity during extinction of conditioned behavior in rats. This reduction was associated with expression of re-learning-related behavior, but not with emotional behaviors. Read the Editorial Highlight for this article on page 485.


Subject(s)
Ceramides/metabolism , Conditioning, Operant/physiology , Extinction, Psychological/physiology , Sphingolipids/metabolism , Animals , Male , Rats , Rats, Wistar , Reaction Time/physiology , Sphingomyelins/metabolism
10.
J Hepatol ; 64(1): 60-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26254847

ABSTRACT

BACKGROUND & AIMS: Exosomes are small membrane vesicles involved in intercellular communication. Hepatocytes are known to release exosomes, but little is known about their biological function. We sought to determine if exosomes derived from hepatocytes contribute to liver repair and regeneration after injury. METHODS: Exosomes derived from primary murine hepatocytes were isolated and characterized biochemically and biophysically. Using cultures of primary hepatocytes, we tested whether hepatocyte exosomes induced proliferation of hepatocytes in vitro. Using models of ischemia/reperfusion injury and partial hepatectomy, we evaluated whether hepatocyte exosomes promote hepatocyte proliferation and liver regeneration in vivo. RESULTS: Hepatocyte exosomes, but not exosomes from other liver cell types, induce dose-dependent hepatocyte proliferation in vitro and in vivo. Mechanistically, hepatocyte exosomes directly fuse with target hepatocytes and transfer neutral ceramidase and sphingosine kinase 2 (SK2) causing increased synthesis of sphingosine-1-phosphate (S1P) within target hepatocytes. Ablation of exosomal SK prevents the proliferative effect of exosomes. After ischemia/reperfusion injury, the number of circulating exosomes with proliferative effects increases. CONCLUSIONS: Our data shows that hepatocyte-derived exosomes deliver the synthetic machinery to form S1P in target hepatocytes resulting in cell proliferation and liver regeneration after ischemia/reperfusion injury or partial hepatectomy. These findings represent a potentially novel new contributing mechanism of liver regeneration and have important implications for new therapeutic approaches to acute and chronic liver disease.


Subject(s)
Exosomes/physiology , Liver Regeneration , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Animals , Cell Proliferation , Hepatocytes/physiology , Male , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine/physiology
11.
Cell Physiol Biochem ; 40(6): 1637-1645, 2016.
Article in English | MEDLINE | ID: mdl-28006772

ABSTRACT

BACKGROUND/AIMS: Ectopic lipid accumulation in hepatocytes has been identified as a risk factor for the progression of liver fibrosis and is strongly associated with obesity. In particular, the saturated fatty acid palmitate is involved in initiation of liver fibrosis via formation of secondary metabolites by hepatocytes that in turn activate hepatic stellate cells (HSCs) in a paracrine manner. METHODS: α-smooth muscle actin-expression (α-SMA) as a marker of liver fibrosis was investigated via western blot analysis and immunofluorescence microscopy in HSCs (LX-2). Sphingolipid metabolism and the generation of the bioactive secondary metabolite sphingosine 1-phosphate (S1P) in response to palmitate were analyzed by LC-MS/MS in hepatocytes (HepG2). To identify the molecular mechanism involved in the progression of liver fibrosis real-time PCR analysis and pharmacological modulation of S1P receptors were performed. RESULTS: Palmitate oversupply increased intra- and extracellular S1P-concentrations in hepatocytes. Conditioned medium from HepG2 cells initiated fibrosis by enhancing α-SMA-expression in LX-2 in a S1P-dependent manner. In accordance, fibrotic response in the presence of S1P was also observed in HSCs. Pharmacological inhibition of S1P receptors demonstrated that S1P3 is the crucial receptor subtype involved in this process. CONCLUSION: S1P is synthesized in hepatocytes in response to palmitate and released into the extracellular environment leading to an activation of HSCs via the S1P3 receptor.


Subject(s)
Lysophospholipids/pharmacology , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Palmitates/adverse effects , Sphingosine/analogs & derivatives , Actins/metabolism , Culture Media, Conditioned/pharmacology , Hep G2 Cells , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology
12.
FASEB J ; 29(8): 3357-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25911610

ABSTRACT

Glucolipotoxic stress has been identified as a key player in the progression of pancreatic ß-cell dysfunction contributing to insulin resistance and the development of type 2 diabetes mellitus (T2D). It has been suggested that bioactive lipid intermediates, formed under lipotoxic conditions, are involved in these processes. Here, we show that sphingosine 1-phosphate (S1P) levels are not only increased in palmitate-stimulated pancreatic ß-cells but also regulate ß-cell homeostasis in a divergent manner. Although S1P possesses a prosurvival effect in ß-cells, an enhanced level of the sphingolipid antagonizes insulin-mediated cell growth and survival via the sphingosine 1-phosphate receptor subtype 2 (S1P2) followed by an inhibition of Akt-signaling. In an attempt to investigate the role of the S1P/S1P2 axis in vivo, the New Zealand obese (NZO) diabetic mouse model, characterized by ß-cell loss under high-fat diet (HFD) conditions, was used. The occurrence of T2D was accompanied by an increase of plasma S1P levels. To examine whether S1P contributes to the morphologic changes of islets via S1P2, the receptor antagonist JTE-013 was administered. Most interestingly, JTE-013 rescued ß-cell damage clearly indicating an important role of the S1P2 in ß-cell homeostasis. Therefore, the present study provides a new therapeutic strategy to diminish ß-cell dysfunction and the development of T2D.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lysophospholipids/metabolism , Mice, Obese/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/methods , Disease Models, Animal , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Male , Mice , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Sphingosine/metabolism
13.
Diabetologia ; 57(2): 373-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24292566

ABSTRACT

AIMS/HYPOTHESIS: Enhanced plasma levels of NEFA have been shown to induce hepatic insulin resistance, which contributes to the development of type 2 diabetes. Indeed, sphingolipids can be formed via a de novo pathway from the saturated fatty acid palmitate and the amino acid serine. Besides ceramides, sphingosine 1-phosphate (S1P) has been identified as a major bioactive lipid mediator. Therefore, our aim was to investigate the generation and function of S1P in hepatic insulin resistance. METHODS: The incorporation of palmitate into sphingolipids was performed by rapid-resolution liquid chromatography-MS/MS in primary human and rat hepatocytes. The influence of S1P and the involvement of S1P receptors in hepatic insulin resistance was examined in human and rat hepatocytes, as well as in New Zealand obese (NZO) mice. RESULTS: Palmitate induced an impressive formation of extra- and intracellular S1P in rat and human hepatocytes. An elevation of hepatic S1P levels was observed in NZO mice fed a high-fat diet. Once generated, S1P was able, similarly to palmitate, to counteract insulin signalling. The inhibitory effect of S1P was abolished in the presence of the S1P2 receptor antagonist JTE-013 both in vitro and in vivo. In agreement with this, the immunomodulator FTY720-phosphate, which binds to all S1P receptors except S1P2, was not able to inhibit insulin signalling. CONCLUSIONS/INTERPRETATION: These data indicate that palmitate is metabolised by hepatocytes to S1P, which acts via stimulation of the S1P2 receptor to impair insulin signalling. In particular, S1P2 inhibition could be considered as a novel therapeutic target for the treatment of insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hepatocytes/physiology , Immunosuppressive Agents/pharmacology , Insulin Resistance , Lysophospholipids/metabolism , Organophosphates/pharmacology , Palmitates/pharmacology , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Chromatography, Liquid , Hepatocytes/drug effects , Immunosuppressive Agents/metabolism , Insulin/metabolism , Male , Mice , Mice, Obese , Organophosphates/metabolism , Rats , Rats, Wistar , Sphingosine/metabolism , Sphingosine/pharmacology
14.
Cell Physiol Biochem ; 34(1): 134-47, 2014.
Article in English | MEDLINE | ID: mdl-24977487

ABSTRACT

Insulin resistance is a complex metabolic disorder in which insulin-sensitive tissues fail to respond to the physiological action of insulin. There is a strong correlation of insulin resistance and the development of type 2 diabetes both reaching epidemic proportions. Dysfunctional lipid metabolism is a hallmark of insulin resistance and a risk factor for several cardiovascular and metabolic disorders. Numerous studies in humans and rodents have shown that insulin resistance is associated with elevations of non-esterified fatty acids (NEFA) in the plasma. Moreover, bioactive lipid intermediates such as diacylglycerol (DAG) and ceramides appear to accumulate in response to NEFA, which may interact with insulin signaling. However, recent work has also indicated that sphingosine 1-phosphate (S1P), a breakdown product of ceramide, modulate insulin signaling in different cell types. In this review, we summarize the current state of knowledge about S1P and insulin signaling in insulin sensitive cells. A specific focus is put on the action of S1P on hepatocytes, pancreatic ß-cells and skeletal muscle cells. In particular, modulation of S1P-signaling can be considered as a potential therapeutic target for the treatment of insulin resistance and type 2 diabetes.


Subject(s)
Diglycerides/metabolism , Insulin Resistance , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Ceramides/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Nonesterified/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Signal Transduction , Sphingosine/metabolism
15.
Cell Physiol Biochem ; 34(1): 27-44, 2014.
Article in English | MEDLINE | ID: mdl-24977479

ABSTRACT

Dendritic cells (DCs) are the cutting edge in innate and adaptive immunity. The major functions of these antigen-presenting cells are the capture, endosomal processing and presentation of antigens, providing them an exclusive ability to provoke adaptive immune responses and to induce and control tolerance. Immature DCs capture and process antigens, migrate towards secondary lymphoid organs where they present antigens to naive T cells in a well-synchronized sequence of procedures referred to as maturation. Indeed, recent research indicated that sphingolipids are modulators of essential steps in DC homeostasis. It has been recognized that sphingolipids not only modulate the development of DC subtypes from precursor cells but also influence functional activities of DCs such as antigen capture, and cytokine profiling. Thus, it is not astonishing that sphingolipids and sphingolipid metabolism play a substantial role in inflammatory diseases that are modulated by DCs. Here we highlight the function of sphingosine 1-phosphate (S1P) on DC homeostasis and the role of S1P and S1P metabolism in inflammatory diseases.


Subject(s)
Dendritic Cells/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Adaptive Immunity , Animals , Antigens/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Immunity, Innate , Skin Diseases/immunology , Skin Diseases/metabolism , Skin Diseases/pathology , Sphingosine/metabolism , Toll-Like Receptors/metabolism , Transcriptome
16.
Handb Exp Pharmacol ; (216): 355-72, 2013.
Article in English | MEDLINE | ID: mdl-23563666

ABSTRACT

Mammalian skin protects our body against external assaults due to a well-organized skin barrier. The formation of the skin barrier is a complex process, in which basal keratinocytes lose their mitotic activity and differentiate to corneocytes. These corneocytes are embedded in intercellular lipid lamellae composed of ceramides, cholesterol, fatty acids, and cholesterol esters. Ceramides are the dominant lipid molecules and their reduction is connected with a transepidermal water loss and an epidermal barrier dysfunction resulting in inflammatory skin diseases. Moreover, bioactive sphingolipid metabolites like ceramide-1-phosphate, sphingosylphosphorylcholine, and sphingosine-1-phosphate are also involved in the biological modulation of keratinocytes and immune cells of the skin. Therefore, it is not astonishing that a dysregulation of sphingolipid metabolism has been identified in inflammatory skin diseases such as atopic dermatitis and psoriasis vulgaris. This chapter will describe not only the specific sphingolipid species and their skin functions but also the dysregulation of sphingolipid metabolism in inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic/metabolism , Psoriasis/metabolism , Signal Transduction , Skin/metabolism , Sphingolipids/metabolism , Animals , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Humans , Langerhans Cells/metabolism , Permeability , Psoriasis/immunology , Psoriasis/pathology , Skin/immunology , Skin/pathology , Skin Absorption
17.
Article in English | MEDLINE | ID: mdl-32565314

ABSTRACT

The bioactive sphingolipid sphingosine 1-phosphate (S1P) has emerged in the last three decades as main regulator of key cellular processes including cell proliferation, survival, migration and differentiation. A crucial role for this sphingolipid has been recognized in skeletal muscle cell biology both in vitro and in vivo. S1P lyase (SPL) is responsible for the irreversible degradation of S1P and together with sphingosine kinases, the S1P producing enzymes, regulates cellular S1P levels. In this study is clearly showed that the blockade of SPL by pharmacological or RNA interference approaches induces myogenic differentiation of C2C12 myoblasts. Moreover, down-regulation of the specific S1P transporter spinster homolog 2 (Spns2) abrogates myogenic differentiation brought about by SPL inhibition or down-regulation, pointing at a role of extracellular S1P in the pro-myogenic action induced by SPL blockade. Furthermore, also S1P2 receptor down-regulation was found to abrogate the pro-myogenic effect of SPL blockade. These results provide further proof that inside-out S1P signaling is critically implicated in skeletal muscle biology and provide support to the concept that the specific targeting of SPL could represent an exploitable strategy to treat skeletal muscle disorders.


Subject(s)
Aldehyde-Lyases/metabolism , Anion Transport Proteins/metabolism , Cell Differentiation , Myoblasts/cytology , Sphingosine-1-Phosphate Receptors/metabolism , Aldehyde-Lyases/antagonists & inhibitors , Animals , Anion Transport Proteins/genetics , Cell Line , Mice , Sphingosine-1-Phosphate Receptors/genetics
18.
Nutrients ; 12(3)2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32183316

ABSTRACT

Recent research has linked sphingolipid (SL) metabolism with cystic fibrosis transmembrane conductance regulator (CFTR) activity, affecting bioactive lipid mediator sphingosine-1-phosphate (S1P). We hypothesize that loss of CFTR function in cystic fibrosis (CF) patients influenced plasma S1P levels. Total and unbound plasma S1P levels were measured in 20 lung-transplanted adult CF patients and 20 healthy controls by mass spectrometry and enzyme-linked immunosorbent assay (ELISA). S1P levels were correlated with CFTR genotype, routine laboratory parameters, lung function and pathogen colonization, and clinical symptoms. Compared to controls, CF patients showed lower unbound plasma S1P, whereas total S1P levels did not differ. A positive correlation of total and unbound S1P levels was found in healthy controls, but not in CF patients. Higher unbound S1P levels were measured in ΔF508-homozygous compared to ΔF508-heterozygous CF patients (p = 0.038), accompanied by higher levels of HDL in ΔF508-heterozygous patients. Gastrointestinal symptoms were more common in ΔF508 heterozygotes compared to ΔF508 homozygotes. This is the first clinical study linking plasma S1P levels with CFTR function and clinical presentation in adult CF patients. Given the emerging role of immunonutrition in CF, our study might pave the way for using S1P as a novel biomarker and nutritional target in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Heterozygote , Homozygote , Intestinal Diseases , Lung Transplantation , Lysophospholipids , Sphingosine/analogs & derivatives , Adult , Cystic Fibrosis/blood , Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Cystic Fibrosis/therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Female , Humans , Intestinal Diseases/blood , Intestinal Diseases/diet therapy , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Lung/immunology , Lung/metabolism , Lysophospholipids/blood , Lysophospholipids/immunology , Male , Middle Aged , Sphingosine/blood , Sphingosine/immunology
19.
Front Immunol ; 10: 1294, 2019.
Article in English | MEDLINE | ID: mdl-31231395

ABSTRACT

Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit.


Subject(s)
Cell Movement/immunology , Dendritic Cells , Measles virus/immunology , Measles , Respiratory Mucosa , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Humans , Measles/immunology , Measles/pathology , Measles/transmission , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
20.
Mol Metab ; 24: 1-17, 2019 06.
Article in English | MEDLINE | ID: mdl-31003944

ABSTRACT

OBJECTIVE: Aging is accompanied by loss of brown adipocytes and a decline in their thermogenic potential, which may exacerbate the development of adiposity and other metabolic disorders. Presently, only limited evidence exists describing the molecular alterations leading to impaired brown adipogenesis with aging and the contribution of these processes to changes of systemic energy metabolism. METHODS: Samples of young and aged murine brown and white adipose tissue were used to compare age-related changes of brown adipogenic gene expression and thermogenesis-related lipid mobilization. To identify potential markers of brown adipose tissue aging, non-targeted proteomic and metabolomic as well as targeted lipid analyses were conducted on young and aged tissue samples. Subsequently, the effects of several candidate lipid classes on brown adipocyte function were examined. RESULTS: Corroborating previous reports of reduced expression of uncoupling protein-1, we observe impaired signaling required for lipid mobilization in aged brown fat after adrenergic stimulation. Omics analyses additionally confirm the age-related impairment of lipid homeostasis and reveal the accumulation of specific lipid classes, including certain sphingolipids, ceramides, and dolichols in aged brown fat. While ceramides as well as enzymes of dolichol metabolism inhibit brown adipogenesis, inhibition of sphingosine 1-phosphate receptor 2 induces brown adipocyte differentiation. CONCLUSIONS: Our functional analyses show that changes in specific lipid species, as observed during aging, may contribute to reduced thermogenic potential. They thus uncover potential biomarkers of aging as well as molecular mechanisms that could contribute to the degradation of brown adipocytes, thereby providing potential treatment strategies of age-related metabolic conditions.


Subject(s)
Adipocytes, Brown/metabolism , Aging/metabolism , Lipid Metabolism , Adipocytes, Brown/cytology , Animals , Biomarkers/metabolism , Cells, Cultured , Ceramides/metabolism , Dolichols/metabolism , Male , Metabolome , Mice , Mice, Inbred C57BL , Proteome/genetics , Proteome/metabolism , Sphingolipids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL