Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Lipid Res ; : 100557, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38719152

ABSTRACT

Dietary sphingomyelin (SM) has been reported to favorably modulate postprandial lipemia. Mechanisms underlying these beneficial effects on cardiovascular risk markers are not fully elucidated. Rodent studies showed that tritiated SM was hydrolyzed in the intestinal lumen into ceramides (Cer), and further to sphingosine (SPH) and fatty acids (FA) that were absorbed by the intestine. Our objective was to investigate in Caco-2/TC7 cells cultured on semi-permeable inserts the uptake and metabolism of SPH and/or C23:0, the main FA of milk SM, as well as lipid secretion. Mixed micelles (MM) consisting of different digested lipids and taurocholate were prepared without or with SPH, SPH and C23:0 (SPH+C23:0) or C23:0. Triglycerides (TG) were quantified in the basolateral medium and sphingolipids were analyzed by tandem mass spectrometry. TG secretion increased 11-fold in all MM-incubated cells compared with lipid-free medium. Apical supply of SPH-enriched MM led to increased concentrations of total Cer in cells and co-addition of C23:0 in SPH-enriched MM led to a preferential increase of C23:0 Cer and C23:0 SM. Complementary experiments using deuterated SPH demonstrated that SPH-d9 was partly converted to sphingosine-1-phosphate-d9, Cer-d9 and SM-d9 within cells incubated with SPH-enriched MM. A few Cer-d9 (2% of added SPH-d9) was recovered in the basolateral medium of (MM+SPH)-incubated cells, especially C23:0 Cer-d9 in (MM+SPH+C23:0)-enriched cells. In conclusion, present results indicate that MM enriched with (SPH+C23:0), such as found in postprandial micelles formed after milk SM ingestion, impact directly sphingolipids endogenous metabolism in enterocytes, resulting in the secretion of TG-rich particles enriched with C23:0 Cer.

2.
J Nutr Biochem ; 117: 109334, 2023 07.
Article in English | MEDLINE | ID: mdl-36965784

ABSTRACT

The adaptive response to overfeeding is associated with profound modifications of gene expression in adipose tissue to support lipid storage and weight gain. The objective of this study was to assess in healthy lean men whether a supplementation with polyphenols could interact with these molecular adaptations. Abdominal subcutaneous adipose tissue biopsies were sampled from 42 subjects participating to an overfeeding protocol providing an excess of 50% of their total energy expenditure for 31 days, and who were supplemented with 2 g/day of grape polyphenols or a placebo. Gene expression profiling was performed by RNA sequencing. Overfeeding led to a modification of the expression of 163 and 352 genes in the placebo and polyphenol groups, respectively. The GO functions of these genes were mostly involved in lipid metabolism, followed by genes involved in adipose tissue remodeling and expansion. In response to overfeeding, 812 genes were differentially regulated between groups. Among them, a set of 41 genes were related to angiogenesis and were down-regulated in the polyphenol group. Immunohistochemistry targeting PECAM1, as endothelial cell marker, confirmed reduced angiogenesis in this group. Finally, quercetin and isorhamnetin, two polyphenol species enriched in the plasma of the volunteers submitted to the polyphenols, were found to inhibit human umbilical vein endothelial cells migration in vitro. Polyphenol supplementation do not prevent the regulation of genes related to lipid metabolism in human adipose tissue during overfeeding, but impact the angiogenesis pathways. This may potentially contribute to a protection against adipose tissue expansion during dynamic phase of weight gain.


Subject(s)
Vitis , Male , Humans , Endothelial Cells/metabolism , Adipose Tissue/metabolism , Obesity/metabolism , Weight Gain/physiology , Dietary Supplements , Polyphenols/pharmacology , Polyphenols/metabolism
3.
Biochimie ; 203: 11-19, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35817131

ABSTRACT

Elevated concentrations of triglyceride-rich lipoproteins (TGRL) in the fasting and postprandial states are risk factors for cardiovascular events, especially in type 2 diabetes (T2D). T2D modifies the lipid composition of plasma and lipoproteins and some sphingolipids (SP) have been validated as potent predictive biomarkers of cardiovascular disease occurrence. The main objectives of the present study were to characterize the plasma SP profile in fasting T2D patients and to determine whether SP are modified in postprandial TGRL from these patients compared to fasting TGRL. In a randomized parallel-group study, 30 T2D women ingested a breakfast including 20g lipids from either hazelnut cocoa palm oil-rich spread (Palm Nut) or Butter. Plasma was collected and TGRL were isolated by ultracentrifugation at fasting and 4h after the meal. Fasting samples of 6 control subjects from another cohort were analyzed for comparison. SP were analyzed by tandem mass spectrometry. Plasma from fasting T2D patients had higher ceramide (Cer) and ganglioside GM3 concentrations, and lower concentrations of sphingosylphosphorylcholine vs healthy subjects. In postprandial TGRL from T2D patients compared to those in the fasting state, Cer concentrations and especially C16:0, C24:1 and C24:0 molecular species, increased after the Palm Nut or Butter breakfast. A positive correlation was observed in the Palm Nut group between changes (Δ4h-fasting) of summed C16:0+C22:0+C24:1+C24:0 Cer concentrations in TGRL, and changes in plasma TG, TGRL-TG and TGRL-C16:0 concentrations. Altogether in T2D, the altered profile of plasma SP and the increased Cer concentrations in postprandial TGRL could contribute to the increased atherogenicity of TGRL.


Subject(s)
Butter , Diabetes Mellitus, Type 2 , Humans , Female , Palm Oil , Sphingolipids , Triglycerides/chemistry , Lipoproteins
4.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33857018

ABSTRACT

BACKGROUNDHigh circulating levels of ceramides (Cer) and sphingomyelins (SM) are associated with cardiometabolic diseases. The consumption of whole fat dairy products, naturally containing such polar lipids (PL), is associated with health benefits, but the impact on sphingolipidome remains unknown.METHODSIn a 4-week randomized controlled trial, 58 postmenopausal women daily consumed milk PL-enriched cream cheese (0, 3, or 5 g of milk PL). Postprandial metabolic explorations were performed before and after supplementation. Analyses included SM and Cer species in serum, chylomicrons, and feces. The ileal contents of 4 ileostomy patients were also explored after acute milk PL intake.RESULTSMilk PL decreased serum atherogenic C24:1 Cer, C16:1 SM, and C18:1 SM species (Pgroup < 0.05). Changes in serum C16+18 SM species were positively correlated with the reduction of cholesterol (r = 0.706), LDL-C (r = 0.666), and ApoB (r = 0.705) (P < 0.001). Milk PL decreased chylomicron content in total SM and C24:1 Cer (Pgroup < 0.001), parallel to a marked increase in total Cer in feces (Pgroup < 0.001). Milk PL modulated some specific SM and Cer species in both ileal efflux and feces, suggesting differential absorption and metabolization processes in the gut.CONCLUSIONMilk PL supplementation decreased atherogenic SM and Cer species associated with the improvement of cardiovascular risk markers. Our findings bring insights on sphingolipid metabolism in the gut, especially Cer, as signaling molecules potentially participating in the beneficial effects of milk PL.TRIAL REGISTRATIONClinicalTrials.gov, NCT02099032, NCT02146339.FUNDINGANR-11-ALID-007-01; PHRCI-2014: VALOBAB, no. 14-007; CNIEL; GLN 2018-11-07; HCL (sponsor).


Subject(s)
Ceramides , Lipid Metabolism/physiology , Milk , Postmenopause/metabolism , Sphingomyelins , Animals , Ceramides/analysis , Ceramides/blood , Ceramides/metabolism , Cheese , Diet , Feces/chemistry , Female , Glycolipids/metabolism , Glycoproteins/metabolism , Humans , Lipid Droplets/metabolism , Overweight , Sphingomyelins/analysis , Sphingomyelins/blood , Sphingomyelins/metabolism
5.
Cells ; 9(12)2020 11 25.
Article in English | MEDLINE | ID: mdl-33255741

ABSTRACT

Following a prolonged exposure to hypoxia-reoxygenation, a partial disruption of the ER-mitochondria tethering by mitofusin 2 (MFN2) knock-down decreases the Ca2+ transfer between the two organelles limits mitochondrial Ca2+ overload and prevents the Ca2+-dependent opening of the mitochondrial permeability transition pore, i.e., limits cardiomyocyte cell death. The impact of the metabolic changes resulting from the alteration of this Ca2+crosstalk on the tolerance to hypoxia-reoxygenation injury remains partial and fragmented between different field of expertise. >In this study, we report that MFN2 loss of function results in a metabolic switch driven by major modifications in energy production by mitochondria. During hypoxia, mitochondria maintain their ATP concentration and, concomitantly, the inner membrane potential by importing cytosolic ATP into mitochondria through an overexpressed ANT2 protein and by decreasing the expression and activity of the ATP hydrolase via IF1. This adaptation further blunts the detrimental hyperpolarisation of the inner mitochondrial membrane (IMM) upon re-oxygenation. These metabolic changes play an important role to attenuate cell death during a prolonged hypoxia-reoxygenation challenge.


Subject(s)
Adenine Nucleotide Translocator 2/metabolism , Adenosine Triphosphate/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Animals , Calcium/metabolism , Cell Death/physiology , Cell Line , Membrane Potential, Mitochondrial/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Myocytes, Cardiac/metabolism , Rats
6.
Biochimie ; 178: 26-38, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32659447

ABSTRACT

Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid (LBPA), is a phospholipid specifically enriched in the late endosome-lysosome compartment playing a crucial role for the fate of endocytosed components. Due to its presence in extracellular fluids during diseases associated with endolysosomal dysfunction, it is considered as a possible biomarker of disorders such as genetic lysosomal storage diseases and cationic amphiphilic drug-induced phospholipidosis. However, there is no true validation of this biomarker in human studies, nor a clear identification of the carrier of this endolysosome-specific lipid in biofluids. The present study demonstrates that in absence of any sign of renal failure, BMP, especially all docosahexaenoyl containing species, are significantly increased in the urine of patients treated with the antiarrhythmic drug amiodarone. Such urinary BMP increase could reflect a generalized drug-induced perturbation of the endolysosome compartment as observed in vitro with amiodarone-treated human macrophages. Noteworthy, BMP was associated with extracellular vesicles (EVs) isolated from human urines and extracellular medium of human embryonic kidney HEK293 cells and co-localizing with classical EV protein markers CD63 and ALIX. In the context of drug-induced endolysosomal dysfunction, increased BMP-rich EV release could be useful to remove excess of undigested material. This first human pilot study not only reveals BMP as a urinary biomarker of amiodarone-induced endolysosomal dysfunction, but also highlights its utility to prove the endosomal origin of EVs, also named as exosomes. This peculiar lipid already known as a canonical late endosome-lysosome marker, may be thus considered as a new lipid marker of urinary exosomes.


Subject(s)
Endosomes/chemistry , Endosomes/metabolism , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Lysophospholipids/metabolism , Monoglycerides/metabolism , Aged , Amiodarone/adverse effects , Animals , Anti-Arrhythmia Agents/adverse effects , Biomarkers/urine , Endosomes/drug effects , Extracellular Vesicles/drug effects , Female , HEK293 Cells , Humans , Kidney Diseases/chemically induced , Lysophospholipids/chemistry , Lysosomes/drug effects , Lysosomes/metabolism , Macrophages/chemistry , Macrophages/drug effects , Macrophages/metabolism , Male , Middle Aged , Monoglycerides/chemistry , Pilot Projects , Rats , THP-1 Cells
7.
Food Chem ; 329: 126927, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32516717

ABSTRACT

Donor human milk, pasteurised for safety reasons, is the first alternative for feeding preterm infants when mothers' own milk is unavailable. Breastmilk pasteurisation impact on lipid digestion and absorption was evaluated by a static in vitro digestion model for preterm infants coupled with intestinal absorption using Caco-2/TC7 cells. Lipid absorption was quantified by digital image analysis of lipid droplets, by measurement of basolateral triglyceride concentration and by analysing the expression of major genes involved. After in vitro digestion, lipolysis extent was 13% lower in pasteurised human milk (PHM) than in raw human milk (RHM). In Caco-2/TC7 cells, the number of lipid droplets was identical for both milk types, while the mean droplet area was 17% smaller with PHM. Altogether, pasteurisation decreased the pre-lipolysis of human milk. This initial difference in free fatty acid amount was only partially buffered by the subsequent processes of in vitro digestion and cellular lipid absorption.


Subject(s)
Lipids/chemistry , Milk, Human/chemistry , Cell Line , Digestion , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa , Intestines , Lipolysis , Pasteurization
8.
Chem Phys Lipids ; 225: 104815, 2019 12.
Article in English | MEDLINE | ID: mdl-31494102

ABSTRACT

Dietary fats are present in the diet under different types of structures, such as spread vs emulsions (notably in processed foods and enteral formula), and interest is growing regarding their digestion and intestinal absorption. In clinical trials, there is often a need to add stable isotope-labeled triacylglycerols (TAGs) as tracers to the ingested fat in order to track its intestinal absorption and further metabolic fate. Because most TAG tracers contain saturated fatty acids, they may modify the physicochemical properties of the ingested labeled fat and thereby its digestion. However, the actual impact of tracer addition on fat crystalline properties and lipolysis by digestive lipases still deserves to be explored. In this context, we monitored the thermal and polymorphic behavior of anhydrous milk fat (AMF) enriched in homogeneous TAGs tracers and further compared it with the native AMF using differential scanning calorimetry and power X-ray diffraction. As tracers, we used a mixture of tripalmitin, triolein and tricaprylin at 2 different concentrations (1.5 and 5.7 wt%, which have been used in clinical trials). The addition of TAG tracers modified the AMF melting profile, especially at the highest tested concentration (5.7 wt%). Both AMF and AMF enriched with 1.5 wt% tracers were completely melted around 37 °C, i.e. close to the body temperature, while the AMF enriched with 5.7 wt% tracers remained partially crystallized at this temperature. Similar trends were observed in both bulk and emulsified systems. Moreover, the kinetics of AMF polymorphic transformation was modified in the presence of tracers. While only ß' form was observed in the native AMF, the ß-form was clearly detected in the AMF containing 5.7 wt% tracers. We further tested the impact of tracers on the lipolysis of AMF in bulk using a static in vitro model of duodenal digestion. Lipolysis of AMF enriched with 5.7 wt% tracers was delayed compared with that of AMF and AMF enriched with 1.5 wt% tracers. Therefore, low amounts of TAG tracers including tripalmitin do not have a high impact on fat digestion, but one has to be cautious when using higher amounts of these tracers.


Subject(s)
Dietary Fats , Temperature , Triglycerides/chemistry , Lipolysis , Molecular Structure
9.
Mol Nutr Food Res ; 63(4): e1801078, 2019 02.
Article in English | MEDLINE | ID: mdl-30628158

ABSTRACT

SCOPE: Enhanced adiposity and metabolic inflammation are major features of obesity associated with altered gut microbiota and intestinal barrier. How these metabolic outcomes can be impacted by milk polar lipids (MPL), naturally containing 25% of sphingomyelin, is investigated in mice fed a mixed high-fat (HF) diet . METHODS AND RESULTS: Male C57Bl/6 mice receive a HF-diet devoid of MPL (21% fat, mainly palm oil, in chow), or supplemented with 1.1% or 1.6% of MPL (HF-MPL1; HF-MPL2) via a total-lipid extract from butterserum concentrate for 8 weeks. HF-MPL2 mice gain less weight versus HF (p < 0.01). Diets do not impact plasma markers of inflammation but in the liver, HF-MPL2 tends to decrease hepatic gene expression of macrophage marker F4/80 versus HF-MPL1 (p = 0.06). Colonic crypt depth is the maximum in HF-MPL2 (p < 0.05). In cecal microbiota, HF-MPL1 increases Bifidobacterium animalis versus HF (p < 0.05). HF-MPL2 decreases Lactobacillus reuteri (p < 0.05), which correlates negatively with the fecal loss of milk sphingomyelin-specific fatty acids (p < 0.05). CONCLUSION: In mice fed a mixed HF diet, MPL can limit HF-induced body weight gain and modulate gut physiology and the abundance in microbiota of bacteria of metabolic interest. This supports further exploration of how residual unabsorbed lipids reaching the colon can impact HF-induced metabolic disorders.


Subject(s)
Fatty Acids/metabolism , Gastrointestinal Microbiome/drug effects , Lipids/pharmacology , Milk/chemistry , Animals , Diet, High-Fat , Fatty Acids/analysis , Feces , Intestinal Absorption , Lipids/administration & dosage , Lipids/analysis , Lipids/chemistry , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Male , Mice, Inbred C57BL , Sphingomyelins/pharmacology , Weight Gain/drug effects
10.
J Nutr Biochem ; 65: 128-138, 2019 03.
Article in English | MEDLINE | ID: mdl-30685581

ABSTRACT

Milk polar lipids (MPL) are specifically rich in milk sphingomyelin (MSM) which represents 24% of MPL. Beneficial effects of MPL or MSM have been reported on lipid metabolism, but information on gut physiology is scarce. Here we assessed whether MPL and MSM can impact tight junction expression. Human epithelial intestinal Caco-2/TC7 cells were incubated with mixed lipid micelles devoid of MSM (Control) or with 0.2 or 0.4 mM of MSM via pure MSM or via total MPL. C57Bl/6 mice received 5 or 10 mg of MSM via MSM or via MPL (oral gavage); small intestinal segments were collected after 4 h. Impacts on tight junction and cytokine expressions were assessed by qPCR; IL-8 and IL-8 murine homologs (Cxcl1, Cxcl2) were analyzed. In vitro, MSM increased tight junction expression (Occludin, ZO-1) vs Control, unlike MPL. However, no differences were observed in permeability assays (FITC-dextran, Lucifer yellow). MSM increased the secretion and gene expression of IL-8 but not of other inflammatory cytokines. Moreover, cell incubation with IL-8 induced an overexpression of tight junction proteins. In mice, mRNA level of Cxcl1 and Cxcl2 in the ileum were increased after gavage with MSM vs NaCl but not with MPL. Altogether, these results suggest a specific action of MSM on intestinal tight junction expression, possibly mediated by IL-8. Our study provides clues to shed light on the beneficial effects of MPL on intestinal functions and supports the need for further mechanistic exploration of the direct vs indirect effects of MSM and IL-8 on the gut barrier.


Subject(s)
Interleukin-8/metabolism , Lipids/pharmacology , Milk/chemistry , Tight Junctions/metabolism , Animals , Caco-2 Cells , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Intestines/cytology , Lipids/chemistry , Male , Mice, Inbred C57BL , Sphingomyelins/administration & dosage , Sphingomyelins/pharmacology , Tight Junction Proteins/genetics
11.
J Nutr Biochem ; 43: 116-124, 2017 05.
Article in English | MEDLINE | ID: mdl-28284063

ABSTRACT

Obesity and type 2 diabetes are nutritional pathologies, characterized by a subclinical inflammatory state. Endotoxins are now well recognized as an important factor implicated in the onset and maintain of this inflammatory state during fat digestion in high-fat diet. As a preventive strategy, lipid formulation could be optimized to limit these phenomena, notably regarding fatty acid profile and PL emulsifier content. Little is known about soybean polar lipid (SPL) consumption associated to oils rich in saturated FA vs. anti-inflammatory omega-3 FA such as α-linolenic acid on inflammation and metabolic endotoxemia. We then investigated in mice the effect of different synthetic diets enriched with two different oils, palm oil or flaxseed oil and containing or devoid of SPL on adipose tissue inflammation and endotoxin receptors. In both groups containing SPL, adipose tissue (WAT) increased compared with groups devoid of SPL and an induction of MCP-1 and LBP was observed in WAT. However, only the high-fat diet in which flaxseed oil was associated with SPL resulted in both higher WAT inflammation and higher circulating sCD14 in plasma. In conclusion, we have demonstrated that LPS transporters LBP and sCD14 and adipose tissue inflammation can be modulated by SPL in high fat diets differing in oil composition. Notably high-flaxseed oil diet exerts a beneficial metabolic impact, however blunted by PL addition. Our study suggests that nutritional strategies can be envisaged by optimizing dietary lipid sources in manufactured products, including fats/oils and polar lipid emulsifiers, in order to limit the inflammatory impact of palatable foods.


Subject(s)
Acute-Phase Proteins/metabolism , Carrier Proteins/metabolism , Glycine max/chemistry , Linseed Oil/pharmacology , Membrane Glycoproteins/metabolism , Palm Oil/pharmacology , Panniculitis/etiology , Animals , Diet, High-Fat , Dietary Supplements , Fatty Acids/analysis , Lipopolysaccharide Receptors/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL
12.
Mol Nutr Food Res ; 60(3): 609-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26592505

ABSTRACT

SCOPE: Enhanced adiposity and metabolic inflammation are major features of obesity that could be impacted by dietary emulsifiers. We investigated in high-fat fed mice the effects of using a new polar lipid (PL) emulsifier from milk (MPL) instead of soybean lecithin (soybean PL [SPL]) on adipose tissue and intestinal mucosa function. METHODS AND RESULTS: Four groups of C57BL6 mice received for 8 wks a low-fat (LF) diet or a high-fat diet devoid of PLs or an high-fat diet including MPL (high-fat-MPL) or SPL (high-fat-SPL). Compared with high-fat diet, high-fat-SPL diet increased white adipose tissue (WAT) mass (p < 0.05), with larger adipocytes (p < 0.05) and increased expression of tumor necrosis factor alpha, monochemoattractant protein-1, LPS-binding protein, and leptin (p < 0.05). This was not observed with high-fat-MPL diet despite similar dietary intakes and increased expression of fatty acid transport protein 4 and microsomal TG transfer protein, involved in lipid absorption, in upper intestine (p < 0.05). High-fat-MPL mice had a lower expression in WAT of cluster of differentiation 68, marker of macrophage infiltration, versus high-fat and high-fat-SPL mice (p < 0.05), and more goblet cells in the colon (p < 0.05). CONCLUSIONS: Unlike SPL, MPL in the high-fat diet did not induce WAT hypertrophy and inflammation but increased colonic goblet cells. This supports further clinical exploration of different sources of dietary emulsifiers in the frame of obesity outbreak.


Subject(s)
Colon/drug effects , Emulsifying Agents/pharmacology , Glycine max/chemistry , Goblet Cells/drug effects , Milk/chemistry , Adipose Tissue, White/drug effects , Adiposity/drug effects , Animals , Caco-2 Cells/drug effects , Colon/cytology , Diet, Fat-Restricted , Diet, High-Fat/adverse effects , Humans , Lecithins/chemistry , Lecithins/pharmacology , Lipids/analysis , Lipids/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Panniculitis/chemically induced , Panniculitis/metabolism
13.
Diabetologia ; 59(3): 614-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660890

ABSTRACT

AIMS/HYPOTHESIS: Mitochondria-associated endoplasmic reticulum membranes (MAMs) are regions of the endoplasmic reticulum (ER) tethered to mitochondria and controlling calcium (Ca(2+)) transfer between both organelles through the complex formed between the voltage-dependent anion channel, glucose-regulated protein 75 and inositol 1,4,5-triphosphate receptor (IP3R). We recently identified cyclophilin D (CYPD) as a new partner of this complex and demonstrated a new role for MAMs in the control of insulin's action in the liver. Here, we report on the mechanisms by which disruption of MAM integrity induces hepatic insulin resistance in CypD (also known as Ppif)-knockout (KO) mice. METHODS: We used either in vitro pharmacological and genetic inhibition of CYPD in HuH7 cells or in vivo loss of CYPD in mice to investigate ER-mitochondria interactions, inter-organelle Ca(2+) exchange, organelle homeostasis and insulin action. RESULTS: Pharmacological and genetic inhibition of CYPD concomitantly reduced ER-mitochondria interactions, inhibited inter-organelle Ca(2+) exchange, induced ER stress and altered insulin signalling in HuH7 cells. In addition, histamine-stimulated Ca(2+) transfer from ER to mitochondria was blunted in isolated hepatocytes of CypD-KO mice and this was associated with an increase in ER calcium store. Interestingly, disruption of inter-organelle Ca(2+) transfer was associated with ER stress, mitochondrial dysfunction, lipid accumulation, activation of c-Jun N-terminal kinase (JNK) and protein kinase C (PKC)ε and insulin resistance in liver of CypD-KO mice. Finally, CYPD-related alterations of insulin signalling were mediated by activation of PKCε rather than JNK in HuH7 cells. CONCLUSIONS/INTERPRETATION: Disruption of IP3R-mediated Ca(2+) signalling in the liver of CypD-KO mice leads to hepatic insulin resistance through disruption of organelle interaction and function, increase in lipid accumulation and activation of PKCε. Modulation of ER-mitochondria Ca(2+) exchange may thus provide an exciting new avenue for treating hepatic insulin resistance.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Insulin Resistance/physiology , Mitochondria/metabolism , Animals , Cell Line , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , Hepatocytes/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout
14.
Diabetes ; 64(6): 2254-64, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25552598

ABSTRACT

Imeglimin is the first in a new class of oral glucose-lowering agents currently in phase 2b development. Although imeglimin improves insulin sensitivity in humans, the molecular mechanisms are unknown. This study used a model of 16-week high-fat, high-sucrose diet (HFHSD) mice to characterize its antidiabetic effects. Six-week imeglimin treatment significantly decreased glycemia, restored normal glucose tolerance, and improved insulin sensitivity without modifying organs, body weights, and food intake. This was associated with an increase in insulin-stimulated protein kinase B phosphorylation in the liver and muscle. In liver mitochondria, imeglimin redirects substrate flows in favor of complex II, as illustrated by increased respiration with succinate and by the restoration of respiration with glutamate/malate back to control levels. In addition, imeglimin inhibits complex I and restores complex III activities, suggesting an increase in fatty acid oxidation, which is supported by an increase in hepatic 3-hydroxyacetyl-CoA dehydrogenase activity and acylcarnitine profile and the reduction of liver steatosis. Imeglimin also reduces reactive oxygen species production and increases mitochondrial DNA. Finally, imeglimin effects on mitochondrial phospholipid composition could participate in the benefit of imeglimin on mitochondrial function. In conclusion, imeglimin normalizes glucose tolerance and insulin sensitivity by preserving mitochondrial function from oxidative stress and favoring lipid oxidation in liver of HFHSD mice.


Subject(s)
Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Triazines/therapeutic use , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL
15.
PLoS One ; 9(10): e110653, 2014.
Article in English | MEDLINE | ID: mdl-25337938

ABSTRACT

Inflammation is now recognized as a major factor contributing to type 2 diabetes (T2D). However, while the mechanisms and consequences associated with white adipose tissue inflammation are well described, very little is known concerning the situation in skeletal muscle. The aim of this study was to investigate, in vitro and in vivo, how skeletal muscle inflammation develops and how in turn it modulates local and systemic insulin sensitivity in different mice models of T2D and in humans, focusing on the role of the chemokine MCP1. Here, we found that skeletal muscle inflammation and macrophage markers are increased and associated with insulin resistance in mice models and humans. In addition, we demonstrated that intra-muscular TNFα expression is exclusively restricted to the population of intramuscular leukocytes and that the chemokine MCP1 was associated with skeletal muscle inflammatory markers in these models. Furthermore, we demonstrated that exposure of C2C12 myotubes to palmitate elevated the production of the chemokine MCP1 and that the muscle-specific overexpression of MCP1 in transgenic mice induced the local recruitment of macrophages and altered local insulin sensitivity. Overall our study demonstrates that skeletal muscle inflammation is clearly increased in the context of T2D in each one of the models we investigated, which is likely consecutive to the lipotoxic environment generated by peripheral insulin resistance, further increasing MCP1 expression in muscle. Consequently, our results suggest that MCP1-mediated skeletal muscle macrophages recruitment plays a role in the etiology of T2D.


Subject(s)
Chemokine CCL2/physiology , Insulin Resistance/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Myositis/immunology , Animals , Cell Line , Cell Movement , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis/metabolism
16.
Circulation ; 128(14): 1555-65, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23983249

ABSTRACT

BACKGROUND: Under physiological conditions, Ca(2+) transfer from the endoplasmic reticulum (ER) to mitochondria might occur at least in part at contact points between the 2 organelles and involves the VDAC1/Grp75/IP3R1 complex. Accumulation of Ca(2+) into the mitochondrial matrix may activate the mitochondrial chaperone cyclophilin D (CypD) and trigger permeability transition pore opening, whose role in ischemia/reperfusion injury is well recognized. We questioned here whether the transfer of Ca(2+) from ER to mitochondria might play a role in cardiomyocyte death after hypoxia-reoxygenation. METHODS AND RESULTS: We report that CypD interacts with the VDAC1/Grp75/IP3R1 complex in cardiomyocytes. Genetic or pharmacological inhibition of CypD in both H9c2 cardiomyoblasts and adult cardiomyocytes decreased the Ca(2+) transfer from ER to mitochondria through IP3R under normoxic conditions. During hypoxia-reoxygenation, the interaction between CypD and the IP3R1 Ca(2+) channeling complex increased concomitantly with mitochondrial Ca(2+) content. Inhibition of either CypD, IP3R1, or Grp75 decreased protein interaction within the complex, attenuated mitochondrial Ca(2+) overload, and protected cells from hypoxia-reoxygenation. Genetic or pharmacological inhibition of CypD provided a similar effect in adult mice cardiomyocytes. Disruption of ER-mitochondria interaction via the downregulation of Mfn2 similarly reduced the interaction between CypD and the IP3R1 complex and protected against hypoxia-reoxygenation injury. CONCLUSIONS: Our data (1) point to a new role of CypD at the ER-mitochondria interface and (2) suggest that decreasing ER-mitochondria interaction at reperfusion can protect cardiomyocytes against lethal reperfusion injury through the reduction of mitochondrial Ca(2+) overload via the CypD/VDAC1/Grp75/IP3R1 complex.


Subject(s)
Calcium Signaling/physiology , Cell Hypoxia/physiology , Endoplasmic Reticulum/physiology , Mitochondria, Heart/physiology , Myocytes, Cardiac/pathology , Oxygen/toxicity , Animals , Cell Line , Cells, Cultured/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Cyclophilins/physiology , HSP70 Heat-Shock Proteins/physiology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intracellular Membranes/physiology , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Patch-Clamp Techniques , Random Allocation , Rats , Voltage-Dependent Anion Channel 1/physiology
17.
Basic Res Cardiol ; 108(4): 363, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744057

ABSTRACT

Preventing cyclophilin D (cypD) translocation to the inner mitochondrial membrane can limit lethal reperfusion injury through the inhibition of the opening of the mitochondrial permeability transition pore. Inhibition or loss of function of cypD may also result into an endoplasmic reticulum (ER) stress that has been shown to alter cell survival. We therefore questioned whether ER stress might play a role in the protection induced by CypD deficiency or inhibition. CypD-KO and NIM811 (a CypD inhibitor)-treated mice were subjected to a prolonged ischemia-reperfusion (I/R). Area at risk and infarct size was measured using blue dye and triphenyltetrazolium chloride staining. ER stress markers were measured in the hearts during the reperfusion phase. As expected, cypD-KO mice exhibited a decreased infarct size when compared to wild-type mice (8 ± 1 vs. 20 ± 4% of left ventricular weight; p < 0.01). CypD-deficient mice displayed an increased expression of ER stress proteins such as eukaryotic initiation factor 2α (eIF2α) or glucose regulated protein 78 (Grp78 or Bip). The ER stress inhibitor TUDCA prevented the infarct size reduction afforded by the loss of cypD function (mean infarct size averaged 21 ± 4% of LV weight, p < 0.01 vs. cypD-KO). Similar results were obtained when NIM811, an analog of cyclosporine A, was used to pharmacologically (instead of genetically) inhibit cypD function. This study suggests that the ER stress induced by the inhibition of cypD function plays a key role in protecting the heart against lethal ischemia-reperfusion injury.


Subject(s)
Cyclophilins/antagonists & inhibitors , Endoplasmic Reticulum Stress/physiology , Heart/physiopathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Cyclophilins/metabolism , Cyclosporine/pharmacology , Cyclosporins/pharmacology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heart/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Taurochenodeoxycholic Acid/pharmacology
18.
Biochem Biophys Res Commun ; 417(1): 439-45, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22177958

ABSTRACT

Endoplasmic reticulum (ER) stress is proposed as a novel link between elevated fatty acids levels, obesity and insulin resistance in liver and adipose tissue. However, it is unknown whether ER stress also contributes to lipid-induced insulin resistance in skeletal muscle, the major tissue responsible of insulin-stimulated glucose disposal. Here, we investigated the possible role of ER stress in palmitate-induced alterations of insulin action, both in vivo, in gastrocnemius of high-palm diet fed mice, and in vitro, in palmitate-treated C(2)C(12) myotubes. We demonstrated that 8 weeks of high-palm diet increased the expression of ER stress markers in muscle of mice, whereas ex-vivo insulin-stimulated PKB phosphorylation was not altered in this tissue. In addition, exposure of C(2)C(12) myotubes to either tuncamycine or palmitate induced ER stress and altered insulin-stimulated PKB phosphorylation. However, alleviation of ER stress by either TUDCA or 4-PBA treatments, or by overexpressing Grp78, did not restore palmitate-induced reduction of insulin-stimulated PKB phosphorylation in C(2)C(12) myotubes. This work highlights that, even ER stress is associated with palmitate-induced alterations of insulin signaling, ER stress is likely not the major culprit of this effect in myotubes, suggesting that the previously proposed link between ER stress and insulin resistance is less important in skeletal muscle than in adipose tissue and liver.


Subject(s)
Diet/adverse effects , Dietary Fats/adverse effects , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/biosynthesis , Insulin Resistance , Muscle Fibers, Skeletal/physiology , Palmitates/adverse effects , Animals , Butylamines/pharmacology , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Heat-Shock Proteins/genetics , Insulin/metabolism , Insulin/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Palmitates/administration & dosage , Palmitates/pharmacology , Taurochenodeoxycholic Acid/pharmacology , Tunicamycin/pharmacology
19.
J Physiol Biochem ; 67(3): 285-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21267801

ABSTRACT

Mitochondria have been shown to be impaired in insulin resistance-related diseases but have not been extensively studied during the first steps of adipose cell development. This study was designed to determine the sequence of changes of the mitochondrial network and function during the first days of adipogenesis. 3T3-L1 preadipocytes were differentiated into adipocytes without using glitazone compounds. At days 0, 3, 6, 9, and 12, mitochondrial network imaging, mitochondrial oxygen consumption, membrane potential, and oxidative phosphorylation efficiency were assessed in permeabilized cells. Gene and protein expressions related to fatty acid metabolism and mitochondrial network were also determined. Compared to preadipocytes (day 0), new adipocytes (days 6 and 9) displayed profound changes of their mitochondrial network that underwent fragmentation and redistribution around lipid droplets. Drp1 and mitofusin 2 displayed a progressive increase in their gene expression and protein content during the first 9 days of differentiation. In parallel with the mitochondrial network redistribution, mitochondria switched to uncoupled respiration with a tendency towards decreased membrane potential, with no variation of mtTFA and NRF1 gene expression. The expression of PGC1α and NRF2 genes and genes involved in lipid oxidation (UCP2, CD36, and CPT1) was increased. Reactive oxygen species (ROS) production displayed a nadir at day 6 with a concomitant increase in antioxidant enzyme gene expression. This 3T3-L1-based in vitro model of adipogenesis showed that mitochondria adapted to the increased number of lipid droplets by network redistribution and uncoupling respiration. The timing and regulation of lipid oxidation-associated ROS production appeared to play an important role in these changes.


Subject(s)
Adipocytes/physiology , Cell Differentiation , Mitochondria/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis , Adiponectin/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Citrate (si)-Synthase/metabolism , Electron Transport Complex IV/metabolism , Enzyme Assays , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Gene Expression , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial , Mice , Microscopy, Video , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Oxygen Consumption , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
20.
Am J Physiol Endocrinol Metab ; 300(3): E581-91, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21224483

ABSTRACT

Reactive oxygen species (ROS) have been widely implicated in the pathogenesis of diabetes and more recently in mitochondrial alterations in skeletal muscle of diabetic mice. However, so far the exact sources of ROS in skeletal muscle have remained elusive. Aiming at better understanding the causes of mitochondrial alterations in diabetic muscle, we designed this study to characterize the sites of ROS production in skeletal muscle of streptozotocin (STZ)-induced diabetic mice. Hyperglycemic STZ mice showed increased markers of systemic and muscular oxidative stress, as evidenced by increased circulating H(2)O(2) and muscle carbonylated protein levels. Interestingly, insulin treatment reduced hyperglycemia and improved systemic and muscular oxidative stress in STZ mice. We demonstrated that increased oxidative stress in muscle of STZ mice is associated with an increase of xanthine oxidase (XO) expression and activity and is mediated by an induction of H(2)O(2) production by both mitochondria and XO. Finally, treatment of STZ mice, as well as high-fat and high-sucrose diet-fed mice, with oxypurinol reduced markers of systemic and muscular oxidative stress and prevented structural and functional mitochondrial alterations, confirming the in vivo relevance of XO in ROS production in diabetic mice. These data indicate that mitochondria and XO are the major sources of hyperglycemia-induced ROS production in skeletal muscle and that the inhibition of XO reduces oxidative stress and improves mitochondrial alterations in diabetic muscle.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/complications , Mitochondria, Muscle/physiology , Muscle, Skeletal/metabolism , Oxidative Stress/physiology , Xanthine Oxidase/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Animals , Antioxidants/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Diet , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Oxypurinol/pharmacology , Protein Carbonylation/drug effects , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...