Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38843936

ABSTRACT

Lipid composition is conserved within sub-cellular compartments to maintain cell function. Lipidomic analyses of liver, muscle, white and brown adipose tissue (BAT) mitochondria revealed substantial differences in their glycerophospholipid (GPL) and free cholesterol (FC) contents. The GPL to FC ratio was 50-fold higher in brown than white adipose tissue mitochondria. Their purity was verified by comparison of proteomes with ER and mitochondria-associated membranes. A lipid signature containing PC and FC, calculated from the lipidomic profiles, allowed differentiation of mitochondria from BAT of mice housed at different temperatures. Elevating FC in BAT mitochondria prevented uncoupling protein (UCP) 1 function, whereas increasing GPL boosted it. Similarly, STARD3 overexpression facilitating mitochondrial FC import inhibited UCP1 function in primary brown adipocytes, whereas a knockdown promoted it. We conclude that the mitochondrial GPL/FC ratio is key for BAT function and propose that targeting it might be a promising strategy to promote UCP1 activity.


Subject(s)
Adipose Tissue, Brown , Cholesterol , Lipidomics , Mitochondria , Uncoupling Protein 1 , Animals , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Mice , Adipose Tissue, Brown/metabolism , Cholesterol/metabolism , Mitochondria/metabolism , Lipidomics/methods , Organ Specificity , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Glycerophospholipids/metabolism , Male , Lipid Metabolism
2.
iScience ; 27(5): 109774, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38711443

ABSTRACT

Ferroptosis is a cell death pathway that can be promoted by peroxidizable polyunsaturated fatty acids in cancer cells. Here, we investigated the mechanisms underlying the toxicity of punicic acid (PunA), an isomer of conjugated linolenic acids (CLnAs) bearing three conjugated double bonds highly prone to peroxidation, on prostate cancer (PCa) cells. PunA induced ferroptosis in PCa cells and triggered massive lipidome remodeling, more strongly in PC3 androgen-negative cells than in androgen-positive cells. The greater sensitivity of androgen-negative cells to PunA was associated with lower expression of glutathione peroxidase 4 (GPX4). We then identified the phospholipase PLA2G7 as a PunA-induced ferroptosis suppressor in PCa cells. Overexpressing PLA2G7 decreased lipid peroxidation levels, suggesting that PLA2G7 hydrolyzes hydroperoxide-containing phospholipids, thus preventing ferroptosis. Importantly, overexpressing both PLA2G7 and GPX4 strongly prevented PunA-induced ferroptosis in androgen-negative PCa cells. This study shows that PLA2G7 acts complementary to GPX4 to protect PCa cells from CLnA-induced ferroptosis.

3.
EXCLI J ; 22: 207-220, 2023.
Article in English | MEDLINE | ID: mdl-36998704

ABSTRACT

There is general consent that with decreasing bone mineral density the amount of marrow adipose tissue increases. While image-based techniques, claim an increase in saturated fatty acids responsible for this effect, this study shows an increase in both saturated and unsaturated fatty acids in the bone marrow. Using fatty acid methyl ester gas chromatography-mass spectrometry, characteristic fatty acid patterns for patients with normal BMD (N = 9), osteopenia (N = 12), and osteoporosis (N = 9) have been identified, which differ between plasma, red bone marrow and yellow bone marrow. Selected fatty acids, e.g. FA10:0, FA14:1, or FA16:1 n-7 in the bone marrow or FA18:0, FA18:1 n-9, FA18:1 n-7, FA20:0, FA20:1 n-9, or FA20:3 n-6 in the plasma, correlated with osteoclast activity, suggesting a possible mechanism how these fatty acids may interfere with BMD. Although several fatty acids correlated well with the osteoclast activity and BMD, there was not a single fatty acid contained in our fatty acid profile that can be claimed for controlling BMD, a fact that may be attributed to the genetic heterogeneity of the patients.

4.
J Hepatol ; 78(4): 820-835, 2023 04.
Article in English | MEDLINE | ID: mdl-36681162

ABSTRACT

BACKGROUND & AIMS: Hepatocyte growth and proliferation depends on membrane phospholipid biosynthesis. Short-chain fatty acids (SCFAs) generated by bacterial fermentation, delivered through the gut-liver axis, significantly contribute to lipid biosynthesis. We therefore hypothesized that dysbiotic insults like antibiotic treatment not only affect gut microbiota, but also impair hepatic lipid synthesis and liver regeneration. METHODS: Stable isotope labeling and 70% partial hepatectomy (PHx) was carried out in C57Bl/6J wild-type mice, in mice treated with broad-spectrum antibiotics, in germ-free mice and mice colonized with minimal microbiota. The microbiome was analyzed by 16S rRNA gene sequencing and microbial culture. Gut content, liver, blood and primary hepatocyte organoids were tested by mass spectrometry-based lipidomics, quantitative reverse-transcription PCR (qRT-PCR), immunoblot and immunohistochemistry for expression of proliferative and lipogenic markers. Matched biopsies from hyperplastic and hypoplastic liver tissue of patients subjected to surgical intervention to induce hyperplasia were analyzed by qRT-PCR for lipogenic enzymes. RESULTS: Three days of antibiotic treatment induced persistent dysbiosis with significantly decreased beta-diversity and richness, but a massive increase of Proteobacteria, accompanied by decreased colonic SCFAs. After PHx, antibiotic-treated mice showed delayed liver regeneration, increased mortality, impaired hepatocyte proliferation and decreased hepatic phospholipid synthesis. Expression of the lipogenic enzyme SCD1 was upregulated after PHx but delayed by antibiotic treatment. Germ-free mice essentially recapitulated the phenotype of antibiotic treatment. Phospholipid biosynthesis, hepatocyte proliferation, liver regeneration and survival were rescued in gnotobiotic mice colonized with a minimal SCFA-producing microbial community. SCFAs induced the growth of murine hepatocyte organoids and hepatic SCD1 expression in mice. Further, SCD1 was required for proliferation of human hepatoma cells and was associated with liver regeneration in human patients. CONCLUSION: Gut microbiota are pivotal for hepatic membrane phospholipid biosynthesis and liver regeneration. IMPACT AND IMPLICATIONS: Gut microbiota affect hepatic lipid metabolism through the gut-liver axis, but the underlying mechanisms are poorly understood. Perturbations of the gut microbiome, e.g. by antibiotics, impair the production of bacterial metabolites, which normally serve as building blocks for membrane lipids in liver cells. As a consequence, liver regeneration and survival after liver surgery is severely impaired. Even though this study is preclinical, its results might allow physicians in the future to improve patient outcomes after liver surgery, by modulation of gut microbiota or their metabolites.


Subject(s)
Cell Membrane , Gastrointestinal Microbiome , Hepatocytes , Liver Regeneration , Phospholipids , Animals , Humans , Mice , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Hyperplasia/metabolism , Hyperplasia/pathology , Liver/pathology , Liver Regeneration/physiology , Mice, Inbred C57BL , Phospholipids/biosynthesis , Phospholipids/metabolism , RNA, Ribosomal, 16S , Hepatocytes/metabolism , Cell Membrane/metabolism
5.
Nutrients ; 14(15)2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35893909

ABSTRACT

The content of polyunsaturated fatty acids (PUFA) in complex lipids essentially influences their physicochemical properties and has been linked to health and disease. To investigate the incorporation of dietary PUFA in the human plasma lipidome, we quantified glycerophospholipids (GPL), sphingolipids, and sterols using electrospray ionization coupled to tandem mass spectrometry of plasma samples obtained from a dietary intervention study. Healthy individuals received foods supplemented with different vegetable oils rich in PUFA. These included sunflower, linseed, echium, and microalgae oil as sources of linoleic acid (LA; FA 18:2 n-6), alpha-linolenic acid (ALA; FA 18:3 n-3), stearidonic acid (SDA; FA 18:4 n-3), and docosahexaenoic acid (DHA; FA 22:6 n-3). While LA and ALA did not influence the species profiles of GPL, sphingolipid, and cholesteryl ester drastically, SDA and DHA were integrated primarily in ethanolamine-containing GPL. This significantly altered phosphatidylethanolamine and plasmalogen species composition, especially those with 38-40 carbons and 6 double bonds. We speculate that diets enriched with highly unsaturated FA more efficiently alter plasma GPL acyl chain composition than those containing primarily di- and tri-unsaturated FA, most likely because of their more pronounced deviation of FA composition from typical western diets.


Subject(s)
Fatty Acids, Omega-3 , Lipidomics , Diet , Docosahexaenoic Acids/metabolism , Ethanolamines , Fatty Acids, Omega-3/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Linoleic Acid , Phosphatidylethanolamines , Plant Oils/chemistry
6.
EBioMedicine ; 79: 104020, 2022 May.
Article in English | MEDLINE | ID: mdl-35490555

ABSTRACT

BACKGROUND: The adipocyte-hypertrophy associated remodeling of fat cell function is considered causal for the development of metabolic disorders. A better understanding of transcriptome and fatty acid (FA) related alterations with adipocyte hypertrophy combined with less-invasive strategies for the detection of the latter can help to increase the prognostic and diagnostic value of adipocyte size and FA composition as markers for metabolic disease. METHODS: To clarify adipocyte-hypertrophy associated transcriptomic alterations, fat cell size was related to RNA-Seq data from white adipose tissue and size-separated adipocytes. The relationship between adipocyte size and adipose tissue FA composition as measured by GC-MS was investigated. MR spectroscopy (MRS) methods for clinical scanning were developed to characterize adipocyte size and FA composition in a fast and non-invasive manner. FINDINGS: With enlarged adipocyte size, substantial transcriptomic alterations of genes involved in mitochondrial function and FA metabolism were observed. Investigations of these two mechanisms revealed a reciprocal relationship between adipocyte size and estimated thermogenic adipocyte content as well as depot-specific correlations of adipocyte size and FA composition. MRS on a clinical scanner was suitable for the in-parallel assessment of adipose morphology and FA composition. INTERPRETATION: The current study provides a comprehensive overview of the adipocyte-hypertrophy associated transcriptomic and FA landscape in both subcutaneous and visceral adipose tissue. MRS represents a promising technique to translate the observed mechanistic, structural and functional changes in WAT with adipocyte hypertrophy into a clinical context for an improved phenotyping of WAT in the context of metabolic diseases. FUNDING: Competence network for obesity (FKZ 42201GI1128), ERC (No 677661, ProFatMRI; No 875488, FatVirtualBiopsy), Else Kröner-Fresenius-Foundation.


Subject(s)
Fatty Acids , Transcriptome , Adipocytes/metabolism , Adipose Tissue/metabolism , Fatty Acids/metabolism , Humans , Hypertrophy/metabolism , Hypertrophy/pathology
7.
Mol Metab ; 61: 101499, 2022 07.
Article in English | MEDLINE | ID: mdl-35470094

ABSTRACT

OBJECTIVE: Classical ATP-independent non-shivering thermogenesis enabled by uncoupling protein 1 (UCP1) in brown adipose tissue (BAT) is activated, but not essential for survival, in the cold. It has long been suspected that futile ATP-consuming substrate cycles also contribute to thermogenesis and can partially compensate for the genetic ablation of UCP1 in mouse models. Futile ATP-dependent thermogenesis could thereby enable survival in the cold even when brown fat is less abundant or missing. METHODS: In this study, we explore different potential sources of UCP1-independent thermogenesis and identify a futile ATP-consuming triglyceride/fatty acid cycle as the main contributor to cellular heat production in brown adipocytes lacking UCP1. We uncover the mechanism on a molecular level and pinpoint the key enzymes involved using pharmacological and genetic interference. RESULTS: ATGL is the most important lipase in terms of releasing fatty acids from lipid droplets, while DGAT1 accounts for the majority of fatty acid re-esterification in UCP1-ablated brown adipocytes. Furthermore, we demonstrate that chronic cold exposure causes a pronounced remodeling of adipose tissues and leads to the recruitment of lipid cycling capacity specifically in BAT of UCP1-knockout mice, possibly fueled by fatty acids from white fat. Quantification of triglyceride/fatty acid cycling clearly shows that UCP1-ablated animals significantly increase turnover rates at room temperature and below. CONCLUSION: Our results suggest an important role for futile lipid cycling in adaptive thermogenesis and total energy expenditure.


Subject(s)
Adipose Tissue, Brown , Thermogenesis , Adenosine Triphosphate/metabolism , Adipose Tissue, Brown/metabolism , Animals , Fatty Acids/metabolism , Mice , Mice, Knockout , Triglycerides/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
8.
Gastroenterology ; 162(2): 658-659, 2022 02.
Article in English | MEDLINE | ID: mdl-34756893
9.
Gastroenterology ; 161(3): 910-923.e19, 2021 09.
Article in English | MEDLINE | ID: mdl-34000281

ABSTRACT

OBJECTIVE: Lipidomic changes were causally linked to metabolic diseases, but the scenario for colorectal cancer (CRC) is less clear. We investigated the CRC lipidome for putative tumor-specific alterations through analysis of 3 independent retrospective patient cohorts from 2 clinical centers, to derive a clinically useful signature. DESIGN: Quantitative comprehensive lipidomic analysis was performed using direct infusion electrospray ionization coupled with tandem mass spectrometry (ESI-MS/MS) and high-resolution mass spectrometry (HR-MS) on matched nondiseased mucosa and tumor tissue in a discovery cohort (n = 106). Results were validated in 2 independent cohorts (n = 28, and n = 20), associated with genomic and clinical data, and lipidomic data from a genetic mouse tumor model (Apc1638N). RESULTS: Significant differences were found between tumor and normal tissue for glycero-, glycerophospho-, and sphingolipids in the discovery cohort. Comparison to the validation collectives unveiled that glycerophospholipids showed high interpatient variation and were strongly affected by preanalytical conditions, whereas glycero- and sphingolipids appeared more robust. Signatures of sphingomyelin and triacylglycerol (TG) species significantly differentiated cancerous from nondiseased tissue in both validation studies. Moreover, lipogenic enzymes were significantly up-regulated in CRC, and FASN gene expression was prognostically detrimental. The TG profile was significantly associated with postoperative disease-free survival and lymphovascular invasion, and was essentially conserved in murine digestive cancer, but not associated with microsatellite status, KRAS or BRAF mutations, or T-cell infiltration. CONCLUSION: Analysis of the CRC lipidome revealed a robust TG-species signature with prognostic potential. A better understanding of the cancer-associated glycerolipid and sphingolipid metabolism may lead to novel therapeutic strategies.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Lipidomics , Lipids/analysis , Metabolome , Adult , Aged , Aged, 80 and over , Animals , Ceramides/analysis , Colectomy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Disease-Free Survival , Female , Genes, APC , Germany , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neoplasm Invasiveness , Reproducibility of Results , Retrospective Studies , Spectrometry, Mass, Electrospray Ionization , Sphingolipids/analysis , Tandem Mass Spectrometry , Triglycerides/analysis
10.
Int J Med Microbiol ; 311(3): 151488, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33662870

ABSTRACT

Gut microbiota significantly influence the plasma and liver lipidome. An interconnecting metabolite is acetate generated after degradation and fermentation of dietary fiber by the gut microbiota, which is metabolized in the liver into longer chain fatty acids and complex lipids reaching the circulation. Whether these systemic changes are accompanied by alternations of the intestinal lipidome is unclear. Therefore, we quantified glycerophospholipids, sphingolipids and sterols in ileum and colon, the two segments containing the highest densities of microbes in the gastrointestinal tract, of germfree and specific pathogen free mice using mass spectrometry-based lipidomics. We found that the presence of gut microbes lowers the free cholesterol content in colon while elevating phosphatidylcholine levels. Further, PUFA-containing phosphatidylcholine and -ethanolamine fractions are increased in ileum and colon of germfree compared to SPF mice. A total fatty acid analysis by GC-MS revealed higher levels of arachidonic and docosahexaenoic acid in the ileum of germfree mice indicating that the gut microbiota inhibits PUFA metabolism in the small intestine.


Subject(s)
Gastrointestinal Microbiome , Animals , Colon , Fatty Acids , Gastrointestinal Tract , Lipidomics , Mice
11.
Cell Rep ; 34(5): 108710, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33535053

ABSTRACT

Diurnal regulation of whole-body lipid metabolism plays a vital role in metabolic health. Although changes in lipid levels across the diurnal cycle have been investigated, the system-wide molecular responses to both short-acting fasting-feeding transitions and longer-timescale circadian rhythms have not been explored in parallel. Here, we perform time-series multi-omics analyses of liver and plasma revealing that the majority of molecular oscillations are entrained by adaptations to fasting, food intake, and the postprandial state. By developing algorithms for lipid structure enrichment analysis and lipid molecular crosstalk between tissues, we find that the hepatic phosphatidylethanolamine (PE) methylation pathway is diurnally regulated, giving rise to two pools of oscillating phosphatidylcholine (PC) molecules in the circulation, which are coupled to secretion of either very low-density lipoprotein (VLDL) or high-density lipoprotein (HDL) particles. Our work demonstrates that lipid molecular timeline profiling across tissues is key to disentangling complex metabolic processes and provides a critical resource for the study of whole-body lipid metabolism.


Subject(s)
Lipid Metabolism/genetics , Liver/physiology , Animals , Circadian Rhythm , Mice
12.
Gut Microbes ; 13(1): 1-21, 2021.
Article in English | MEDLINE | ID: mdl-33382950

ABSTRACT

Extibacter muris is a newly described mouse gut bacterium which metabolizes cholic acid (CA) to deoxycholic acid (DCA) via 7α-dehydroxylation. Although bile acids influence metabolic and inflammatory responses, few in vivo models exist for studying their metabolism and impact on the host. Mice were colonized from birth with the simplified community Oligo-MM12 with or without E. muris. As the metabolism of bile acids is known to affect lipid homeostasis, mice were fed either a low- or high-fat diet for eight weeks before sampling and analyses targeting the gut and liver. Multiple Oligo-MM12 strains were capable of deconjugating primary bile acids in vitro. E. muris produced DCA from CA either as pure compound or in mouse bile. This production was inducible by CA in vitro. Ursodeoxycholic, chenodeoxycholic, and ß-muricholic acid were not metabolized under the conditions tested. All gnotobiotic mice were stably colonized with E. muris, which showed higher relative abundances after HF diet feeding. The presence of E. muris had minor, diet-dependent effects on Oligo-MM12 communities. The secondary bile acids DCA and surprisingly LCA and their taurine conjugates were detected exclusively in E. muris-colonized mice. E. muris colonization did not influence body weight, white adipose tissue mass, liver histopathology, hepatic aspartate aminotransferase, or blood levels of cholesterol, insulin, and paralytic peptide (PP). However, proteomics revealed shifts in hepatic pathways involved in amino acid, glucose, lipid, energy, and drug metabolism in E. muris-colonized mice. Liver fatty acid composition was substantially altered by dietary fat but not by E. muris.In summary, E. muris stably colonized the gut of mice harboring a simplified community and produced secondary bile acids, which affected proteomes in the liver. This new gnotobiotic mouse model can now be used to study the pathophysiological role of secondary bile acids in vivo.


Subject(s)
Bile Acids and Salts/metabolism , Clostridiales/metabolism , Gastrointestinal Microbiome/physiology , Liver/physiology , Animals , Biotransformation , Clostridiales/growth & development , Diet, High-Fat , Germ-Free Life , Intestines/microbiology , Liver/metabolism , Mice
13.
Mol Cancer Res ; 18(12): 1849-1862, 2020 12.
Article in English | MEDLINE | ID: mdl-32859692

ABSTRACT

Recent studies indicate that adipose tissue in obesity promotes breast cancer progression by secreting protumorigenic chemokines, growth factors, and fatty acids. However, the detailed mechanisms by which hypertrophic adipose tissue influences breast cancer cells are still not well understood. Here we show that co-culture with adipose tissue from high-fat diet induced obese C57BL/6 mice alters transcriptome profiles in triple-negative breast cancer (TNBC) cells, leading to upregulation of genes involved in inflammation and lipid metabolism, such as IL1B, PLIN2, and ANGPTL4. Similar results were obtained by treating TNBC cells with adipose tissue conditioned media (ACM) generated from fat tissue of obese female patients. Many of the upregulated genes were activated by PPAR nuclear receptors, as shown by pathway analyses and gene expression experiments using PPAR agonists and antagonists. Metabolic analysis revealed that TNBC cells cultivated with ACM had significantly higher levels of ß-oxidation. Furthermore, ACM-treated TNBC cells displayed a pronounced aggressive cell phenotype, with enhanced wound healing, proliferation, and invasion capabilities. ACM-induced invasion was dependent on the PPAR-target ANGPTL4 and activated FAK signaling, as shown by ANGPTL4 depletion and FAK inhibition. Together, our data suggest that factors released by adipose tissue change PPAR-regulated gene expression and lipid metabolism and induce a more aggressive TNBC cell phenotype. These effects are, at least in parts, mediated by fatty acids provided by the adipose tissue. IMPLICATIONS: Adipose tissue provides factors for increased progression of TNBC cells, identifying PPAR- and FAK-signaling as potential novel targets for treatment of TNBC, especially in obese women.


Subject(s)
Adipose Tissue/cytology , Angiopoietin-Like Protein 4/metabolism , Breast Neoplasms/metabolism , Focal Adhesion Kinase 1/metabolism , Obesity/metabolism , PPAR alpha/metabolism , Adipose Tissue/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Coculture Techniques , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Mice , Obesity/chemically induced , Obesity/complications , Obesity/genetics , PPAR alpha/genetics
14.
PLoS Biol ; 17(8): e3000412, 2019 08.
Article in English | MEDLINE | ID: mdl-31369546

ABSTRACT

Lipid species patterns are conserved within cells to maintain physicochemical properties of membranes and cellular functions. We present the lipidome, including sterols, glycerolipids (GLs), glycerophospholipids (GPLs), and sphingolipids (SLs), of primary ex vivo differentiated (I) white, (II) brite, and (III) brown adipocytes derived from primary preadipocytes isolated from (I) epididymal white, (II) inguinal white, and (III) intrascapular brown adipose tissue. Quantitative lipidomics revealed significantly decreased fractions of phosphatidylcholine (PC) and phosphatidylethanolamine (PE), with longer (C > 36) and more polyunsaturated species, as well as lower levels of cardiolipin (CL) in white than in brite and brown adipocytes. Together, the brite and brown lipidome was comparable and indicates differences in membrane lipid packing density compared with white adipocytes. Changes in ceramide species profile could be related to the degree of browning. Beta-adrenergic stimulation of brown adipocytes led to generation of saturated lyso-PC (LPC) increasing uncoupling protein (UCP) 1-mediated leak respiration. Application of stable isotope labeling showed that LPC formation was balanced by an increased de novo synthesis of PC.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adrenergic Agents , Animals , Cell Differentiation , Lipid Metabolism/physiology , Lipidomics/methods , Lipids/physiology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
15.
Biomolecules ; 9(4)2019 03 28.
Article in English | MEDLINE | ID: mdl-30925749

ABSTRACT

Short chain fatty acids (SCFAs) are generated by the degradation and fermentation of complex carbohydrates, (i.e., dietary fiber) by the gut microbiota relevant for microbe⁻host communication. Here, we present a method for the quantification of SCFAs in fecal samples by liquid chromatography tandem mass spectrometry (LC-MS/MS) upon derivatization to 3-nitrophenylhydrazones (3NPH). The method includes acetate, propionate, butyrate, and isobutyrate with a run time of 4 min. The reproducible (coefficients of variation (CV) below 10%) quantification of SCFAs in human fecal samples was achieved by the application of stable isotope labelled internal standards. The specificity was demonstrated by the introduction of a quantifier and qualifier ions. The method was applied to investigate the pre-analytic stability of SCFAs in human feces. Concentrations of SCFA may change substantially within hours; the degree and kinetics of these changes revealed huge differences between the donors. The fecal SCFA level could be preserved by the addition of organic solvents like isopropanol. An analysis of the colon content of mice either treated with antibiotics or fed with a diet containing a non-degradable and -fermentable fiber source showed decreased SCFA concentrations. In summary, this fast and reproducible method for the quantification of SCFA in fecal samples provides a valuable tool for both basic research and large-scale studies.


Subject(s)
Fatty Acids, Volatile/analysis , Animals , Chromatography, Liquid , Humans , Mice , Mice, Inbred C57BL , Quality Control , Tandem Mass Spectrometry
16.
Front Chem ; 6: 483, 2018.
Article in English | MEDLINE | ID: mdl-30420949

ABSTRACT

Species of the genus Wolffia are traditionally used as human food in some of the Asian countries. Therefore, all 11 species of this genus, identified by molecular barcoding, were investigated for ingredients relevant to human nutrition. The total protein content varied between 20 and 30% of the freeze-dry weight, the starch content between 10 and 20%, the fat content between 1 and 5%, and the fiber content was ~25%. The essential amino acid content was higher or close to the requirements of preschool-aged children according to standards of the World Health Organization. The fat content was low, but the fraction of polyunsaturated fatty acids was above 60% of total fat and the content of n-3 polyunsaturated fatty acids was higher than that of n-6 polyunsaturated fatty acids in most species. The content of macro- and microelements (minerals) not only depended on the cultivation conditions but also on the genetic background of the species. This holds true also for the content of tocopherols, several carotenoids and phytosterols in different species and even intraspecific, clonal differences were detected in Wolffia globosa and Wolffia arrhiza. Thus, the selection of suitable clones for further applications is important. Due to the very fast growth and the highest yield in most of the nutrients, Wolffia microscopica has a high potential for practical applications in human nutrition.

17.
Nat Commun ; 9(1): 3760, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218046

ABSTRACT

Interactions between the gut microbial ecosystem and host lipid homeostasis are highly relevant to host physiology and metabolic diseases. We present a comprehensive multi-omics view of the effect of intestinal microbial colonization on hepatic lipid metabolism, integrating transcriptomic, proteomic, phosphoproteomic, and lipidomic analyses of liver and plasma samples from germfree and specific pathogen-free mice. Microbes induce monounsaturated fatty acid generation by stearoyl-CoA desaturase 1 and polyunsaturated fatty acid elongation by fatty acid elongase 5, leading to significant alterations in glycerophospholipid acyl-chain profiles. A composite classification score calculated from the observed alterations in fatty acid profiles in germfree mice clearly differentiates antibiotic-treated mice from untreated controls with high sensitivity. Mechanistic investigations reveal that acetate originating from gut microbial degradation of dietary fiber serves as precursor for hepatic synthesis of C16 and C18 fatty acids and their related glycerophospholipid species that are also released into the circulation.


Subject(s)
Acetates/metabolism , Acetyltransferases/metabolism , Dietary Fiber/metabolism , Fatty Acids/metabolism , Gastrointestinal Microbiome/physiology , Liver/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Fatty Acid Elongases , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression Profiling , Germ-Free Life , Lipid Metabolism , Mice , Proteomics
18.
Microbiome ; 6(1): 134, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30071904

ABSTRACT

BACKGROUND: As the gut microbiota contributes to metabolic health, it is important to determine specific diet-microbiota interactions that influence host metabolism. Bile acids and dietary fat source can alter phenotypes of diet-induced obesity, but the interplay with intestinal microorganisms is unclear. Here, we investigated metabolic consequences of diets enriched in primary bile acids with or without addition of lard or palm oil, and studied gut microbiota structure and functions in mice. RESULTS: In combination with bile acids, dietary lard fed to male C57BL/6N mice for a period of 8 weeks enhanced fat mass accumulation in colonized, but not in germ-free mice when compared to palm oil. This was associated with impaired glucose tolerance, lower fasting insulin levels, lower counts of enteroendocrine cells, fatty liver, and elevated amounts of hepatic triglycerides, cholesteryl esters, and monounsaturated fatty acids. Lard- and bile acid-fed mice were characterized by shifts in dominant gut bacterial communities, including decreased relative abundances of Lachnospiraceae and increased occurrence of Desulfovibrionaceae and the species Clostridium lactatifermentans and Flintibacter butyricus. Metatranscriptomic analysis revealed shifts in microbial functions, including lipid and amino acid metabolism. CONCLUSIONS: Caution is required when interpreting data from diet-induced obesity models due to varying effects of dietary fat source. Detrimental metabolic consequences of a diet enriched with lard and primary bile acids were dependent on microbial colonization of the host and were linked to hepatic lipid rearrangements and to alterations of dominant bacterial communities in the cecum.


Subject(s)
Bacteria/classification , Bile Acids and Salts/analysis , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Obesity/chemically induced , Amino Acids/metabolism , Animals , Bacteria/drug effects , Bacteria/genetics , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Dietary Fats/adverse effects , Gene Expression Profiling , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Palm Oil/adverse effects , Phylogeny , RNA, Ribosomal, 16S/genetics
19.
J Lipid Res ; 56(10): 2029-39, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26239049

ABSTRACT

Acylcarnitines are intermediates of fatty acid and amino acid oxidation found in tissues and body fluids. They are important diagnostic markers for inherited diseases of peroxisomal and mitochondrial oxidation processes and were recently described as biomarkers of complex diseases like the metabolic syndrome. Quantification of acylcarnitine species can become challenging because various species occur as isomers and/or have very low concentrations. Here we describe a new LC-MS/MS method for quantification of 56 acylcarnitine species with acyl-chain lengths from C2 to C18. Our method includes amino acid-derived positional isomers, like methacrylyl-carnitine (2-M-C3:1-CN) and crotonyl-carnitine (C4:1-CN), and odd-numbered carbon species, like pentadecanoyl-carnitine (C15:0-CN) and heptadecanoyl-carnitine (C17:0-CN), occurring at very low concentrations in plasma and tissues. Method validation in plasma and liver samples showed high sensitivity and excellent accuracy and precision. In an application to samples from streptozotocin-treated diabetic mice, we identified significantly increased concentrations of acylcarnitines derived from branched-chain amino acid degradation and of odd-numbered straight-chain species, recently proposed as potential biomarkers for the metabolic syndrome. In conclusion, the LC-MS/MS method presented here allows robust quantification of isomeric acylcarnitine species and extends the palette of acylcarnitines with diagnostic potential derived from fatty acid and amino acid metabolism.


Subject(s)
Carnitine/analogs & derivatives , Chromatography, Liquid/methods , Fatty Acids/analysis , Tandem Mass Spectrometry/methods , Amino Acids/metabolism , Animals , Carnitine/analysis , Carnitine/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Fatty Acids/blood , Fatty Acids/metabolism , Isomerism , Liver/chemistry , Liver/metabolism , Mice , Mice, Inbred C57BL , Oxidation-Reduction
20.
Diabetologia ; 58(9): 2133-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26058503

ABSTRACT

AIMS/HYPOTHESIS: Metabolomics approaches in humans have identified around 40 plasma metabolites associated with insulin resistance (IR) and type 2 diabetes, which often coincide with those for obesity. We aimed to separate diabetes-associated from obesity-associated metabolite alterations in plasma and study the impact of metabolically important tissues on plasma metabolite concentrations. METHODS: Two obese mouse models were studied; one exclusively with obesity (ob/ob) and another with type 2 diabetes (db/db). Both models have impaired leptin signalling as a cause for obesity, but the different genetic backgrounds determine the susceptibility to diabetes. In these mice, we profiled plasma, liver, skeletal muscle and adipose tissue via semi-quantitative GC-MS and quantitative liquid chromatography (LC)-MS/MS for a wide range of metabolites. RESULTS: Metabolite profiling identified 24 metabolites specifically associated with diabetes but not with obesity. Among these are known markers such as 1,5-anhydro-D-sorbitol, 3-hydroxybutyrate and the recently reported marker glyoxylate. New metabolites in the diabetic model were lysine, O-phosphotyrosine and branched-chain fatty acids. We also identified 33 metabolites that were similarly altered in both models, represented by branched-chain amino acids (BCAA) as well as glycine, serine, trans-4-hydroxyproline, and various lipid species and derivatives. Correlation analyses showed stronger associations for plasma amino acids with adipose tissue metabolites in db/db mice compared with ob/ob mice, suggesting a prominent contribution of adipose tissue to changes in plasma in a diabetic state. CONCLUSIONS/INTERPRETATION: By studying mice with metabolite signatures that resemble obesity and diabetes in humans, we have found new metabolite entities for validation in appropriate human cohorts and revealed their possible tissue of origin.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Metabolome , Obesity/genetics , 3-Hydroxybutyric Acid/metabolism , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/blood , Fatty Acids/metabolism , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation , Glyoxylates/metabolism , Insulin Resistance , Leptin/metabolism , Liver/metabolism , Lysine/metabolism , Male , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Phosphotyrosine/metabolism , Signal Transduction , Sorbitol/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...