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

Publication year range
1.
Metab Eng ; 81: 197-209, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072356

ABSTRACT

Ricinoleic acid (C18:1-OH, RA) is a valuable hydroxy fatty acid with versatile applications. The current industrial source of RA relies on the hydrolysis of castor bean oil. However, the coexistence of the toxic compound ricin and the unstable supply of this plant have led to an exploration of promising alternatives: generating RA in heterologous plants or microorganisms. In this study, we engineered the oleaginous yeast Yarrowia lipolytica to produce RA in the form of free fatty acids (FFA). First, we overexpressed fungal Δ12 oleate hydroxylase gene (CpFAH12) from Claviceps purpurea while deleting genes related to fatty acid degradation (MEF1 and PEX10) and oleic acid desaturation (FAD2). Since Δ12 oleate hydroxylase converts oleic acid (C18:1) located at the sn-2 position of phosphatidylcholine (PC), we next focused on increasing the PC pool containing oleic acid. This objective was achieved thorough implementing metabolic engineering strategies designed to enhance the biosynthesis of PC and C18 fatty acids. To increase the PC pool, we redirected the flux towards phospholipid biosynthesis by deleting phosphatidic acid phosphatase genes (PAH1 and APP1) and diacylglycerol acyltransferase gene (DGA1), involved in the production of diacylglycerol and triacylglycerol, respectively. Furthermore, the PC biosynthesis via the CDP-DAG pathway was enhanced through the overexpression of CDS1, PSD1, CHO2, and OPI3 genes. Subsequently, to increase the oleic acid content within PC, we overexpressed the heterologous fatty acid elongase gene (MaC16E) involved in the conversion of C16 to C18 fatty acids. As RA production titer escalated, the produced RA was mainly found in the FFA form, leading to cell growth inhibition. The growth inhibition was mitigated by inducing RA secretion via Triton X-100 treatment, a process that simultaneously amplified RA production by redirecting flux towards RA synthesis. The final engineered strain JHYL-R146 produced 2.061 g/L of free RA in a medium treated with 5% Triton X-100, constituting 74% of the total FFAs produced. Generating free RA offers the added benefit of bypassing the hydrolysis stage required when employing castor bean oil as an RA source. This achievement represents the highest level of RA synthesis from glucose reported thus far, underscoring the potential of Y. lipolytica as a host for sustainable RA production.


Subject(s)
Fatty Acids, Nonesterified , Yarrowia , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Oleic Acid/genetics , Oleic Acid/metabolism , Ricinoleic Acids/metabolism , Octoxynol/metabolism , Fatty Acids/metabolism , Mixed Function Oxygenases/genetics , Metabolic Engineering
2.
Pflugers Arch ; 472(4): 473-480, 2020 04.
Article in English | MEDLINE | ID: mdl-32248286

ABSTRACT

Diabetic osteoporosis is a severe and chronic complication of diabetes in the bone and joint system, and its pathogenesis is needed to be explored. In the present study, we examined the effect and underlying mechanism of miR-155 on osteogenic differentiation in human bone marrow-derived mesenchymal stem cells (hBMSCs) under high glucose and free fatty acids (HG-FFA) conditions. It was shown that miR-155 levels in hBMSCs increased corresponding to the time of exposure to HG-FFA treatment. MiR-155 expression was altered by transfecting miR-155 mimic or miR-155 inhibitor. HG-FFA exposure resulted in an obviously decrease in cell viability and alkaline phosphatase (ALP) activity, and downregulated the expressionof runt-related transcription factor 2 (Runx2) and osteocalcin (OCN) in hBMSCs. Transfection of miR-155 mimic further exacerbated HG-FFA-induced inhibitory effect on osteogenic differentiation, and miR-155 inhibitor neutralized this inhibitory effect. Luciferase assays confirmed that SIRT1 was a direct target of miR-155 and can be negatively modulated by miR-155. Furthermore, SIRT1 siRNA partially counteracted miR-155 inhibitor-induced upregulation of SIRT1in HG-FFA-treated hBMSCs. SIRT1 siRNA also reversed the promotional effect of the miR-155 inhibitor on ALP activity and expression of the Runx2 and OCN proteins under HG-FFA conditions. In conclusion, the results suggest that miR-155 suppression promoted osteogenic differentiation of hBMSCs under HG-FFA conditions by targeting SIRT1. Inhibition of MiR-155 may provide a new therapeutic method for the prevention and treatment of diabetic osteoporosis.


Subject(s)
Bone Marrow Cells/cytology , Fatty Acids, Nonesterified/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Sirtuin 1/genetics , Bone and Bones/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Down-Regulation , Fatty Acids, Nonesterified/metabolism , Humans , Osteogenesis/drug effects , Osteogenesis/physiology , Signal Transduction/physiology , Transcriptional Activation/genetics
3.
PLoS Genet ; 13(11): e1007059, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29121639

ABSTRACT

Fat represents a calorically potent food source that yields approximately twice the amount of energy as carbohydrates or proteins per unit of mass. The highly palatable taste of free fatty acids (FAs), one of the building blocks of fat, promotes food consumption, activates reward circuitry, and is thought to contribute to hedonic feeding underlying many metabolism-related disorders. Despite a role in the etiology of metabolic diseases, little is known about how dietary fats are detected by the gustatory system to promote feeding. Previously, we showed that a broad population of sugar-sensing taste neurons expressing Gustatory Receptor 64f (Gr64f) is required for reflexive feeding responses to both FAs and sugars. Here, we report a genetic silencing screen to identify specific populations of taste neurons that mediate fatty acid (FA) taste. We find neurons identified by expression of Ionotropic Receptor 56d (IR56d) are necessary and sufficient for reflexive feeding response to FAs. Functional imaging reveals that IR56d-expressing neurons are responsive to short- and medium-chain FAs. Silencing IR56d neurons selectively abolishes FA taste, and their activation is sufficient to drive feeding responses. Analysis of co-expression with Gr64f identifies two subpopulations of IR56d-expressing neurons. While physiological imaging reveals that both populations are responsive to FAs, IR56d/Gr64f neurons are activated by medium-chain FAs and are sufficient for reflexive feeding response to FAs. Moreover, flies can discriminate between sugar and FAs in an aversive taste memory assay, indicating that FA taste is a unique modality in Drosophila. Taken together, these findings localize FA taste within the Drosophila gustatory center and provide an opportunity to investigate discrimination between different categories of appetitive tastants.


Subject(s)
Drosophila Proteins/genetics , Fatty Acids, Nonesterified/genetics , Receptors, Cell Surface/genetics , Sensory Receptor Cells/metabolism , Taste Perception/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation , Receptors, Cell Surface/metabolism , Receptors, Ionotropic Glutamate/genetics , Receptors, Ionotropic Glutamate/metabolism , Sugars/metabolism , Taste/genetics , Taste/physiology
4.
J Recept Signal Transduct Res ; 38(1): 76-82, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29369009

ABSTRACT

INTRODUCTION/AIMS: In recent years, it has been shown that free fatty acids receptors (FFAR) of whose function in the cell surface plays a significant role in the regulation of cell function and nutrition as well are activated by various endogenous ligands, but mainly by fatty acids. Within FFAR of our interest are GPR 41, 43 and 120. The functions of these receptors are varied and dependent on the tissue where they are. The activation and signaling of these receptors, FFAR, are involved in many physiological processes, and currently the target of many drugs in metabolic disorders like obesity, diabetes and atherosclerosis. MATERIAL AND METHODS: Obesity was induced with hypercaloric diet (HD) in male Wistar rats for 20 weeks (n = 10). At the end, adipose tissue (abdominal and subcutaneous) was taken to perform assays for relative quantification mRNA expression by end-point RT-PCR and protein level expression by Western blot. RESULTS: These present data have shown for the first time that total mRNA isolation and protein expression from both adipose tissues (abdominal and subcutaneous) of rat in obesity condition yield significative statistical difference among the control versus obese groups, showing that the diet high in carbohydrates modifies the total presence of mRNA and protein level expression of the receptors GPR41, 43 and 120. CONCLUSIONS: Further comparative methods are in process to clarify whether or not the obesity changes the functional receptors in these two tissues for new pharmacological approaches.


Subject(s)
Obesity/drug therapy , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Adipose Tissue/metabolism , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Diet, High-Fat , Disease Models, Animal , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation/genetics , Humans , Insulin/genetics , Insulin/metabolism , Obesity/metabolism , Obesity/pathology , Rats , Receptors, G-Protein-Coupled/metabolism
5.
J Proteome Res ; 16(10): 3741-3752, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28849941

ABSTRACT

The nonmetabolizable lysophosphatidylcholine (LysoPC) analogue edelfosine is the prototype of a class of compounds being investigated for their potential as selective chemotherapeutic agents. Edelfosine targets membranes, disturbing cellular homeostasis. Is not clear at this point how membrane alterations are communicated between intracellular compartments leading to growth inhibition and eventual cell death. In the present study, a combined metabolomics/lipidomics approach for the unbiased identification of metabolic pathways altered in yeast treated with sublethal concentrations of the LysoPC analogue was employed. Mass spectrometry of polar metabolites, fatty acids, and lipidomic profiling was used to study the effects of edelfosine on yeast metabolism. Amino acid and sugar metabolism, the Krebs cycle, and fatty acid profiles were most disrupted, with polar metabolites and short-medium chain fatty acid changes preceding long and very long-chain fatty acid variations. Initial increases in metabolites such as trehalose, proline, and γ-amino butyric acid with a concomitant decrease in metabolites of the Krebs cycle, citrate and fumarate, are interpreted as a cellular attempt to offset oxidative stress in response to mitochondrial dysfunction induced by the treatment. Notably, alanine, inositol, and myristoleic acid showed a steady increase during the period analyzed (2, 4, and 6 h after treatment). Of importance was the finding that edelfosine induced significant alterations in neutral glycerolipid metabolism resulting in a significant increase in the signaling lipid diacylglycerol.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Lipid Metabolism/genetics , Metabolomics , Phospholipid Ethers/metabolism , Citric Acid Cycle/genetics , Dietary Fats/metabolism , Fatty Acids/chemistry , Fatty Acids/genetics , Fatty Acids/metabolism , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/genetics , Lysophosphatidylcholines/chemistry , Lysophosphatidylcholines/metabolism , Oxidative Stress/genetics , Phospholipid Ethers/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
6.
Biosci Biotechnol Biochem ; 80(9): 1829-35, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26998626

ABSTRACT

Free fatty acids are useful as source materials for the production of biodiesel fuel and various chemicals such as pharmaceuticals and dietary supplements. Previously, we attained a 9.2-fold increase in free fatty acid productivity by disrupting a predicted acyl-CoA synthetase gene (faaA, AO090011000642) in Aspergillus oryzae. In this study, we achieved further increase in the productivity by overexpressing a predicted transketolase gene of the pentose phosphate pathway in the faaA disruptant. The A. oryzae genome is predicted to have three transketolase genes and overexpression of AO090023000345, one of the three genes, resulted in phenotypic change and further increase (corresponding to an increased production of 0.38 mmol/g dry cell weight) in free fatty acids at 1.4-fold compared to the faaA disruptant. Additionally, the biomass of hyphae increased at 1.2-fold by the overexpression. As a result, free fatty acid production yield per liter of liquid culture increased at 1.7-fold by the overexpression.


Subject(s)
Aspergillus oryzae/genetics , Coenzyme A Ligases/genetics , Fatty Acids, Nonesterified/biosynthesis , Fungal Proteins/genetics , Aspergillus oryzae/enzymology , Aspergillus oryzae/growth & development , Coenzyme A Ligases/biosynthesis , Fatty Acids, Nonesterified/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Hyphae/enzymology , Hyphae/genetics , Hyphae/growth & development , Pentose Phosphate Pathway/genetics , Transketolase/genetics
7.
Toxicol Appl Pharmacol ; 284(2): 101-12, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25748669

ABSTRACT

Developmental bisphenol A (BPA) exposure increases adulthood hepatic steatosis with reduced mitochondrial function. To investigate the potential epigenetic mechanisms behind developmental BPA-induced hepatic steatosis, pregnant Sprague-Dawley rats were dosed with vehicle (oil) or BPA (100µg/kg/day) from gestational day 6 until postnatal day (PND) 21. After weaning, offspring were either challenged with a high-fat (HF; 45% fat) or remained on a control (C) diet until PND110. From PND60 to 90, both BPA and HF diet increased the fat/lean ratio in males only, and the combination of BPA and HF diet appeared to cause the highest ratio. On PND110, Oil-HF, BPA-C, and BPA-HF males had higher hepatic lipid accumulation than Oil-C, with microvesicular steatosis being marked in the BPA-HF group. Furthermore, on PND1, BPA increased and modified hepatic triglyceride (TG) and free fatty acid (FFA) compositions in males only. In PND1 males, BPA increased hepatic expression of FFA uptake gene Fat/Cd36, and decreased the expression of TG synthesis- and ß-oxidation-related genes (Dgat, Agpat6, Cebpα, Cebpß, Pck1, Acox1, Cpt1a, Cybb). BPA altered DNA methylation and histone marks (H3Ac, H4Ac, H3Me2K4, H3Me3K36), and decreased the binding of several transcription factors (Pol II, C/EBPß, SREBP1) within the male Cpt1a gene, the key ß-oxidation enzyme. In PND1 females, BPA only increased the expression of genes involved in FFA uptake and TG synthesis (Lpl, Fasn, and Dgat). These data suggest that developmental BPA exposure alters and reprograms hepatic ß-oxidation capacity in males, potentially through the epigenetic regulation of genes, and further alters the response to a HF diet.


Subject(s)
Benzhydryl Compounds/toxicity , Diet, High-Fat/adverse effects , Fatty Liver/genetics , Liver/drug effects , Liver/physiology , Phenols/toxicity , Prenatal Exposure Delayed Effects/genetics , Animals , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression/drug effects , Liver/metabolism , Male , Oxidation-Reduction/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism , Triglycerides/genetics , Triglycerides/metabolism
8.
J Sci Food Agric ; 95(14): 2892-900, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25452231

ABSTRACT

BACKGROUND: Recent technological advances to improve the quality of virgin olive oil (VOO) have been focused on olive breeding programs by selecting outstanding cultivars and target progenies. Fatty acid (FA) composition, with special emphasis on oleic acid (C18:1) and palmitic acid (C16:0), is one of the most critical quality factors to be evaluated in VOO. For this reason, the profile of FAs is frequently used as a decision tool in olive breeding programs. RESULTS: A method based on gas chromatography with flame ionization detection (GC-FID) was used to study the influence of genotype on the concentration of ten of the most important FAs in VOOs from target crosses Arbequina × Arbosana, Picual × Koroneiki and Sikitita × Arbosana and their corresponding genitors Arbequina, Arbosana, Koroneiki, Picual and Sikitita. For this purpose, a targeted approach was selected for determination of esterified FAs (EFAs) and non-esterified FAs (NEFAs) in a dual analysis by the same chromatographic method. A Pearson analysis revealed correlations between pairs of FAs, which allowed detecting metabolic connections through desaturation and elongation enzymes. An ANOVA test (with P < 0.01) led to identification of C16:0 EFA, C16:1 EFA and C18:1 EFA and also C16:1 NEFA and C18:0 NEFA as the FAs more influenced by cross breeding. Statistical analysis was carried out by unsupervised analysis using principal component analysis (PCA) and cluster analysis (CA) to look for variability sources. CONCLUSION: Crosses with a common genitor (Arbequina × Arbosana and Sikitita × Arbosana) were partially overlapped in the PCAs using the profile of FAs. The CA results revealed clear differences between Sikitita × Arbosana and Picual × Koroneiki crosses in the composition of the most significant FAs, while Arbequina × Arbosana was not properly discriminated from the other crosses.


Subject(s)
Fatty Acids/analysis , Genotype , Olea/chemistry , Olive Oil/chemistry , Plant Breeding , Breeding , Chromatography, Gas , Fatty Acids/genetics , Fatty Acids, Nonesterified/analysis , Fatty Acids, Nonesterified/genetics , Flame Ionization , Humans , Olea/genetics , Oleic Acid/analysis , Oleic Acid/genetics , Palmitic Acid/analysis , Species Specificity
9.
Metab Eng ; 25: 82-91, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25014174

ABSTRACT

Crude glycerol, generated as waste by-product in biodiesel production process, has been considered as an important carbon source for converting to value-added bioproducts recently. Free fatty acids (FFAs) can be used as precursors for the production of biofuels or biochemicals. Microbial biosynthesis of FFAs can be achieved by introducing an acyl-acyl carrier protein thioesterase into Escherichia coli. In this study, the effect of metabolic manipulation of FFAs synthesis cycle, host genetic background and cofactor engineering on FFAs production using glycerol as feed stocks was investigated. The highest concentration of FFAs produced by the engineered stain reached 4.82g/L with the yield of 29.55% (g FFAs/g glycerol), about 83% of the maximum theoretical pathway value by the type II fatty acid synthesis pathway. In addition, crude glycerol from biodiesel plant was also used as feedstock in this study. The FFA production was 3.53g/L with a yield of 24.13%. The yield dropped slightly when crude glycerol was used as a carbon source instead of pure glycerol, while it still can reach about 68% of the maximum theoretical pathway yield.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/physiology , Fatty Acids, Nonesterified/biosynthesis , Glycerol/metabolism , Metabolic Engineering/methods , NADP Transhydrogenases/metabolism , Phosphotransferases/metabolism , Computer Simulation , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fatty Acids, Nonesterified/genetics , Genetic Enhancement/methods , Models, Biological , NADP Transhydrogenases/genetics , Phosphotransferases/genetics
10.
Metab Eng ; 14(4): 380-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22480945

ABSTRACT

Microbial biosynthesis of fatty acid like chemicals from renewable carbon sources has attracted significant attention in recent years. Free fatty acids can be used as precursors for the production of fuels or chemicals. Wild type E. coli strains produce fatty acids mainly for the biosynthesis of lipids and cell membranes and do not accumulate free fatty acids as intermediates in lipid biosynthesis. However, free fatty acids can be produced by breaking the fatty acid elongation through the overexpression of an acyl-ACP thioesterase. Since acetyl-CoA might be an important factor for fatty acid synthesis (acetate formation pathways are the main competitive pathways in consuming acetyl-CoA or pyruvate, a precursor of acetyl-CoA), and the long chain fatty acid CoA-ligase (FadD) plays a pivotal role in the transport and activation of exogenous fatty acids prior to their subsequent degradation, we examined the composition and the secretion of the free fatty acids in four different strains including the wild type MG1655, a mutant strain with inactivation of the fatty acid beta-oxidation pathway (fadD mutant (ML103)), and mutant strains with inactivation of the two major acetate production pathways (an ack-pta (acetate kinase/phosphotransacetylase), poxB (pyruvate oxidase) double mutant (ML112)) and a fadD, ack-pta, poxB triple mutant (ML115). The engineered E. coli cells expressing acyl-ACP thioesterase with glucose yield is higher than 40% of theoretical yield. Compared to MG1655(pXZ18) and ML103(pXZ18), acetate forming pathway deletion strains such as ML112(pXZ18) and ML115(pXZ18) produced similar quantity of total free fatty acids, which indicated that acetyl-CoA availability does not appear to be limiting factor for fatty acid production in these strains. However, these strains did show significant differences in the composition of free fatty acids. Different from MG1655(pXZ18) and ML103(pXZ18), acetate formation pathway deletion strains such as ML112(pXZ18) and ML115(pXZ18) produced similar level of C14, C16:1 and C16 free fatty acids, and the free fatty acid compositions of both strains did not change significantly with time. In addition, the strains bearing the fadD mutation showed significant differences in the quantities of free fatty acids found in the broth. Finally, we examined two potential screening methods for selecting and isolating high free fatty acids producing cells.


Subject(s)
Acetates/metabolism , Coenzyme A Ligases/metabolism , Escherichia coli/metabolism , Fatty Acids, Nonesterified/biosynthesis , Palmitoyl-CoA Hydrolase/biosynthesis , Ricinus/enzymology , Acetate Kinase/genetics , Acetate Kinase/metabolism , Escherichia coli/genetics , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Mutation , Palmitoyl-CoA Hydrolase/genetics , Phosphate Acetyltransferase/genetics , Phosphate Acetyltransferase/metabolism , Pyruvate Oxidase/genetics , Pyruvate Oxidase/metabolism , Ricinus/genetics
11.
Biotechnol Bioeng ; 109(9): 2190-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22473793

ABSTRACT

The direct conversion of carbon dioxide into biofuels by photosynthetic microorganisms is a promising alternative energy solution. In this study, a model cyanobacterium, Synechococcus elongatus PCC 7942, is engineered to produce free fatty acids (FFA), potential biodiesel precursors, via gene knockout of the FFA-recycling acyl-ACP synthetase and expression of a thioesterase for release of the FFA. Similar to previous efforts, the engineered strains produce and excrete FFA, but the yields are too low for large-scale production. While other efforts have applied additional metabolic engineering strategies in an attempt to boost FFA production, we focus on characterizing the engineered strains to identify the physiological effects that limit cell growth and FFA synthesis. The strains engineered for FFA-production show reduced photosynthetic yields, chlorophyll-a degradation, and changes in the cellular localization of the light-harvesting pigments, phycocyanin and allophycocyanin. Possible causes of these physiological effects are also identified. The addition of exogenous linolenic acid, a polyunsaturated FFA, to cultures of S. elongatus 7942 yielded a physiological response similar to that observed in the FFA-producing strains with only one notable difference. In addition, the lipid constituents of the cell and thylakoid membranes in the FFA-producing strains show changes in both the relative amounts of lipid components and the degree of saturation of the fatty acid side chains. These changes in lipid composition may affect membrane integrity and structure, the binding and diffusion of phycobilisomes, and the activity of membrane-bound enzymes including those involved in photosynthesis. Thus, the toxicity of unsaturated FFA and changes in membrane composition may be responsible for the physiological effects observed in FFA-producing S. elongatus 7942. These issues must be addressed to enable the high yields of FFA synthesis necessary for large-scale biofuel production.


Subject(s)
Fatty Acids, Nonesterified/biosynthesis , Metabolic Engineering/methods , Synechococcus/genetics , Synechococcus/metabolism , Biofuels , Biomass , Cell Membrane/chemistry , Cell Membrane/metabolism , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Photosynthesis/drug effects , Phycocyanin/analysis , Phycocyanin/metabolism , Plant Oils/analysis , Plant Oils/metabolism , Synechococcus/chemistry , Synechococcus/drug effects
12.
Mol Cell Biol ; 42(1): e0032721, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34694913

ABSTRACT

The activity of AMP-activated protein kinase α (AMPKα) is reduced in type 2 diabetes, and type 2 diabetes is associated with muscular atrophy. To date, there is little known about the mechanism by which free fatty acid (FFA) participates in muscular impairment. The purpose of the present study was to explore whether FFA damages myogenesis through the AMPKα-histone deacetylase 4 (HDAC4)-microRNA 206 (miR-206) pathway. The results showed that 1 mM FFA produced lipid accumulation, significantly impaired the insulin signaling pathway, and decreased the myogenic differentiation of C2C12 myoblast cells. FFA reduced the LKB1-AMPKα pathway, and the activation of AMPKα rescued the myogenic impairment caused by FFA (P < 0.05). AMPKα promoted myogenesis by regulating the expression of miR-206 through HDAC4 (P < 0.05) and affected the cell cycle and cell proliferation to promote myogenesis by regulating miR-206 and miR-206's target cyclin D1 gene. In addition, AICAR (5-aminoimidazole-4-carboxamide 1-ß-d-ribofuranoside) and HDAC4 small interfering RNA (siRNA) promoted myogenic differentiation compared with the FFA group; however, this positive effect was significantly downregulated after transfection with the miR-206 inhibitor. In summary, AMPKα plays positive roles in myogenic differentiation and myogenesis, and FFA decreased myogenic differentiation and myotube formation through the AMPKα-HDAC4-miR-206 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Nonesterified/metabolism , MicroRNAs/genetics , Animals , Cell Differentiation/drug effects , Cell Proliferation/genetics , Diabetes Mellitus, Type 2/genetics , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/pharmacology , Mice , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , RNA, Small Interfering/metabolism
13.
Curr Mol Med ; 21(3): 257-264, 2021.
Article in English | MEDLINE | ID: mdl-32338219

ABSTRACT

BACKGROUND: Elevation of plasma free fatty acids as a principal aspect of type 2 diabetes maintains etiologically insulin insensitivity in target cells. TNF-α inhibitory effects on key insulin signaling pathway elements remain to be verified in insulinresistant hepatic cells. Thus, TNF-α knockdown effects on the key elements of insulin signaling were investigated in the palmitate-induced insulin-resistant hepatocytes. The Akt serine kinase, a key protein of the insulin signaling pathway, phosphorylation was monitored to understand the TNF-α effect on probable enhancing of insulin resistance. METHODS: Insulin-resistant HepG2 cells were produced using 0.5 mM palmitate treatment and shRNA-mediated TNF-α gene knockdown and its down-regulation confirmed using ELISA technique. Western blotting analysis was used to assess the Akt protein phosphorylation status. RESULTS: Palmitate-induced insulin resistance caused TNF-α protein overexpression 1.2-, 2.78, and 2.25- fold as compared to the control cells at post-treatment times of 8 h, 16 h, and 24 h, respectively. In the presence of palmitate, TNF-α expression showed around 30% reduction in TNF-α knockdown cells as compared to normal cells. In the TNF-α down-regulated cell, Akt phosphorylation was approximately 62% more than control cells after treatment with 100 nM insulin in conjugation with 0.5 mM palmitate. CONCLUSIONS: The obtained data demonstrated that TNF-α protein expression reduction improved insulin-stimulated Akt phosphorylation in the HepG2 cells and decreased lipidinduced insulin resistance of the diabetic hepatocytes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Proto-Oncogene Proteins c-akt/genetics , Tumor Necrosis Factor-alpha/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Hep G2 Cells , Humans , Insulin/genetics , Palmitates/metabolism , Phosphorylation/genetics , Signal Transduction/genetics
14.
J Biol Chem ; 284(52): 36312-36323, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19778901

ABSTRACT

Intracellular lipid accumulation in the heart is associated with cardiomyopathy, yet the precise role of triglyceride (TG) remains unclear. With exercise, wild type hearts develop physiologic hypertrophy. This was associated with greater TG stores and a marked induction of the TG-synthesizing enzyme diacylglycerol (DAG) acyltransferase 1 (DGAT1). Transgenic overexpression of DGAT1 in the heart using the cardiomyocyte- specific alpha-myosin heavy chain (MHC) promoter led to approximately a doubling of DGAT activity and TG content and reductions of approximately 35% in cardiac ceramide, 26% in DAG, and 20% in free fatty acid levels. Cardiac function assessed by echocardiography and cardiac catheterization was unaffected. These mice were then crossed with animals expressing long-chain acyl-CoA synthetase via the MHC promoter (MHC-ACS), which develop lipotoxic cardiomyopathy. MHC-DGAT1XMHC-ACS double transgenic male mice had improved heart function; fractional shortening increased by 74%, and diastolic function improved compared with MHC-ACS mice. The improvement of heart function correlated with a reduction in cardiac DAG and ceramide and reduced cardiomyocyte apoptosis but increased fatty acid oxidation. In addition, the survival of the mice was improved. Our study indicates that TG is not likely to be a toxic lipid species directly, but rather it is a feature of physiologic hypertrophy and may serve a cytoprotective role in lipid overload states. Moreover, induction of DGAT1 could be beneficial in the setting of excess heart accumulation of toxic lipids.


Subject(s)
Cardiomyopathies/enzymology , Diacylglycerol O-Acyltransferase/biosynthesis , Myocardium/enzymology , Triglycerides/metabolism , Animals , Cardiomyopathies/genetics , Ceramides/genetics , Ceramides/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diglycerides/genetics , Diglycerides/metabolism , Enzyme Induction , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Promoter Regions, Genetic/genetics , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
15.
Lipids Health Dis ; 9: 146, 2010 Dec 27.
Article in English | MEDLINE | ID: mdl-21187011

ABSTRACT

BACKGROUND: Abnormalities in lipid metabolism and transport are hallmarks in analbuminemic Nagase rats (NAR) and humans. Triglyceridemia is nearly 3- to 5-fold higher in female NAR than in control Sprague-Dawley rats (SDR). Also, NAR present with a severe plasma free fatty acid (FFA) deficit. There are conflicting results regarding the mechanisms underlying NAR hypertriglyceridemia. OBJECTIVE: We aimed at investigating whether liver lipogenesis and triglyceride secretion rates into the plasma contribute to the hypertriglyceridemia in NAR. We also studied whether heparin or albumin administration would release the hypothesized lipolysis inhibition in NAR. METHODS: The incorporation of tritiated water into lipids and the linear accumulation rate of plasma triglycerides after Triton WR1339 injection were the measures of liver lipogenesis and triglyceride secretion rates. RESULTS: Lipogenesis (596 ± 40 vs. 929 ± 124 µmol 3H2O/g/h) and triglyceride (4.25 ± 1.00 vs. 7.04 ± 1.68 mg/dL/min) secretion rates were slower (P ≤ 0.05) in fasted NAR than in control SDR. The injection of either heparin or albumin elicited an increase in NAR plasma FFA levels over time. FFA levels reached control levels 90 min after the albumin administration, increasing from 0.36 ± 0.05 to 1.34 ± 0.16 mEq/L (P ≤ 0.05). These results indicate that the lack of plasma albumin inhibits intravascular lipolysis and causes the FFA deficit observed in NAR. CONCLUSION: NAR hepatic triglyceride synthesis and output do not contribute to NAR hypertriglyceridemia. We propose that the lack of albumin diminishes intravascular lipolysis which reduces the plasma triglyceride removal rate and explain both NAR hypertriglyceridemia and FFA deficiency.


Subject(s)
Fatty Acids, Nonesterified , Hypertriglyceridemia , Serum Albumin , Triglycerides , Animals , Deficiency Diseases/blood , Deficiency Diseases/genetics , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Heparin/administration & dosage , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Hypertriglyceridemia/metabolism , Lipogenesis/drug effects , Lipogenesis/genetics , Lipolysis/drug effects , Lipolysis/genetics , Polyethylene Glycols/administration & dosage , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Serum Albumin/deficiency , Serum Albumin/genetics , Triglycerides/blood , Triglycerides/genetics , Triglycerides/metabolism
16.
J Clin Invest ; 130(4): 1931-1947, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917686

ABSTRACT

Visceral adipose tissue plays a critical role in numerous diseases. Although imaging studies often show adipose involvement in abdominal diseases, their outcomes may vary from being a mild self-limited illness to one with systemic inflammation and organ failure. We therefore compared the pattern of visceral adipose injury during acute pancreatitis and acute diverticulitis to determine its role in organ failure. Acute pancreatitis-associated adipose tissue had ongoing lipolysis in the absence of adipocyte triglyceride lipase (ATGL). Pancreatic lipase injected into mouse visceral adipose tissue hydrolyzed adipose triglyceride and generated excess nonesterified fatty acids (NEFAs), which caused organ failure in the absence of acute pancreatitis. Pancreatic triglyceride lipase (PNLIP) increased in adipose tissue during pancreatitis and entered adipocytes by multiple mechanisms, hydrolyzing adipose triglyceride and generating excess NEFAs. During pancreatitis, obese PNLIP-knockout mice, unlike obese adipocyte-specific ATGL knockouts, had lower visceral adipose tissue lipolysis, milder inflammation, less severe organ failure, and improved survival. PNLIP-knockout mice, unlike ATGL knockouts, were protected from adipocyte-induced pancreatic acinar injury without affecting NEFA signaling or acute pancreatitis induction. Therefore, during pancreatitis, unlike diverticulitis, PNLIP leaking into visceral adipose tissue can cause excessive visceral adipose tissue lipolysis independently of adipocyte-autonomous ATGL, and thereby worsen organ failure.


Subject(s)
Adipocytes/enzymology , Intra-Abdominal Fat/enzymology , Lipase/metabolism , Pancreatitis/enzymology , Signal Transduction , Acute Disease , Adipocytes/pathology , Animals , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Intra-Abdominal Fat/pathology , Lipase/genetics , Male , Mice , Mice, Knockout , Pancreatitis/genetics , Pancreatitis/pathology
17.
Genes (Basel) ; 11(2)2020 02 19.
Article in English | MEDLINE | ID: mdl-32093082

ABSTRACT

Ketosis is one of the most prevalent transition metabolic disorders in dairy cows, and has been intrinsically influenced by both genetic and nutritional factors. However, altered gene expression with respective to dairy cow ketosis has not been addressed yet, especially at the genome-wide level. In this study, we recruited nine Holsteins diagnosed with clinical ketosis and ten healthy controls, for which whole blood samples were collected at both prepartum and postpartum. Four groups of blood samples were defined: from cows with ketosis at prepartum (PCK, N = 9) and postpartum (CK, N = 9), respectively, and controls at prepartum (PHC, N = 10) and postpartum (HC, N = 10). RNA-Seq approach was used for investigating gene expression, by which a total of 27,233 genes were quantified with four billion high-quality reads. Subsequently, we revealed 75 and four differentially expressed genes (DEGs) between sick and control cows at postpartum and prepartum, respectively, which indicated that sick and control cows had similar gene expression patterns at prepartum. Meanwhile, there were 95 DEGs between postpartum and prepartum for sick cows, which showed depressed changes of gene expression during this transition period in comparison with healthy cows (428 DEGs). Functional analyses revealed the associated DEGs with ketosis were mainly involved in biological stress response, ion homeostasis, AA metabolism, energy signaling, and disease related pathways. Finally, we proposed that the expression level of STX1A would be potentially used as a new biomarker because it was the only gene that was highly expressed in sick cows at both prepartum and postpartum. These results could significantly help us to understand the underlying molecular mechanisms for incidence and progression of ketosis in dairy cows.


Subject(s)
Cattle Diseases/metabolism , Ketosis/genetics , Ketosis/metabolism , Animal Nutritional Physiological Phenomena , Animals , Cattle/genetics , Cattle Diseases/genetics , Diet , Energy Metabolism/genetics , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Genome-Wide Association Study , Lactation , Milk/metabolism , Parturition/metabolism , Peripartum Period/genetics , Peripartum Period/metabolism , Postpartum Period/genetics , Postpartum Period/metabolism , Pregnancy
18.
Biochim Biophys Acta Gen Subj ; 1864(1): 129422, 2020 01.
Article in English | MEDLINE | ID: mdl-31491457

ABSTRACT

BACKGROUND: Previous studies suggested that fibrillar human IAPP (hIAPP) is more likely to deposit in ß-cells, resulting in ß-cell injury. However, the changes in the conformation of hIAPP in lipid environment and the mechanism involved in ß-cell damage are unclear. METHODS: Synthetic hIAPP was incubated with five types of free fatty acids and phospholipids 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-l-serine (POPS), which constitute the cell membrane. Thioflavin-T fluorescence assay was conducted to analyze the degree of hIAPP fibrosis, and circular dichroism spectroscopy was performed to detect the ß-fold formation of hIAPP. Furthermore, INS-1 cells were infected with human IAPP delivered by a GV230-EGFP plasmid. The effects of endogenous hIAPP overexpression induced by sodium palmitate on the survival, endoplasmic reticulum (ER) stress, and apoptosis of INS-1 cells were evaluated. RESULTS: The five types of free fatty acids can accelerate the fibrosis of hIAPP. Sodium palmitate also maintained the stability of fibrillar hIAPP. POPS, not POPC, accelerated hIAPP fibrosis. Treatment of INS-1 cells with sodium palmitate increased the expression of hIAPP, activated ER stress and ER stress-dependent apoptosis signaling pathways, and increased the apoptotic rate. CONCLUSION: Free fatty acids and anionic phospholipid can promote ß-fold formation and fibrosis in hIAPP. High lipid induced the overexpression of hIAPP and aggravated ER stress and apoptosis in INS-1 cells, which caused ß-cell death in high lipid environment. GENERAL SIGNIFICANCE: Our study reveals free fatty acids and hIAPP synergistically implicated in endoplasmic reticulum stress and apoptosis of islet ß-cells.


Subject(s)
Apoptosis/genetics , Fibrosis/genetics , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/genetics , Amyloid/genetics , Amyloid/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum Stress/genetics , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Gene Expression Regulation/genetics , Humans , Insulin-Secreting Cells/pathology , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/ultrastructure , Lipid Metabolism/genetics , Lipids/genetics , Palmitic Acid/metabolism , Phosphatidylcholines/genetics , Phosphatidylcholines/metabolism , Phosphatidylserines/genetics , Phosphatidylserines/metabolism , Protein Conformation, beta-Strand , Protein Folding
19.
Biomed Res Int ; 2019: 2596914, 2019.
Article in English | MEDLINE | ID: mdl-31467878

ABSTRACT

According to our previous studies, bta-miR-124a was differentially expressed in breast tissue between high-fat and low-fat dairy cows. However, the function of bta-miR-124a in lipid metabolism of dairy cows and the identification of its target genes have not been reported. Therefore, this study will identify the target gene of bta-miR-124a and explore its role in the regulation of milk lipid metabolism. First, preliminary bioinformatics prediction of bta-miR-124a candidate target genes was performed, and quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze relative expression changes of bta-miR-124a and its candidate target genes and the expression level of the downstream gene of the target gene in the lipid metabolism signaling pathway in dairy mammary epithelial cell lines (Mac-T), using the dual luciferase reporter system for the identification of the targeting relationship between bta-miR-124a and the candidate target gene. Then, the effect of transfection of bta-miR-124a mimics and inhibitors on triglyceride (TG) and free fatty acid (FFA) levels was analyzed. The results indicate that bta-miR-124a directly interacts with the 3'-untranslated region of peroxisomal trans-2-enoyl-CoA reductase (PECR) to downregulate its expression in Mac-T cells. Further, bta-mir-124a regulates the expression of PECR and the downstream gene extension of very long chain fatty acid protein 2 (ELOVL2) through an unsaturated fatty acid biosynthesis signaling pathway. In conclusion, bta-miR-124a is involved in lipid metabolism by directly downregulating the PECR gene and affecting the expression of the downstream gene ELOVL2 and regulates the content of some key secretory elements such as TG and FFA. The function of bta-miR-124a has a certain effect on the synthesis and secretion of milk fat in the mammary epithelial cells of dairy cows.


Subject(s)
Lipid Metabolism/genetics , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , Milk/metabolism , Animals , Cattle , Cell Line , Fatty Acid Elongases/genetics , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Regulation/genetics , Humans , Mammary Glands, Animal/cytology , Oxidoreductases Acting on CH-CH Group Donors , Transfection , Triglycerides/genetics , Triglycerides/metabolism
20.
PLoS One ; 14(10): e0223024, 2019.
Article in English | MEDLINE | ID: mdl-31647849

ABSTRACT

AIM: To examine global gene expression response to profound metabolic and hormonal stress induced by acute sprint exercise. METHODS: Healthy men and women (n = 14) performed three all-out cycle sprints interspersed by 20 min recovery. Muscle biopsies were obtained before the first, and 2h and 20 min after last sprint. Microarray analysis was performed to analyse acute gene expression response and repeated blood samples were obtained. RESULTS: In skeletal muscle, a set of immediate early genes, FOS, NR4A3, MAFF, EGR1, JUNB were markedly upregulated after sprint exercise. Gene ontology analysis from 879 differentially expressed genes revealed predicted activation of various upstream regulators and downstream biofunctions. Gene signatures predicted an enhanced turnover of skeletal muscle mass after sprint exercise and some novel induced genes such as WNT9A, FZD7 and KLHL40 were presented. A substantial increase in circulating free fatty acids (FFA) was noted after sprint exercise, in parallel with upregulation of PGC-1A and the downstream gene PERM1 and gene signatures predicting enhanced lipid turnover. Increase in growth hormone and insulin in blood were related to changes in gene expressions and both hormones were predicted as upstream regulators. CONCLUSION: This is the first study reporting global gene expression in skeletal muscle in response to acute sprint exercise and several novel findings are presented. First, in line with that muscle hypertrophy is not a typical finding after a period of sprint training, both hypertrophy and atrophy factors were regulated. Second, systemic FFA and hormonal and exposure might be involved in the sprint exercise-induced changes in gene expression.


Subject(s)
Fatty Acids, Nonesterified/genetics , Muscle Proteins/genetics , Muscle, Skeletal/physiopathology , Transcriptome/genetics , Adult , Blood Glucose/metabolism , Exercise/physiology , Fatty Acids, Nonesterified/metabolism , Female , Human Growth Hormone/genetics , Human Growth Hormone/metabolism , Humans , Insulin/metabolism , Male , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Running/physiology
SELECTION OF CITATIONS
SEARCH DETAIL