Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
Methods Mol Biol ; 2754: 471-481, 2024.
Article in English | MEDLINE | ID: mdl-38512683

ABSTRACT

Neuroinflammation is the brain condition that occurs due to the hyper-activation of brain's immune cells and microglia, over the stimulation of extracellular aggregated proteins such as amyloid plaques and by extracellular Tau as well. The phenotypic changes of microglia from inflammatory to anti-inflammatory can be triggered by many factors, which also includes dietary fatty acids. The classes of omega-3 fatty acids are the majorly responsible in maintaining the anti-inflammatory phenotype of microglia. The enhanced phagocytic ability of microglia might induce the clearance of extracellular aggregated proteins, such as amyloid beta and Tau. In this study, we emphasized on the effect of α-linolenic acid (ALA) on the activation of microglia and internalization of the extracellular Tau seed in microglia.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/metabolism , alpha-Linolenic Acid/therapeutic use , Microglia/metabolism , Anti-Inflammatory Agents/pharmacology , tau Proteins/metabolism
2.
Adv Nutr ; 14(6): 1644-1655, 2023 11.
Article in English | MEDLINE | ID: mdl-37778442

ABSTRACT

Overweight and obesity are highly prevalent worldwide and are associated with cardiovascular disease (CVD) risk factors, including systematic inflammation, dyslipidemia, and hypertension. Alpha-linolenic acid (ALA) is a plant-based essential polyunsaturated fatty acid associated with reduced CVD risks. This systematic review and meta-analysis aimed to investigate the effects of supplementation with ALA compared with the placebo on CVD risk factors in people with obesity or overweight (International Prospective Register of Systematic Reviews Registration No. CRD42023429563). This review included studies with adults using oral supplementation or food or combined interventions containing vegetable sources of ALA. All studies were randomly assigned trials with parallel or crossover designs. The Cochrane Collaboration tool was used for assessing the risk of bias (Version 1). PubMed, Web of Science, Embase, and Cochrane library databases were searched from inception to April 2023. Nineteen eligible randomized controlled trials, including 1183 participants, were included in the meta-analysis. Compared with placebo, dietary ALA supplementation significantly reduced C-reactive protein concentration (standardized mean difference [SMD] = -0.38 mg/L; 95% confidence interval [CI]: -0.72, -0.04), tumor necrosis factor-α concentration (SMD = -0.45 pg/mL; 95% CI: -0.73, -0.17), triglyceride in serum (SMD = -4.41 mg/dL; 95% CI: -5.99, -2.82), and systolic blood pressure (SMD = -0.37 mm Hg; 95% CI: -0.66, -0.08); but led to a significant increase in low-density lipoprotein cholesterol concentrations (SMD = 1.32 mg/dL; 95% CI: 0.05, 2.59). ALA supplementation had no significant effect on interleukin-6, diastolic blood pressure, total cholesterol, or high-density lipoprotein cholesterol (all P ≥ 0.05). Subgroup analysis revealed that ALA supplementation at a dose of ≥3 g/d from flaxseed and flaxseed oil had a more prominent effect on improving CVD risk profiles, particularly where the intervention duration was ≥12 wk and where the baseline CVD profile was poor.


Subject(s)
Cardiovascular Diseases , Adult , Humans , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/drug therapy , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use , Overweight/complications , Overweight/drug therapy , Randomized Controlled Trials as Topic , Cholesterol, HDL , Obesity/complications , Obesity/drug therapy , Dietary Supplements
3.
Int J Mol Sci ; 24(18)2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37762621

ABSTRACT

Cardiovascular diseases (CVDs) represent the leading cause of global mortality with 1.7 million deaths a year. One of the alternative systems to drug therapy to minimize the risk of CVDs is represented by alpha-linolenic acid (ALA), an essential fatty acid of the omega-3 series, known for its cholesterol-lowering effect. The main purpose of this review is to analyze the effects of ALA and investigate the relevant omega-6/omega-3 ratio in order to maintain functionally beneficial effects. Concerning the lipid-lowering preventive effects, ALA may favorably affect the values of LDL-C and triglycerides in both adult and pediatric populations. Furthermore, ALA has shown protective effects against hypertension, contributing to balancing blood pressure through customary diet. According to the 2009 EFSA statement, dietary ALA may contribute to reducing the risk of CVDs, thanks to anti-hypertensive, anti-atherosclerotic and cardioprotective effects.


Subject(s)
Cardiovascular Diseases , Fatty Acids, Omega-3 , Hypertension , Adult , Child , Humans , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Antihypertensive Agents , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control
4.
Antiviral Res ; 216: 105666, 2023 08.
Article in English | MEDLINE | ID: mdl-37429528

ABSTRACT

Zika virus (ZIKV) has garnered global attention due to its association with severe congenital defects including microcephaly. However, there are no licensed vaccines or drugs against ZIKV infection. Pregnant women have the greatest need for treatment, making drug safety crucial. Alpha-linolenic acid (ALA), a polyunsaturated ω-3 fatty acid, has been used as a health-care product and dietary supplement due to its potential medicinal properties. Here, we demonstrated that ALA inhibits ZIKV infection in cells without loss of cell viability. Time-of-addition assay revealed that ALA interrupts the binding, adsorption, and entry stages of ZIKV replication cycle. The mechanism is probably that ALA disrupts membrane integrity of the virions to release ZIKV RNA, inhibiting viral infectivity. Further examination revealed that ALA inhibited DENV-2, HSV-1, influenza virus and SARS-CoV-2 infection dose-dependently. ALA is a promising broad-spectrum antiviral agent.


Subject(s)
COVID-19 , Dengue , Herpes Simplex , Orthomyxoviridae , Zika Virus Infection , Zika Virus , Female , Humans , Pregnancy , Zika Virus Infection/drug therapy , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use , Antiviral Agents/therapeutic use , SARS-CoV-2 , Dengue/drug therapy , Herpes Simplex/drug therapy , Virus Replication
5.
Viruses ; 15(7)2023 06 24.
Article in English | MEDLINE | ID: mdl-37515117

ABSTRACT

Emergent Coronaviridae viruses, such as SARS-CoV-1 in 2003, MERS-CoV in 2012, and SARS-CoV-2 (CoV-2) in 2019, have caused millions of deaths. These viruses have added to the existing respiratory infection burden along with respiratory syncytial virus (RSV) and influenza. There are limited therapies for respiratory viruses, with broad-spectrum treatment remaining an unmet need. Since gut fermentation of fiber produces short-chain fatty acids (SCFA) with antiviral potential, developing a fatty acid-based broad-spectrum antiviral was investigated. Molecular docking of fatty acids showed α-linolenic acid (ALA) is likely to interact with CoV-2-S, NL63-CoV-S, and RSV-F, and an ALA-containing liposome interacted with CoV-2 directly, degrading the particle. Furthermore, a combination of ALA and a SCFA-acetate synergistically inhibited CoV2-N expression and significantly reduced viral plaque formation and IL-6 and IL-1ß transcript expression in Calu-3 cells, while increasing the expression of IFN-ß. A similar effect was also observed in RSV-infected A549 cells. Moreover, mice infected with a murine-adapted SARS-CoV-2 (MA10) and treated with an ALA-liposome encapsulating acetate showed significant reductions in plaque-forming units present in lung tissue and in infection-associated lung inflammation and cytokines. Taken together, these results demonstrate that the ALA liposome-encapsulating acetate can be a promising broad antiviral therapy against respiratory infections.


Subject(s)
COVID-19 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Animals , Mice , SARS-CoV-2 , Liposomes , alpha-Linolenic Acid/therapeutic use , Molecular Docking Simulation , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Acetates
6.
J Nutr Biochem ; 119: 109408, 2023 09.
Article in English | MEDLINE | ID: mdl-37336331

ABSTRACT

Increasing evidence demonstrated that pyroptosis and subsequent inflammation played an important role in the pathological process of non-alcoholic steatohepatitis (NASH). Plant sterol ester of α-linolenic acid (PS-ALA) was beneficial for non-alcoholic fatty liver disease, but the underlying mechanisms are still not fully understood. This study aims to investigate whether PS-ALA can protect against proptosis via regulating SIRT1. Thirty male C57BL/6J mice were fed a normal diet, a high-fat and high-cholesterol diet (HFCD), or a HFCD supplemented with either 1.3%ALA, 2%PS, or 3.3% PS-ALA for 24 weeks. Hepatocytes were treated with oleic acid and cholesterol (OA/Cho) with or without PS-ALA. We found that PS-ALA ameliorated NASH in HFCD-fed mice. In addition, PS-ALA decreased the expression of NLRP3 and ASC and reduced the co-localization of NLRP3 and cleave-Caspase-1. Also, PS-ALA protected against pyroptosis as evidenced by decreased co-localization of GSDMD and propidium iodide (PI) positive cells. Mechanistically, we revealed that the inhibitory action of PS-ALA on the pyroptosis was mediated by SIRT1. This was demonstrated by the fact that silencing SIRT1 with small interfering RNA or inhibition of SIRT1 with its inhibitor abolished the inhibition effect of PS-ALA on the expression of NLRP3 and GSDMD cleavage. Collectively, the data from the present study reveals a novel mechanism that PS-ALA inhibits pyroptosis and it triggered inflammation via stimulating SIRT1, which provides new insights into the beneficial effect of PS-ALA on NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Phytosterols , Mice , Animals , Non-alcoholic Fatty Liver Disease/pathology , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Sirtuin 1/genetics , Sirtuin 1/metabolism , Mice, Inbred C57BL , Cholesterol/pharmacology , Phytosterols/pharmacology , Inflammation , Esters/pharmacology
7.
Endocr Regul ; 57(1): 18-24, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36753666

ABSTRACT

Objective. The intent of the present study was to test two hypotheses. The primary hypothesis was that there would be differences between blood serum individual free fatty acids (SIFFA) and serum individual total fatty acids (SITFA) in terms of their different relationships (correlations) to each of homeostatic model assessment-individual insulin resistance (HOMA-IR) and homeostatic model assessment-individual insulin resistance-percentage ß-cell function (HOMA-% ß) remaining in human type 2 diabetic patients with pre-flaxseed oil (FXO) and pre-safflower oil (SFO) administration. The secondary hypothesis was that FXO (rich in alpha-linolenic acid, ALA) supplementation would alter these correlations differently in the SIFFA and STIFFA pools in comparison with the placebo SFO (poor in ALA). Methods. Patients were recruited via a newspaper advertisement and two physicians. All patients came to visit 1 and three months later to visit 2. At visit 2, the subjects were randomly assigned (double-blind) to flaxseed or safflower oil (placebo) treatment for three months until visit 3. Results. There were pre-intervention differences in the SIFFA and STIFA pool's relationships with each of HOMA-IR and HOMA-% ß. These relatioships remained either unchanged or became significant after intervention (treatment or placebo). There was a negative correlation found between HOMA-IR and serum free ALA (SFALA) mol % after FXO. Serum total ALA (STALA) mol % had no significant correlations with HOMA-IR and HOMA- % ß before and after flaxseed oil administration. Conclusions. The SIFFA and SITFA pools have different relationships with HOMA-IR and HOMA-% ß for each of pre- and post-intervention. It is concluded that the data support both the primary and the secondary hypotheses indicating that they are correct.


Subject(s)
Insulin Resistance , Linseed Oil , Humans , Linseed Oil/pharmacology , Linseed Oil/therapeutic use , Fatty Acids , Safflower Oil , Serum , Dietary Supplements , alpha-Linolenic Acid/therapeutic use
8.
Front Cell Infect Microbiol ; 12: 865320, 2022.
Article in English | MEDLINE | ID: mdl-35656035

ABSTRACT

Helicobacter pylori (H. pylori) infection is associated with a variety of gastrointestinal diseases. Here, we focused on the activity of a novel nanomedicine-liposomal linolenic acid (LipoLLA) against H. pylori and its impact on human fecal bacteria in vitro. The minimum inhibitory concentrations (MICs) of LipoLLA against 30 H. pylori clinical strains were determined in combination with amoxicillin (AMX), metronidazole (MTZ), levofloxacin (LVFX) and clarithromycin (CAM). Bactericidal activity was measured by generating concentration-bactericidal curves at different times and pH values. Leakage of glucose (GLU) and aspartate aminotransferase (AST) was detected, combined with detection of changes in morphology by electron microscopy, to study the mechanism of action of LipoLLA against H. pylori. The effect of LipoLLA on human fecal bacteria was studied by high-throughput sequencing of fecal samples. We observed a synergistic or additive effect when LipoLLA was combined with AMX, MTZ, LVFX and CAM. The concentration-sterilization curves were pH and time dependent. After treatment with LipoLLA, GLU and AST levels were increased (P<0.05), and the morphology of H. pylori changed significantly. Moreover, LipoLLA activity led to no significant changes in the intestinal flora in terms of alpha diversity, species composition, beta diversity, etc. In conclusion, LipoLLA showed good anti-H. pylori effects. It destroyed the outer membrane barrier and caused leakage of the bacterial contents to achieve anti-H. pylori effects. And LipoLLA had little effect on human fecal bacteria in vitro.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Amoxicillin/pharmacology , Amoxicillin/therapeutic use , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Humans , Liposomes/chemistry , Metronidazole/pharmacology , Microbial Sensitivity Tests , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use
9.
J Nutr Biochem ; 107: 109072, 2022 09.
Article in English | MEDLINE | ID: mdl-35660097

ABSTRACT

Apoptosis is a feature of progressions steatosis to nonalcoholic steatohepatitis (NASH) and can be explained by endoplasmic reticulum stress (ERS). The present study aimed to investigate the protective effects of plant sterol ester of α-linolenic acid (PS-ALA) on ERS-triggered apoptosis in high fat diet-fed mice and oleic acid-induced hepatocytes, and further explore the underlying mechanisms. Our results showed that PS-ALA improved Non-alcoholic fatty liver disease (NAFLD) in both in vivo and in vitro models. Moreover, PS-ALA treatment can attenuate ERS and associated apoptosis via inhibiting IRE1α/TRAF2/JNK signal pathway. Furthermore, we found that the protective effect of PS-ALA on ERS-triggered apoptosis was mediated by activation of AMP-activated protein kinase (AMPK) as pretreatment with Compound C, an AMPK inhibitor, abolished the anti-apoptotic effect of PS-ALA. Taken together, our results illustrate that PS-ALA attenuating ERS-mediated apoptosis via activating AMPK, which provided new insights into the protective effect of PS-ALA in NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Phytosterols , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Endoribonucleases/pharmacology , Esters/metabolism , Esters/pharmacology , Liver/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Phytosterols/metabolism , Protein Serine-Threonine Kinases , alpha-Linolenic Acid/metabolism , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use
10.
Lipids Health Dis ; 21(1): 20, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35144649

ABSTRACT

BACKGROUND: Dietary fatty acids intake affects the composition of erythrocyte fatty acids, which is strongly correlated with glycolipid metabolism disorders. This study aimed at investigating the different effects of marine-derived and plant-derived omega-3 polyunsaturated fatty acid (n-3 PUFA) on the fatty acids of erythrocytes and glycolipid metabolism in patients with type 2 diabetes mellitus (T2DM). METHODS: The randomized double-blinded trial that was performed on 180 T2DM patients. The participants were randomly assigned to three groups for the six-month intervention. The participants were randomly assigned to three groups for the six-month intervention. The fish oil (FO) group was administered with FO at a dose of 3 g/day containing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the perilla oil (PO) group was administered with PO at a dose of 3 g/day containing α-linolenic (ALA), the linseed and fish oil (LFO) group was administered with mixed linseed and fish oil at a dose of 3 g/day containing EPA, DHA and ALA. Demographic information were collected and anthropometric indices, glucose and lipid metabolism indexes, erythrocyte fatty acid composition were measured. Statistical analyses were performed using two-way ANOVA. RESULTS: A total of 150 patients finished the trial, with 52 of them in the FO group, 50 in the PO group and 48 in the LFO group. There were significant effects of time × treatment interaction on fast blood glucose (FBG), insulin, HOMA-IR and C-peptide, TC and triglyceride (TG) levels (P < 0.001). Glucose and C-peptide in PO and LFO groups decreased significantly and serum TG in FO group significantly decreased (P < 0.001) after the intervention. Erythrocyte C22: 5 n-6, ALA, DPA, n-6/n-3 PUFA, AA/EPA levels in the PO group were significantly higher than FO and LFO groups, while EPA, total n-3 PUFA and Omega-3 index were significantly higher in the FO and LFO groups compared to PO group. CONCLUSION: Supplementation with perilla oil decreased FBG while fish oil supplementation decreased the TG level. Marine-based and plant-based n-3 PUFAs exhibit different effects on fatty acid compositions of erythrocytes and regulated glycolipid metabolism. TRIAL REGISTRATION: This trial was recorded under Chinese Clinical Trial Registry Center (NO: ChiCTR-IOR-16008435 ) on May 28 2016.


Subject(s)
Diabetes Mellitus, Type 2/blood , Erythrocytes/drug effects , Fatty Acids, Omega-3/therapeutic use , Fatty Acids/blood , Fish Oils/therapeutic use , alpha-Linolenic Acid/therapeutic use , Double-Blind Method , Dyslipidemias/drug therapy , Erythrocytes/chemistry , Female , Humans , Male , Middle Aged , Plant Oils/therapeutic use
11.
J Thromb Haemost ; 20(2): 375-386, 2022 02.
Article in English | MEDLINE | ID: mdl-34758193

ABSTRACT

BACKGROUND: Sickle cell disease (SCD) is a genetic hemoglobinopathy associated with high morbidity and mortality. The primary cause of hospitalization in SCD is vaso-occlusive crisis (VOC), mediated by alteration of red blood cells, platelets, immune cells and a pro-adhesive endothelium. OBJECTIVES: We investigated the potential therapeutic use of the plant-derived omega-3 alpha-linolenic acid (ALA) in SCD. METHODS: Berkeley mice were fed a low- or high-ALA diet for 4 weeks, followed by analysis of liver fibrosis, endothelial activation, platelet activation and formation of platelet-neutrophils aggregates. Aggregation of platelets over collagen under flow after high-ALA was compared to a blocking P-selectin Fab. RESULTS: Dietary high-ALA was able to reduce the number of sickle cells in blood smear, liver fibrosis, and the expression of adhesion molecules on the endothelium of aorta, lungs, liver and kidneys (VCAM-1, ICAM-1 and vWF). Specific parameters of platelet activation were blunted after high-ALA feeding, notably P-selectin exposure and the formation of neutrophil-platelet aggregates, along with a correspondingly reduced expression of PSGL-1 on neutrophils. By comparison, in vivo treatment of SCD mice with the anti-P-selectin Fab was able to similarly reduce the formation of neutrophil-platelet aggregates, but did not reduce GpIbα shedding nor the activation of the αIIb ß3 integrin in response to thrombin. Both ALA feeding and P-selectin blocking significantly reduced collagen-mediated cell adhesion under flow. CONCLUSIONS: Dietary ALA is able to reduce the pro-inflammatory and pro-thrombotic state occurring in the SCD mouse model and may represent a novel, inexpensive and readily available therapeutic strategy for SCD.


Subject(s)
Anemia, Sickle Cell , alpha-Linolenic Acid , Anemia, Sickle Cell/drug therapy , Animals , Cell Adhesion , Collagen , Diet , Mice , Platelet Activation , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use
12.
Nutr Neurosci ; 25(7): 1400-1412, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33345728

ABSTRACT

OBJECTIVES: The brains of individuals with Down syndrome (DS) present defects in neurogenesis and synaptogenesis during prenatal and early postnatal stages that are partially responsible for their cognitive disabilities. Because oleic and linolenic fatty acids enhance neurogenesis, synaptogenesis, and cognitive abilities in rodents and humans, in this study we evaluated the ability of these compounds to restore these altered phenotypes in the Ts65Dn (TS) mouse model of DS during early postnatal stages. METHODS: TS and euploid mice were treated with oleic or linolenic acid from PD3 to PD15, and the short- and long- term effects of these acids on neurogenesis and synaptogenesis were evaluated. The effects of these treatments on the cognitive abilities of TS mice during early adulthood were also evaluated. RESULTS: Administration of oleic or linolenic acid did not modify cell proliferation immediately after treatment discontinuation or several weeks later. However, oleic acid increased the total number of DAPI+ cells (+ 26%), the percentage of BrdU+ cells that acquired a neural phenotype (+ 9.1%), the number of pre- (+ 29%) and post-synaptic (+ 32%) terminals and the cognitive abilities of TS mice (+ 18.1%). In contrast, linolenic acid only produced a slight cognitive improvement in TS mice. (+12.1%). DISCUSSION: These results suggest that early postnatal administration of oleic acid could palliate the cognitive deficits of DS individuals.


Subject(s)
Down Syndrome , Animals , Cognition , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/therapy , Female , Hippocampus , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oleic Acid , Pregnancy , alpha-Linolenic Acid/therapeutic use
13.
BMC Cardiovasc Disord ; 21(1): 532, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34749668

ABSTRACT

BACKGROUND: The cardioprotective ability of n-3 polyunsaturated fatty acids (PUFAs) is controversial. Most studies suggest a specific role for PUFAs in cardioprotection from ischemic heart disease (IHD). However, few studies have used genetic biomarkers of n-3 PUFAs to examine their potential relationships with IHD. This study aimed to use Mendelian randomization to evaluate whether genetically-predicted n-3 PUFAs affect IHD and cardiometabolic risk factors (CRFs). METHODS: Genetic variants strongly (p < 5 × 10-8) and independently (r2 > 0.1) associated with n-3 PUFAs were derived from the CHARGE Consortium (including 8,866 subjects of European ancestry) and were used as instrumental variables (IVs) for evaluating the effect of n-3 PUFAs, including α-linolenic acid (ALA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA). Data on the associations between the IVs and IHD, myocardial infarction, and CRFs (including diabetes, lipids, blood pressure, body mass index, and waist-to-hip ratio (WHR)) were obtained from the UK Biobank SOFT CAD GWAS with the CARDIoGRAMplusC4D 1000 Genomes-based GWAS (113,937 IHD cases and 339,115 controls), the Myocardial Infarction Genetics and CARDIoGRAM Exome consortia (42,335 MI cases and 78,240 controls), the DIAbetes Genetics Replication And Meta-analysis consortium (26,676 diabetes mellitus cases and 132,532 controls), the Global Lipids Genetics Consortium (n = 196,475), the International Consortium for Blood Pressure (n = 69,395), and the meta-analysis of GWAS for body fat distribution in the UK Biobank and Genetic Investigation of Anthropometric Traits (n = 694,649). RESULTS: Genetically-predicted higher ALA was associated with lower risk of IHD, type 2 diabetes (T2D), and lower serum lipids. The effect size per 0.05-unit increase (about 1 standard deviation) in plasma ALA level) was - 1.173 (95% confidence interval - 2.214 to - 0.133) for IHD. DPA and EPA had no association with IHD but were associated with a higher risk of T2D, higher levels of lipids or WHR. DHA had no association with IHD or CRFs. CONCLUSIONS: Our study suggests a benefit of ALA for IHD and its main risk factors. DHA, DPA, and EPA had no association with IHD but were partly associated with increasing cardiometabolic risk factors.


Subject(s)
Cardiometabolic Risk Factors , Fatty Acids, Omega-3/therapeutic use , Myocardial Ischemia/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Lipids/blood , Mendelian Randomization Analysis , Meta-Analysis as Topic , Myocardial Ischemia/blood , Myocardial Ischemia/therapy , alpha-Linolenic Acid/therapeutic use
14.
Nutrients ; 13(9)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34579018

ABSTRACT

BACKGROUND: High-fat diet (HFD) consumption induced gut dysbiosis, inflammation, obese-insulin resistance. Perilla seed oil (PSO) is a rich source of omega-3 polyunsaturated fatty acids with health promotional effects. However, the effects of PSO on gut microbiota/inflammation and metabolic disturbance in HFD-induced obesity have not been investigated. Therefore, we aimed to compare the effects of different doses of PSO and metformin on gut microbiota/inflammation, and metabolic parameters in HFD-fed rats. METHODS: Thirty-six male Wistar rats were fed either a normal diet or an HFD for 24 weeks. At week 13, HFD-fed rats received either 50, 100, and 500 mg/kg/day of PSO or 300 mg/kg/day metformin for 12 weeks. After 24 weeks, the metabolic parameters, gut microbiota, gut barrier, inflammation, and oxidative stress were determined. RESULTS: HFD-fed rats showed gut dysbiosis, gut barrier disruption with inflammation, increased oxidative stress, metabolic endotoxemia, and insulin resistance. Treatment with PSO and metformin not only effectively attenuated gut dysbiosis, but also improved gut barrier integrity and decreased gut inflammation. PSO also decreased oxidative stress, metabolic endotoxemia, and insulin resistance in HFD-fed rats. Metformin had greater benefits than PSO. CONCLUSION: PSO and metformin had the beneficial effect on attenuating gut inflammation and metabolic disturbance in obese-insulin resistance.


Subject(s)
Dysbiosis/drug therapy , alpha-Linolenic Acid/therapeutic use , Animals , Blotting, Western , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Insulin Resistance , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/blood , Male , Metformin/therapeutic use , Oxidative Stress , Plant Oils/therapeutic use , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
15.
Neurotoxicology ; 85: 245-253, 2021 07.
Article in English | MEDLINE | ID: mdl-34111468

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects many older people around the world. Numerous studies are underway to evaluate the protective effects of natural products in AD. Alpha-linoleic acid (ALA) is an essential unsaturated fatty acid that exhibits neuroprotective outcomes in rat models of ischemic stroke and Parkinson's disease. This research aimed to investigate the effect of ALA on oxidative stress, neuroinflammation, neuronal death, and memory deficit induced by amyloid-beta (Aß) peptide. After intrahippocampal injection of Aß1-42, rats received ALA (150 µg/kg, subcutaneously) for 14 consecutive days. ALA decreased the levels of malondialdehyde and nitric oxide, enhanced glutathione content, and increased the activity of catalase in the hippocampus of the rat model of AD. It also reduced the expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, nuclear factor-kappa B, and N-methyl-d-aspartate receptor subunits NR2A and NR2B mRNAs in the hippocampus, prevented the neuronal loss in the CA1 region, and enhanced the expression of α7 nicotinic acetylcholine receptor. In addition, ALA allowed Aß1-42-injected rats to spend less time and distance to reach the hidden platform in the Morris water maze test and to swim longer in the target quadrant. We concluded that ALA reduces the biochemical, molecular, histological, and behavioral changes caused by Aß1-42 and it may be an effective option for treating AD.


Subject(s)
Hippocampus/drug effects , Memory Disorders/prevention & control , Oxidative Stress/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , alpha-Linolenic Acid/therapeutic use , alpha7 Nicotinic Acetylcholine Receptor , Amyloid beta-Peptides/toxicity , Animals , Gene Expression , Hippocampus/metabolism , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/chemically induced , Memory Disorders/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Peptide Fragments/toxicity , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , alpha-Linolenic Acid/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , alpha7 Nicotinic Acetylcholine Receptor/genetics
16.
Article in English | MEDLINE | ID: mdl-34051428

ABSTRACT

INTRODUCTION: Consumption of omega-3 polyunsaturated fatty acids (n-3 PUFAs) has been reported to provide health benefits, but it remains unknown whether the fatty acids themselves or their oxygenated metabolites, oxylipins, are responsible for the beneficial effects. PURPOSE: This paper describes the design and rationale of a randomized, double-blinded, cross-over study comparing the effects of α-linolenic acid (ALA)-rich flax oil and docosahexaenoic acid (DHA)-rich fish oil supplementation on circulating oxylipin profiles in females with obesity, in relation to obesity-induced inflammation. METHODS AND ANALYSIS: Pre-menopausal females (n = 24) aged 20-55 with a BMI ≥30, will consume capsules containing flaxseed oil (4 g ALA/day) or fish oil (4 g DHA + 0.8 g EPA/day) during 4-week supplementation phases, with a minimum 4-week washout. The primary outcome is alterations in plasma oxylipin profiles. Secondary outcomes include effects of supplementation on circulating markers of inflammation, adipokines, plasma fatty acid composition, blood lipid profile, anthropometrics, oxylipin and cytokine profiles of stimulated immune cells, monocyte glucose metabolism, blood pressure and pulse wave velocity. ETHICS AND SIGNIFICANCE: This trial has been approved by the University of Manitoba Biomedical Research Ethics Board and the St. Boniface Hospital Research Review Committee. The study will provide information regarding the effects of ALA and DHA supplementation on oxylipin profiles in obese but otherwise healthy females. Additionally, it will improve our understanding of the response of circulating inflammatory mediators originating from immune cells, adipose tissue and the liver to n-3 PUFA supplementation in relation to the metabolic features of obesity.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Fish Oils/therapeutic use , Inflammation/metabolism , Linseed Oil/therapeutic use , Obesity/drug therapy , Oxylipins/metabolism , alpha-Linolenic Acid/therapeutic use , Adipokines/metabolism , Adult , Blood Glucose/metabolism , Cross-Over Studies , Cytokines/metabolism , Dietary Supplements , Double-Blind Method , Fatty Acids, Omega-3 , Female , Humans , Middle Aged , Obesity/metabolism , Randomized Controlled Trials as Topic , Young Adult
17.
Int J Food Sci Nutr ; 72(3): 386-390, 2021 May.
Article in English | MEDLINE | ID: mdl-32746658

ABSTRACT

Consumption of omega-3 fatty acids, including the precursor α-linolenic acid (ALA) is often sub-optimal and not in line with international guidelines. Supplementation is debatable, but some individuals, e.g., pre-diabetic, low-grade inflammation, cardiometabolic yet otherwise healthy subjects, might benefit from supra-physiological omega-3 intake, particularly to lessen inflammation. We explored the feasibility of a large clinical trial by performing a pilot study to evaluate adherence, palatability, and self-reported side effects of ALA administration in a group of volunteers. We enrolled 12 individuals with borderline dyslipidemia or overweight, treated with dietary advice according to international guidelines and who had insufficient intakes of essential fatty acids. Subjects were followed for nutritional counselling and were matched with appropriate controls. Patients were administered 6 g/day of ALA, for two months. We report the absence of side effects. such as fishy aftertaste and gastrointestinal distress, in addition to a slight decrease of C-reactive protein concentrations (Identifier: ISRCTN13118704).


Subject(s)
Dietary Supplements , Inflammation/drug therapy , alpha-Linolenic Acid/administration & dosage , alpha-Linolenic Acid/therapeutic use , Adult , Aged , Blood Pressure , C-Reactive Protein , Diet , Fatty Acids, Essential/metabolism , Feasibility Studies , Female , Heart , Humans , Male , Middle Aged , Overweight/drug therapy , Patient Compliance , Pilot Projects
18.
Am J Physiol Endocrinol Metab ; 319(5): E877-E892, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32893672

ABSTRACT

Free fatty acid (FFA) receptors FFA1 and FFA4 are omega-3 molecular targets in metabolic diseases; however, their function in cancer cachexia remains unraveled. We assessed the role of FFA1 and FFA4 receptors in the mouse model of cachexia induced by Lewis lung carcinoma (LLC) cell implantation. Naturally occurring ligands such as α-linolenic acid (ALA) and docosahexaenoic acid (DHA), the synthetic FFA1/FFA4 agonists GW9508 and TUG891, or the selective FFA1 GW1100 or FFA4 AH7614 antagonists were tested. FFA1 and FFA4 expression and other cachexia-related parameters were evaluated. GW9508 and TUG891 decreased tumor weight in LLC-bearing mice. Regarding cachexia-related end points, ALA, DHA, and the preferential FFA1 agonist GW9508 rescued body weight loss. Skeletal muscle mass was reestablished by ALA treatment, but this was not reflected in the fiber cross-sectional areas (CSA) measurement. Otherwise, TUG891, GW1100, or AH7614 reduced the muscle fiber CSA. Treatments with ALA, GW9508, GW1100, or AH7614 restored white adipose tissue (WAT) depletion. As for inflammatory outcomes, ALA improved anemia, whereas GW9508 reduced splenomegaly. Concerning behavioral impairments, ALA and GW9508 rescued locomotor activity, whereas ALA improved motor coordination. Additionally, DHA improved grip strength. Notably, GW9508 restored abnormal brain glucose metabolism in different brain regions. The GW9508 treatment increased leptin levels, without altering uncoupling protein-1 downregulation in visceral fat. LLC-cachectic mice displayed FFA1 upregulation in subcutaneous fat, but not in visceral fat or gastrocnemius muscle, whereas FFA4 was unaltered. Overall, the present study shed new light on FFA1 and FFA4 receptors' role in metabolic disorders, indicating FFA1 receptor agonism as a promising strategy in mitigating cancer cachexia.


Subject(s)
Body Weight/drug effects , Cachexia/drug therapy , Carcinoma, Lewis Lung/metabolism , Docosahexaenoic Acids/therapeutic use , Receptors, G-Protein-Coupled/metabolism , alpha-Linolenic Acid/therapeutic use , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cachexia/etiology , Cachexia/metabolism , Carcinoma, Lewis Lung/complications , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Methylamines/pharmacology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Neoplasm Transplantation , Phenylpropionates/pharmacology , Propionates/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Sulfonamides/pharmacology , Xanthenes/pharmacology , alpha-Linolenic Acid/pharmacology
19.
Oxid Med Cell Longev ; 2020: 8908901, 2020.
Article in English | MEDLINE | ID: mdl-32832007

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder. Amyloid ß- (Aß-) induced mitochondrial dysfunction may be a primary process triggering all the cascades of events that lead to AD. Therefore, identification of natural factors and endogenous mechanisms that protect neurons against Aß toxicity is needed. In the current study, we investigated whether alpha-linolenic acid (ALA), as a natural product, would increase insulin and IGF-I (insulin-like growth factor I) release from astrocytes. Moreover, we explored the protective effect of astrocytes-derived insulin/IGF-I on Aß-induced neurotoxicity, with special attention paid to their impact on mitochondrial function of differentiated SH-SY5Y cells. The results showed that ALA induced insulin and IGF-I secretion from astrocytes. Our findings demonstrated that astrocyte-derived insulin/insulin-like growth factor I protects differentiated SH-SY5Y cells against Aß 1-42-induced cell death. Moreover, pretreatment with conditioned medium (CM) and ALA-preactivated CM (ALA-CM) protected the SH-SY5Y cells against Aß 1-42-induced mitochondrial dysfunction by reducing the depolarization of the mitochondrial membrane, increasing mitochondrial biogenesis, restoring the balance between fusion and fission processes, and regulation of mitophagy and autophagy processes. Our study suggested that astrocyte-derived insulin/insulin-like growth factor I suppresses Aß 1-42-induced cytotoxicity in the SH-SY5Y cells by protecting against mitochondrial dysfunction. Moreover, the neuroprotective effects of CM were intensified by preactivation with ALA.


Subject(s)
Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/drug effects , Astrocytes/metabolism , Neurodegenerative Diseases/drug therapy , Oxidative Stress/drug effects , alpha-Linolenic Acid/therapeutic use , Cell Differentiation , Cell Line, Tumor , Humans , Signal Transduction
20.
Biochem Pharmacol ; 180: 114152, 2020 10.
Article in English | MEDLINE | ID: mdl-32679125

ABSTRACT

α-Linolenic acid (ALA), an essential fatty acid, has anticancer activity in breast cancer, but the mechanism of its effects in triple-negative breast cancer (TNBC) remains unclear. We investigated the effect of ALA on Twist1, which is required to initiate epithelial-mesenchymal transition (EMT) and promotes tumor metastasis, and Twist1-mediated migration in MDA-MB231, MDA-MB468 and Hs578T cells. Twist1 protein was constitutively expressed in these TNBC cells, particularly MDA-MB-231 cells. Treatment with 100 µM ALA and Twist1 siRNA markedly decreased the Twist1 protein level and cell migration. Moreover, ALA transiently attenuated the nuclear accumulation of STAT3α as well as Twist1 mRNA expression. Treatment with ALA significantly attenuated the phosphorylation of JNK, ERK and Akt and decreased the phosphorylation of Twist1 at serine 68 in MDA-MB-231 cells. ALA accelerated Twist1 degradation in the presence of cycloheximide, whereas the ubiquitination and degradation of Twist1 by ALA was suppressed by MG-132. Pretreatment with ALA mimicked Twist1 siRNA, increased the protein expression of epithelial markers such as E-cadherin, and decreased the protein expression of mesenchymal markers including Twist1, Snail2, N-cadherin, vimentin, and fibronectin. Our findings suggest that ALA can be used not only to abolish EMT but also to suppress Twist1-mediated migration in TNBC cells.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/biosynthesis , alpha-Linolenic Acid/pharmacology , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Nuclear Proteins/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Twist-Related Protein 1/genetics , alpha-Linolenic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...