Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Brain Behav Immun ; 118: 192-201, 2024 May.
Article in English | MEDLINE | ID: mdl-38432599

ABSTRACT

Despite decades of research on the pathophysiology of depression, the development of new therapeutic interventions has been slow, and no biomarkers of treatment response have been clinically implemented. Several lines of evidence suggest that the clinical and biological heterogeneity among patients with major depressive disorder (MDD) has hampered progress in this field. MDD with low-grade inflammation - "inflamed depression" - is a subtype of depression that may be associated with a superior antidepressant treatment response to anti-inflammatory compounds. Omega-3 fatty acid eicosapentaenoic acid (EPA) has anti-inflammatory properties, and preliminary data suggest that it may be particularly efficacious in inflamed depression. In this study we tested the hypothesis that add-on EPA has greater antidepressant efficacy in MDD patients with high baseline high-sensitivity C-reactive protein (hs-CRP) compared to MDD patients with low hs-CRP. All subjects received 2.2 g EPA, 400 mg docosahexaenoic acid and 800 mg of other fatty acids daily for 8 weeks, added to stable ongoing antidepressant treatment. The primary outcome was change in the 17-item Hamilton Depression Rating Scale (HAMD-17). Patients and raters were blind to baseline hs-CRP status. In an intention-to-treat analysis including all subjects with at least one post baseline visit (n = 101), ahs-CRPcut-off of ≥1 mg/L, but not ≥3 mg/L, was associated with a greater improvement in HAMD-17 total score. In addition to a general antidepressant effect among patients with hs-CRP ≥ 1 mg/L, adjuvant EPA treatment improved symptoms putatively related to inflamed depression such as fatigue and sleep difficulties. This adds to the mounting evidence that delineation of MDD subgroups based on inflammation may be clinically relevant to predict treatment response to anti-inflammatory interventions.


Subject(s)
Depressive Disorder, Major , Fatty Acids, Omega-3 , Humans , Fatty Acids, Omega-3/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/diagnosis , Depression/drug therapy , C-Reactive Protein/metabolism , Eicosapentaenoic Acid/therapeutic use , Docosahexaenoic Acids/therapeutic use , Antidepressive Agents/therapeutic use , Inflammation/drug therapy , Inflammation/chemically induced , Anti-Inflammatory Agents/therapeutic use
2.
Nutrients ; 16(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38474759

ABSTRACT

Inefficient glucose metabolism and decreased ATP production in the brain are linked to ageing, cognitive decline, and neurodegenerative diseases (NDDs). This study employed thermodynamic analysis to assess the effect of fish oil supplementation on glucose metabolism in ageing brains. Data from previous studies on glucose metabolism in the aged human brain and grey mouse lemur brains were examined. The results demonstrated that Omega-3 fish oil supplementation in grey mouse lemurs increased entropy generation and decreased Gibbs free energy across all brain regions. Specifically, there was a 47.4% increase in entropy generation and a 47.4 decrease in Gibbs free energy in the whole brain, indicating improved metabolic efficiency. In the human model, looking at the specific brain regions, supplementation with Omega-3 polyunsaturated fatty acids (n-3 PUFAs) reduced the entropy generation difference between elderly and young individuals in the cerebellum and particular parts of the brain cortex, namely the anterior cingulate and occipital lobe, with 100%, 14.29%, and 20% reductions, respectively. The Gibbs free energy difference was reduced only in the anterior cingulate by 60.64%. This research underscores that the application of thermodynamics is a comparable and powerful tool in comprehending the dynamics and metabolic intricacies within the brain.


Subject(s)
Fatty Acids, Omega-3 , Fish Oils , Humans , Aged , Fish Oils/metabolism , Glucose/metabolism , Fatty Acids, Omega-3/metabolism , Brain/metabolism , Diet , Thermodynamics , Dietary Supplements
3.
J Nutr Biochem ; 127: 109603, 2024 May.
Article in English | MEDLINE | ID: mdl-38373507

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disease that causes progressive cognitive decline. A major pathological characteristic of AD brain is the presence of senile plaques composed of ß-amyloid (Aß), the accumulation of which induces toxic cascades leading to synaptic dysfunction, neuronal apoptosis, and eventually cognitive decline. Dietary n-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are beneficial for patients with early-stage AD; however, the mechanisms are not completely understood. In this study, we investigated the effects of n-3 PUFAs on Aß-induced toxicity in a transgenic AD Caenorhabditis elegans (C. elegans) model. The results showed that EPA and DHA significantly inhibited Aß-induced paralytic phenotype and decreased the production of reactive oxygen species while reducing the levels of Aß in the AD worms. Further studies revealed that EPA and DHA might reduce the accumulation of Aß by restoring the activity of proteasome. Moreover, treating worms with peroxisome proliferator-activated receptor (PPAR)-γ inhibitor GW9662 prevented the inhibitory effects of n-3 PUFAs on Aß-induced paralytic phenotype and diminished the elevation of proteasomal activity by n-3 PUFAs, suggesting that PPARγ-mediated signals play important role in the protective effects of n-3 PUFAs against Aß-induced toxicity.


Subject(s)
Alzheimer Disease , Fatty Acids, Omega-3 , Neurodegenerative Diseases , Animals , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/pharmacology , PPAR gamma/genetics , Disease Models, Animal
4.
Nutrients ; 16(4)2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38398839

ABSTRACT

BACKGROUND: Plant-based dietary patterns are a source of different amounts and proportions of fatty acids (FAs) from those in traditional diets. Information about the full FAs profile provided by plant-based diets is widely lacking. The aim of this study was to present the exact serum profiles of FAs among people on a plant-based diet compared with omnivorous subjects. METHODS: FAs compositions and inflammation statuses (based on serum C-reactive protein (CRP) levels) were studied in serum samples obtained from 102 female volunteers (divided into four groups: vegans, vegetarians, pescatarians, and omnivores). The quality of the volunteers' diets was assessed based on seven-day dietary records. RESULTS: Both vegans and vegetarians had lower total n-3 PUFAs, EPA, and DHA serum levels than omnivores. Decreased levels of these FAs presumably did not cause inflammation in vegetarians and vegans, as vegetarians had similar serum levels of CRP compared to omnivores, and vegans had even lower levels. CONCLUSION: The analysis of serum FAs and CRP levels in vegetarians and vegans suggests that factors other than diet alone influence inflammation and overall health status. Further research on long-term plant-based diet users is needed to better understand this issue, and supplementation with EPA and DHA is worth considering in vegans and vegetarians.


Subject(s)
Diet, Vegetarian , Fatty Acids , Humans , Female , Diet , Vegetarians , Diet, Vegan , Health Status , Inflammation
5.
Curr Top Med Chem ; 24(1): 45-59, 2024.
Article in English | MEDLINE | ID: mdl-37907485

ABSTRACT

Cancer ranks as the second leading cause of mortality in high-income countries, underscoring the critical need for effective therapeutic strategies. One prominent approach, chemotherapy, is widely employed for treating solid tumors. However, the significant adverse effects associated with chemotherapy, notably myeloablation and osteonecrosis, impart considerable challenges by compromising immune function and diminishing patients' quality of life. Furthermore, the emergence of chemotherapy resistance poses a formidable hurdle in achieving successful cancer treatment outcomes. In this context, the focus is on exploring alternative approaches to enhance the efficacy of cancer treatment and mitigate its adverse consequences. Among these approaches, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), two n-3 polyunsaturated fatty acids (PUFAs), have garnered substantial interest. These PUFAs exhibit the potential to influence membrane lipid composition and modulate critical gene expressions associated with cancer, such as Bcl-2, PI3K, NF-κB, and phosphorylated Akt, thereby potentially reducing cancer risk. Moreover, emerging evidence highlights their ability to augment chemotherapy efficacy, particularly in drug-resistant cancer cells. Importantly, both preclinical and clinical investigations have provided compelling evidence supporting the protective effects of n-3 PUFAs on healthy cells. Leveraging these findings, there has been growing attention on the exploration of n-3 PUFAs as adjuvants to chemotherapy. This strategic approach holds promise in mitigating the adverse effects linked to chemotherapy, notably myeloablation and osteonecrosis, while simultaneously enhancing its effectiveness in combating cancer. This comprehensive review delves into the multifaceted attributes of n-3 PUFAs, encompassing their cytotoxic properties, potential as chemopreventive agents, and their prospective role in ameliorating the adverse effects commonly associated with chemotherapy, with a particular emphasis on myeloablation and osteonecrosis. By elucidating the intricate interplay between n-3 PUFAs and cancer treatment paradigms, this review contributes to the expanding body of knowledge aimed at refining cancer therapeutic strategies and enhancing patient outcomes.


Subject(s)
Fatty Acids, Omega-3 , Neoplasms , Osteonecrosis , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Quality of Life , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Neoplasms/drug therapy , Osteonecrosis/drug therapy
6.
Colloids Surf B Biointerfaces ; 234: 113702, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38113752

ABSTRACT

Promising findings have been emerged from studies utilizing n3 polyunsaturated fatty acids (PUFA) supplementation in animal models of inflammatory bowel disease (IBD). Introduction of marine phospholipids which combine n3 PUFA with phosphatidylcholine in a nanoliposome formulation offers enhanced pharmacological efficacy due to physical stability, improved bioavailability, and specific targeting to inflamed colitis tissues. In the present study, a marine phospholipid-based nanoliposome formulation was developed and optimized, resulting in nanovesicles of approximately 107.7 ± 1.3 nm in size, 0.18 ± 0.01 PDI, and - 32.03 ± 3.16 mV ZP. The nanoliposomes exhibited spherical vesicles with stable properties upon incubation at SGF as shown by the TEM, DLS, and turbidity measurements over 3 h. MPL nanoliposomes were cytocompatible until the concentration of 500 µg/mL as per MTT assay and taken by macrophages through macropinocytosis and caveolae pathways, and demonstrated significant inhibitory activity against reactive oxygen species (ROS) in LPS-stimulated macrophages. They were also shown to be blood-compatible and safe for administration in healthy mice. In a colitis mouse model, the nanoliposomes displayed preferential distribution in the inflamed gut, delaying the onset of colitis when administered prophylactically. These findings highlight the potential of marine phospholipid nanoliposomes as a promising therapeutic approach for managing inflammatory bowel disease.


Subject(s)
Colitis , Fatty Acids, Omega-3 , Inflammatory Bowel Diseases , Animals , Mice , Phospholipids , Inflammatory Bowel Diseases/drug therapy , Colitis/chemically induced , Colitis/drug therapy , Phosphatidylcholines , Liposomes
7.
Pediatr Allergy Immunol ; 34(9): e14024, 2023 09.
Article in English | MEDLINE | ID: mdl-37747750

ABSTRACT

BACKGROUND: Asthma is an inflammatory disease. The potential of n-3 polyunsaturated fatty acids (PUFAs) to alleviate asthma symptoms through their anti-inflammatory effects and immune modulation has been explored. However, the precise role of dietary n-3 PUFAs in childhood and adolescent asthma remains unclear. OBJECTIVE: This study aimed to evaluate the association between dietary n-3 PUFAs intake and asthma in children and adolescents in the United States. METHODS: We conducted a cross-sectional analysis of 8543 children and adolescents from the National Health and Nutrition Examination Survey (NHANES) between 2013 and 2020 by adjusting for covariates and using multivariate logistic regression, restricted cubic spline, threshold effects, and subgroup analyses. RESULTS: Among 8354 participants, 1456 (16.5%) self-reported diagnosis of asthma by a healthcare provider. After adjusting for potential confounding factors, compared with individuals in the lowest n-3 PUFA consumption group (T1, <26.07 mg/kg/day), the adjusted odds ratio (OR) for asthma was 0.71 (95% CI: 0.6-0.84, p < .001) in the second group (T2, 26.07-48.93 mg/kg/day) and 0.58 (95% CI: 0.47-0.73, p < .001) in the third group (T3, >48.93 mg/kg/day). Furthermore, a nonlinear (L-shaped) relationship was observed between n-3 PUFA intake and asthma (p = .009), with subgroup and sensitivity analyses confirming the stability of the results. In the threshold analysis, a critical turning point was observed at approximately 59.0 mg/kg/day (OR = 0.984, 95% CI: 0.977-0.991, p < .001). CONCLUSION: Dietary intake of n-3 PUFAs exhibited an L-shaped relationship with asthma in children and adolescents in the United States, with a critical turning point observed at approximately 59.0 mg/kg/day.


Subject(s)
Asthma , Fatty Acids, Omega-3 , Adolescent , Humans , Child , Cross-Sectional Studies , Nutrition Surveys , Asthma/epidemiology , Fatty Acids
8.
Biomolecules ; 13(9)2023 09 19.
Article in English | MEDLINE | ID: mdl-37759812

ABSTRACT

Psoriasis is a skin disease characterized by epidermal hyperplasia and an inappropriate activation of the adaptive immunity. A dysregulation of the skin's lipid mediators is reported in the disease with a predominance of the inflammatory cascade derived from n-6 polyunsaturated fatty acids (n-6 PUFAs). Bioactive lipid mediators derived from arachidonic acid (AA) are involved in the inflammatory functions of T cells in psoriasis, whereas n-3 PUFAs' derivatives are anti-inflammatory metabolites. Here, we sought to evaluate the influence of a supplementation of the culture media with eicosapentaenoic acid (EPA) on the lipid profile of a psoriatic skin model produced with polarized T cells. Healthy and psoriatic skin substitutes were produced following the auto-assembly technique. Psoriatic skin substitutes produced with or without T cells presented increased epidermal and dermal linolenic acid (LA) and AA levels. N-6 PUFA lipid mediators were strongly measured in psoriatic substitutes, namely, 13-hydroxyoctadecadienoic acid (13-HODE), prostaglandin E2 (PGE2) and 12-hydroxyeicosatetraenoic acid (12-HETE). The added EPA elevated the amounts of EPA, n-3 docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA) in the epidermal and dermal phospholipids. The EPA supplementation balanced the production of epidermal lipid mediators, with an increase in prostaglandin E3 (PGE3), 12-hydroxyeicosapentaenoic acid (12-HEPE) and N-eicosapentaenoyl-ethanolamine (EPEA) levels. These findings show that EPA modulates the lipid composition of psoriatic skin substitutes by encouraging the return to a cutaneous homeostatic state.


Subject(s)
Fatty Acids, Omega-3 , Psoriasis , Skin Diseases , Humans , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/metabolism , T-Lymphocytes/metabolism , Fatty Acids, Omega-6 , Eicosanoids , Arachidonic Acid/metabolism , Dinoprostone
9.
Psychopharmacology (Berl) ; 240(9): 1877-1889, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37612456

ABSTRACT

RATIONALE: The growing evidence has demonstrated the importance of endoplasmic reticulum stress (ERS) in the pathophysiology of depression. ERS genes were considered to be potential novel therapeutic targets for depression. OBJECTIVES: To clarify the mechanisms of the chronic unpredictable mild stress (CUMS)-induced ERS response and the potential contributing pathways in depression, and further investigate the potential link between N-3 polyunsaturated fatty acids (PUFAs) and stress-induced ERS disturbances. METHODS: This study analyzed the expression of ERS-related genes including GRP78, ATF-4, ATF-6, XBP-1, and CHOP, and sigma-1R with real-time PCR in peripheral blood mononuclear cell (PBMC) RNA samples from participants. All of the rats except for those in the control groups were subjected to 5 consecutive weeks of CUMS to establish the depression model, and the antidepressant effects of N-3 PUFAs were observed by behavior tests. Moreover, the effect of diet and stress on the ERS pathways was also investigated using the western blot. RESULTS: Blood CHOP, ATF-4, and XBP-1 levels were notably elevated in depressed patients relative to healthy individuals. Moreover, increased sigma-1R and decreased ATF-6 implied the protective role of sigma-1R through modulating ERS in patients with depression. Animal studies disclosed the novel findings that supplementary N-3 PUFAs in rats alleviated CUMS-induced disturbance of ERS through the ATF-4/XBP-1/CHOP pathway, implying its potential strategy for depression. CONCLUSION: CUMS-induced depressive-like behaviors are related to the disturbance of ERS. Furthermore, supplementary N-3 PUFAs might be an effective way to alleviate ERS.


Subject(s)
Fatty Acids, Omega-3 , Animals , Rats , Fatty Acids, Omega-3/pharmacology , Leukocytes, Mononuclear , Inhibition, Psychological , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Endoplasmic Reticulum Stress
10.
Mar Drugs ; 21(6)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37367679

ABSTRACT

The growth and development of the fetus and newborn throughout pregnancy and lactation are directly related to the nutritional status of the mother, which has a significant impact on the health of the offspring. The purpose of this experiment was to investigate the susceptibility of n-3 polyunsaturated fatty acid deficiency in early life to seizures in adulthood. The n-3 PUFAs-deficient mice's offspring were established and then fed with α-LNA diet, DHA-enriched ethyl ester, and DHA-enriched phospholipid-containing diets for 17 days at the age of eight weeks. During this period, animals received intraperitoneal injections of 35 mg/kg of pentylenetetrazol (PTZ) every other day for eight days. The results showed that dietary n-3 PUFA-deficiency in early life could aggravate PTZ-induced epileptic seizures and brain disorders. Notably, nutritional supplementation with n-3 PUFAs in adulthood for 17 days could significantly recover the brain n-3 fatty acid and alleviate the epilepsy susceptibility as well as raise seizure threshold to different levels by mediating the neurotransmitter disturbance and mitochondria-dependent apoptosis, demyelination, and neuroinflammation status of the hippocampus. DHA-enriched phospholipid possessed a superior effect on alleviating the seizure compared to α-LNA and DHA-enriched ethyl ester. Dietary n-3 PUFA deficiency in early life increases the susceptibility to PTZ-induced epilepsy in adult offspring, and nutritional supplementation with n-3 PUFAs enhances the tolerance to the epileptic seizure.


Subject(s)
Epilepsy , Fatty Acids, Omega-3 , Female , Pregnancy , Mice , Animals , Pentylenetetrazole/toxicity , Docosahexaenoic Acids , Fatty Acids, Omega-3/pharmacology , Diet , Phospholipids , Dietary Supplements , Epilepsy/chemically induced , Epilepsy/drug therapy , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control
11.
Foods ; 12(9)2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37174357

ABSTRACT

Previous attempts to increase the level of flaxseed in hens' diet for the production of n-3 polyunsaturated fatty acids (n-3 PUFAs)-enriched eggs have been commonly associated with undesirable effects on production efficiency, lipid health indices, and oxidative stability of eggs, requiring adequate research attention. This study investigated the effects of feeding a moderate level of flaxseed (FS) and plant polyphenol extracts (PPEs) on fatty acid content, oxidative stability, and lipid health indices in eggs of slow-growing Sasso T451A laying hens. One hundred and five hens were assigned to five groups (seven replicates of three) and fed on FS (75 g flaxseed and no antioxidants), VE8 (75 g flaxseed and 800 mg vitamin E), TS8 (75 g flaxseed and 800 mg Thymus schimperi), DA8 (75 g flaxseed and 800 mg Dodonaea angustifolia), and CD8 (75 g flaxseed and 800 mg Curcuma domestica) extract per kg diets. The egg yolk content of eicosapentaenoic acid (EPA, C20:5 n-3) in the DA8, TS8, and CD8 diets and docosahexaenoic acid (DHA, C22:6 n-3) in TS8 and CD8 diets significantly (p < 0.05) increased compared with the FS diet. The FS diet significantly increased the malondialdehyde (MDA) content in egg yolks, whereas the TS8 diet decreased it by 67% (p < 0.05). Little difference was observed in yolk fatty acid content between cooked and raw eggs. Production of n-3 PUFA-enriched eggs with favorable lipid health indices was possible through inclusion of PPEs extracted from local plant species grown in Ethiopia and a moderate dose of flaxseed in the diet of laying hens.

12.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047706

ABSTRACT

The quantitative polymerase chain reaction (qRT-PCR) technique gives promising opportunities to detect and quantify RNA targets and is commonly used in many research fields. This study aimed to identify suitable reference genes for physical exercise and omega-3 fatty acids supplementation intervention. Forty healthy, physically active men were exposed to a 12-week eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) supplementation and standardized endurance training protocol. Blood samples were collected before and after the intervention and mRNA levels of six potential reference genes were tested in the leukocytes of 18 eligible participants using the qRT-PCR method: GAPDH (Glyceraldehyde-3-phosphate dehydrogenase), ACTB (Beta actin), TUBB (Tubulin Beta Class I), RPS18 (Ribosomal Protein S18), UBE2D2 (Ubiquitin-conjugating enzyme E2 D2), and HPRT1 (Hypoxanthine Phosphoribosyltransferase 1). The raw quantification cycle (Cq) values were then analyzed using RefFinder, an online tool that incorporates four different algorithms: NormFinder, geNorm, BestKeeper, and the comparative delta-Ct method. Delta-Ct, NormFinder, BestKeeper, and RefFinder comprehensive ranking have found GAPDH to be the most stably expressed gene. geNorm has identified TUBB and HPRT as the most stable genes. All algorithms have found ACTB to be the least stably expressed gene. A combination of the three most stably expressed genes, namely GAPDH, TUBB, and HPRT, is suggested for obtaining the most reliable results.


Subject(s)
Fatty Acids, Omega-3 , Hypoxanthine Phosphoribosyltransferase , Male , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Polymerase Chain Reaction , Exercise , Dietary Supplements , Real-Time Polymerase Chain Reaction/methods , Gene Expression Profiling/methods , Reference Standards
13.
Nutr Res ; 110: 74-86, 2023 02.
Article in English | MEDLINE | ID: mdl-36689814

ABSTRACT

Obesity is a chronic metabolic disease that involves excessive accumulation of fat in white adipose tissue (WAT). Apart from storing excess fats, WAT also serves as an important endocrine organ secreting adipocytokines such as adiponectin and leptin. Adiponectin and leptin bind to their transmembrane receptors adiponectin receptor 1 (AdipoR1)/adiponectin receptor 2 (AdipoR2) and Ob-R, respectively, and mediate their effect on metabolism by regulating multiple downstream targets. Dietary fat is considered the main culprit behind obesity development. Numerous preclinical studies have highlighted role of essential polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, in prevention of obesity. Despite emerging data, there still is no clear understanding of the mechanism of action of n-3 PUFAs and n-6 PUFAs on adipose tissue function in two functionally and anatomically different depots of WAT: visceral and subcutaneous. We designed this study using a high fat diet (HFD) fed rodent model of obesity to test our hypothesis that n-3 and n-6 PUFAs possibly differentially modulate adipokine secretion and downstream metabolic pathways such as peroxisome proliferator-activated receptor-γ (PPAR-γ), protein kinase B (AKT)-forkhead box O1 (FOXO1), and Janus kinase-signal transducer and activator of transcription in obesity. The results of the current study showed that n-3 PUFAs upregulate the expression of AdipoR1/R2 and ameliorate the effects of HFD by modulating adipogenesis via PPAR-γ and by improving glucose tolerance and lipid metabolism via AKT-FOXO1 axis in fish oil fed rats. However, n-6 PUFAs did not show any remarkable change compared with HFD fed animals. Our study highlights that n-3 PUFAs modulate expression of various targets in adiponectin and leptin signaling cascade, bringing about an overall reduction in obesity and improvement in adipose tissue function in HFD induced obesity.


Subject(s)
Diet, High-Fat , Fatty Acids, Omega-3 , Rats , Animals , Diet, High-Fat/adverse effects , Adiponectin , Leptin/metabolism , Rats, Wistar , Proto-Oncogene Proteins c-akt/metabolism , Fatty Acids, Omega-6/pharmacology , Receptors, Adiponectin/metabolism , Receptors, Adiponectin/therapeutic use , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Obesity/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Adipokines/metabolism , Fatty Acids, Unsaturated/metabolism , Signal Transduction
14.
Arch Gynecol Obstet ; 307(1): 249-262, 2023 01.
Article in English | MEDLINE | ID: mdl-35348829

ABSTRACT

BACKGROUND: Maternal omega-3 consumption during pregnancy has been positively linked with a positive impact on maternal health and fetal growth. However, the results of individual studies are inconsistent and conflicting. OBJECTIVE: Examine the effect of supplementation with DHA, and/or EPA, and/or ALA throughout pregnancy on offspring's growth and pregnancy outcomes. DESIGN: A systematic review and meta-analysis. POPULATION: Pregnant women. METHODS: According to (PRISMA) statement and the Cochrane Handbook guidelines. Human trials (RCT or quasi-RCT) which involved oral omega-3 supplementation at least twice a week during pregnancy were included and comparing it with control groups with no supplementation or placebo administration. Data were extracted and directed using RevMan software. Fifty-nine randomized controlled trials were eligible for inclusion in the meta-analysis. Performed in MEDLINE, PubMed, Scopus, Google Scholar, and the Cochrane Library comparing omega 3 with control groups, from 1990 to 2020. THE MAIN OUTCOME MEASURES: The primary outcome measures were pregnancy-induced hypertension, preeclampsia, gestational duration, preterm birth, early preterm birth, birth weight, low birth weight, neonatal length, and head circumference. The secondary outcomes were neonatal intensive care unit, infant death, prenatal death, and cesarean section. RESULTS: In 24 comparisons (21,919 women) n-3 fatty acids played a protective role against the risk of preeclampsia (RR = 0.84, 95% CI 0.74-0.96 p = 0.008; I2 = 24%). In 46 comparisons (16,254 women) n-3 fatty acids were associated with a significantly greater duration of pregnancy (MD = 1.35, 95% CI 0.65-2.05, p = 0.0002; I2 = 59%). 27 comparisons (15,510 women) was accompanied by a significant decrease in pre-term birth less than 37 weeks (RR = 0.86, 95% CI 0.77-0.95, p = 0.005; I2 = 0%). 12 comparisons (11,774 women) was accompanied by a significant decrease in early pre-term birth less than 34 weeks (RR = 0.77, 95% CI 0.63-0.95, p = 0.01; I2 = 40%). 38 comparisons (16,505 infants) had a significant increase in birth weight (MD = 49.19, 95% CI 28.47-69.91, p < 0.00001; I2 = 100%). Finally, 14 comparisons (8,449 infants) had a borderline significance in increase in low birth weight (RR = 0.88, 95% CI 0.78-1.00, p = 0.05; I2 = 28%). CONCLUSIONS: Supplementation with omega-3 in prgnancy can prevent preeclampsia, increase gestational duration, increase birth weight and decrease the risk of low birth weight and preterm birth.


Subject(s)
Fatty Acids, Omega-3 , Pre-Eclampsia , Premature Birth , Infant , Pregnancy , Female , Infant, Newborn , Humans , Pre-Eclampsia/prevention & control , Premature Birth/prevention & control , Dietary Supplements , Cesarean Section , Birth Weight , Maternal Health , Pregnancy Outcome
15.
Biomed Pharmacother ; 157: 114091, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36481403

ABSTRACT

Rosacea is a facial chronic inflammatory skin disease with dysfunction of immune and neurovascular system and treatments for rosacea are challenging. N-3 polyunsaturated fatty acids (PUFAs), one of essential fatty acids, are needed for health maintenance and exert anti-inflammation and immunomodulatory effects in a series of cutaneous diseases such as atopic dermatitis and photoaging through dietary supplementation. However, the role of n-3 PUFAs on rosacea remains to be elucidated. In this study, KEGG enrichment analysis and GO analysis indicated that the biological process and signaling pathways, including chemokine signaling pathway, regulated by n-3 PUFAs highly overlapped with those in the pathogenic biological process of rosacea, especially the erythema telangiectasia type. Next, mice were randomized to fed with a customized n-3 PUFAs diet. We showed that n-3 PUFAs ameliorated skin erythema, inhibited dermal inflammatory cell infiltration (mast cells, neutrophils, and CD4 +T cells) and suppressed elevated pro-inflammatory cytokines in LL37-induced rosacea-like mice. Besides, n-3 PUFAs were also verified to repress angiogenesis in LL37-induced mice skin. Further investigation revealed that n-3 PUFAs attenuated LL37-induced inflammation via TLR2/ MyD88/ NF-κB pathway both in mice and in keratinocytes. In conclusion, our findings underscore that dietary supplementation of n-3 PUFAs have the potential to become an efficient and safe clinical therapeutic candidate for rosacea.


Subject(s)
Fatty Acids, Omega-3 , Rosacea , Animals , Mice , Dietary Supplements , Erythema , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Rosacea/chemically induced , Rosacea/drug therapy , Toll-Like Receptor 2/metabolism
16.
Mar Drugs ; 22(1)2023 Dec 27.
Article in English | MEDLINE | ID: mdl-38248642

ABSTRACT

Inflammation is a conserved process that involves the activation of immune and non-immune cells aimed at protecting the host from bacteria, viruses, toxins and injury. However, unresolved inflammation and the permanent release of pro-inflammatory mediators are responsible for the promotion of a condition called "low-grade systemic chronic inflammation", which is characterized by tissue and organ damage, metabolic changes and an increased susceptibility to non-communicable diseases. Several studies have demonstrated that different dietary components may influence modifiable risk factors for diverse chronic human pathologies. Marine n-3 polyunsaturated fatty acids (n-3 PUFAs), mainly eicosapentaenoic (EPA) and docosahexaenoic acid (DHA), are well-recognized anti-inflammatory and immunomodulatory agents that are able to influence many aspects of the inflammatory process. The aim of this article is to review the recent literature that relates to the modulation of human disease, such as rheumatoid arthritis, by n-3 PUFAs.


Subject(s)
Arthritis, Rheumatoid , Fatty Acids, Omega-3 , Humans , Arthritis, Rheumatoid/drug therapy , Fatty Acids, Unsaturated , Inflammation/drug therapy , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Risk Factors
17.
Molecules ; 27(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36234919

ABSTRACT

High-fat (HF) diets and low-grade chronic inflammation contribute to the development of insulin resistance and type 2 diabetes (T2D), whereas n-3 polyunsaturated fatty acids (PUFAs), due to their anti-inflammatory effects, protect against insulin resistance. Interleukin (IL)-1ß is implicated in insulin resistance, yet how n-3 PUFAs modulate IL-1ß secretion and attenuate HF diet-induced insulin resistance remains elusive. In this study, a HF diet activated NLRP3 inflammasome via inducing reactive oxygen species (ROS) generation and promoted IL-1ß production primarily from adipose tissue preadipocytes, but not from adipocytes and induced insulin resistance in wild type (WT) mice. Interestingly, endogenous synthesized n-3 polyunsaturated fatty acids (PUFAs) reversed this process in HF diet-fed fat-1 transgenic mice although the HF diet induced higher weight gain in fat-1 mice, compared with the control diet. Mechanistically, palmitic acid (PA), the main saturated fatty acid in an HF diet inactivated AMPK and led to decreased GSK-3ß phosphorylation, at least partially through reducing Akt activity, which ultimately blocked the Nrf2/Trx1 antioxidant pathway and induced TXNIP cytoplasm translocation and NLRP3 inflammasome activation, whereas docosahexaenoic acid (DHA), the most abundant n-3 PUFA in fat-1 adipose tissue, reversed this process via inducing Akt activation. Our GSK-3ß shRNA knockdown study further revealed that GSK-3ß played a pivot role between the upstream AMPK/Akt pathway and downstream Nrf2/Trx1/TXNIP pathway. Given that NLRP3 inflammasome is implicated in the development of most inflammatory diseases, our results suggest the potential of n-3 PUFAs in the prevention or adjuvant treatment of NLRP3 inflammasome-driven diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Fatty Acids, Omega-3 , Insulin Resistance , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Carrier Proteins , Diet, High-Fat/adverse effects , Docosahexaenoic Acids/pharmacology , Fatty Acids/pharmacology , Fatty Acids, Omega-3/pharmacology , Glycogen Synthase Kinase 3 beta , Inflammasomes/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Palmitic Acid/pharmacology , Proto-Oncogene Proteins c-akt , RNA, Small Interfering , Reactive Oxygen Species , Thioredoxins
18.
Immun Inflamm Dis ; 10(11): e707, 2022 11.
Article in English | MEDLINE | ID: mdl-36301036

ABSTRACT

INTRODUCTION: n-3 polyunsaturated fatty acids (PUFAs) are believed to be implicated in the pathogenesis of many inflammation-related diseases, including depression. METHODS: The mouse model of depression was established through chronic unpredictable mild stress (CUMS), the mice were intervened with n-3 PUFAs, and then the expression of toll-like receptor 4 (TLR4) was stimulated with lipopolysaccharides (LPS). Tail suspension test (TST), forced swimming test (FST) and sucrose preference test were performed to monitor the depression behavior of mice. Microglia activation was detected by Iba1 immunofluorescence, and neuronal injury was detected by Nissl staining. Concentrations of tumor necrosis factor (TNF)-α, Interleukin (IL)-6 and IL-1ß in the hippocampus were assessed via enzyme linked immunosorbent assay (ELISA). Quantitative real time polymerase chain reaction was used to detect IL-6, IL-1ß and TNF-α messenger RNA levels. Western blot was utilized for detection of TLR4 protein expression. RESULTS: CUMS significantly reduced the sucrose preference in mice, while increased the immobility time in FST and TST. Moreover, CUMS significantly aggravated microglia activation and neuronal damage in mice and increased the levels of IL-6, IL-1ß and TNF-α in hippocampal tissues, however, intervention with n-3 PUFAs could improve the above effects. Further, the increased TLR4 induced by LPS partially reversed the inhibition of n-3 PUFAs on depression-like behaviors, microglial activation and inflammatory injury of hippocampal neurons. CONCLUSION: n-3 PUFAs may ameliorate depression-like behaviors via reducing hippocampal neuroinflammation in CUMS-induced mice by regulating TLR4 expression, suggesting that n-3 PUFAs may be an effective antidepressant, which provides evidence for future treatment of depression.


Subject(s)
Fatty Acids, Omega-3 , Toll-Like Receptor 4 , Mice , Animals , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Depression/drug therapy , Depression/etiology , Depression/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/metabolism , Lipopolysaccharides/toxicity , Interleukin-6/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , Behavior, Animal , Hippocampus/metabolism , Sucrose/metabolism , Sucrose/pharmacology
19.
Front Nutr ; 9: 985451, 2022.
Article in English | MEDLINE | ID: mdl-36061895

ABSTRACT

Several meta-analyses have revealed that n-3 PUFAs can lower blood pressure, but the findings are conflicting. In this regard, the present umbrella meta-analysis aimed was performed to clarify whether n-3 PUFAs have effects on blood pressure. PubMed, Scopus, Embase, Web of Science, and Google Scholar were used as international databases from inception to May 2022. To examine the effects of n-3 PUFA supplementation on blood pressure, a random-effects model was applied. The leave-one-out method was performed for the sensitivity analysis. The pooled estimate of 10 meta-analyses with 20 effect sizes revealed significant reductions in both systolic (ES = -1.19 mmHg; 95% CI: -1.76, -0.62, p < 0.001) and diastolic blood pressure (ES = -0.91 mmHg, 95% CI: -1.35, -0.47; p < 0.001) following n-3 PUFAs supplementation. In studies with a sample size of ≤ 400 participants and a mean age over 45, SBP and DBP were found to be substantially reduced. Overall, this umbrella meta-analysis indicates that n-3 PUFAs supplementation might play a role in improving DBP and SBP.

20.
Int Immunopharmacol ; 111: 109104, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35914448

ABSTRACT

BACKGROUND: Existing meta-analyses on omega-3 polyunsaturated fatty acids and their anti-inflammatory effects have reported uncertain findings. The current umbrella meta-analysis aimed to assess the findings of multiple meta-analyses on the efficacy of n-3 PUFAs on inflammatory biomarkers in adults with different health conditions. METHODS: Using suitable keywords, articles published until December 2021 were searched in PubMed/Medline, Web of Science, Scopus, EMBASE, and Google Scholar. Meta-analyses investigating the impact of supplementation of n-3 PUFAs on inflammatory biomarkers in adults were included. We performed this meta-analysis using a random-effects model. RESULTS: Overall, 32 meta-analyses were qualified in this umbrella meta-analysis. Our findings demonstrated that the n-3 PUFA supplementation significantly reduced serum C-reactive protein (CRP) (ES = -0.40; 95 % CI: -0.56, -0.24, p < 0.001; I2 = 89.5 %, p < 0.001), Tumour necrosis factor α (TNFα) (ES = -0.23; 95 % CI: -0.37, -0.08, p = 0.002; I2 = 60.1 %, p < 0.001), and interleukin 6 (IL-6) concentrations (ES = -0.22; 95 % CI: -0.39, -0.05, p = 0.010; I2 = 66.2 %, p < 0.001). CONCLUSION: The current umbrella meta-analysis found that supplementation of n-3 PUFAs in adults can improve CRP, TNF-α, and IL-6 concentrations under various health conditions. n-3 PUFAs can be recommended as adjuvant anti-inflammatory agents.


Subject(s)
Fatty Acids, Omega-3 , Interleukin-6 , Biomarkers , C-Reactive Protein/analysis , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Humans , Inflammation/drug therapy , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL