Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.797
1.
Sci Rep ; 14(1): 10947, 2024 05 13.
Article En | MEDLINE | ID: mdl-38740811

The immunomodulatory effects of omega-3 and omega-6 fatty acids are a crucial subject of investigation for sustainable fish aquaculture, as fish oil is increasingly replaced by terrestrial vegetable oils in aquafeeds. Unlike previous research focusing on fish oil replacement with vegetable alternatives, our study explored how the omega-6 to omega-3 polyunsaturated fatty acid (PUFA) ratio in low-fish oil aquafeeds influences Atlantic salmon's antiviral and antibacterial immune responses. Atlantic salmon were fed aquafeeds rich in soy oil (high in omega-6) or linseed oil (high in omega-3) for 12 weeks and then challenged with bacterial (formalin-killed Aeromonas salmonicida) or viral-like (polyriboinosinic polyribocytidylic acid) antigens. The head kidneys of salmon fed high dietary omega-3 levels exhibited a more anti-inflammatory fatty acid profile and a restrained induction of pro-inflammatory and neutrophil-related genes during the immune challenges. The high-omega-3 diet also promoted a higher expression of genes associated with the interferon-mediated signaling pathway, potentially enhancing antiviral immunity. This research highlights the capacity of vegetable oils with different omega-6 to omega-3 PUFA ratios to modulate specific components of fish immune responses, offering insights for future research on the intricate lipid nutrition-immunity interplay and the development of novel sustainable low-fish oil clinical aquaculture feeds.


Aeromonas salmonicida , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Fish Diseases , Salmo salar , Animals , Salmo salar/immunology , Fatty Acids, Omega-6/pharmacology , Fatty Acids, Omega-3/pharmacology , Aeromonas salmonicida/immunology , Fish Diseases/immunology , Fish Diseases/prevention & control , Fish Diseases/virology , Head Kidney/immunology , Animal Feed , Soybean Oil/pharmacology , Fish Oils/pharmacology , Aquaculture/methods
2.
Biomed Khim ; 70(2): 83-88, 2024 Apr.
Article En | MEDLINE | ID: mdl-38711407

The toxic effect of ethanol on the cerebral cortex and protective effects of omega-3 fatty acids against this neurotoxicity were investigated. Twenty eight male Wistar-albino rats were divided into 4 groups. Rats of the ethanol and ethanol withdrawal groups were treated with ethanol (6 g/kg/day) for 15 days. Animals of the ethanol+omega-3 group received omega-3 fatty acids (400 mg/kg daily) and ethanol. In rats of the ethanol group SOD activity was lower than in animals of the control group. In rats treated with omega-3 fatty acids along with ethanol SOD, activity increased. GSH-Px activity and MDA levels in animals of all groups were similar. In ethanol treated rats NO levels significantly decreased as compared to the animals of the control group (6.45±0.24 nmol/g vs 11.05±0.53 nmol/g, p.


Cerebral Cortex , Ethanol , Fatty Acids, Omega-3 , Nitric Oxide , Rats, Wistar , Superoxide Dismutase , Animals , Male , Rats , Fatty Acids, Omega-3/pharmacology , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Nitric Oxide/metabolism , Superoxide Dismutase/metabolism , Glutathione Peroxidase/metabolism , Antioxidants/pharmacology , Malondialdehyde/metabolism
3.
Integr Cancer Ther ; 23: 15347354241243024, 2024.
Article En | MEDLINE | ID: mdl-38708673

Colorectal cancer (CRC) is the third leading cause of cancer-related death in the world. Multiple evidence suggests that there is an association between excess fat consumption and the risk of CRC. The long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are essential for human health, and both in vitro and in vivo studies have shown that these fatty acids can prevent CRC development through various molecular mechanisms. These include the modulation of arachidonic acid (AA) derived prostaglandin synthesis, alteration of growth signaling pathways, arrest of the cell cycle, induction of cell apoptosis, suppression of angiogenesis and modulation of inflammatory response. Human clinical studies found that LC n-3 PUFA combined with chemotherapeutic agents can improve the efficacy of treatment and reduce the dosage of chemotherapy and associated side effects. In this review, we discuss comprehensively the anti-cancer effects of LC n-3 PUFA on CRC, with a main focus on the underlying molecular mechanisms.


Colorectal Neoplasms , Fatty Acids, Omega-3 , Humans , Colorectal Neoplasms/drug therapy , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/administration & dosage , Animals , Apoptosis/drug effects , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/administration & dosage , Signal Transduction/drug effects , Docosahexaenoic Acids/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
4.
Int J Mol Sci ; 25(7)2024 Mar 28.
Article En | MEDLINE | ID: mdl-38612589

Lung cancer is the leading cause of cancer death worldwide. Polycyclic aromatic hydrocarbons (PAHs) are metabolized by the cytochrome P450 (CYP)1A and 1B1 to DNA-reactive metabolites, which could lead to mutations in critical genes, eventually resulting in cancer. Omega-3 fatty acids, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are beneficial against cancers. In this investigation, we elucidated the mechanisms by which omega-3 fatty acids EPA and DHA will attenuate PAH-DNA adducts and lung carcinogenesis and tumorigenesis mediated by the PAHs BP and MC. Adult wild-type (WT) (A/J) mice, Cyp1a1-null, Cyp1a2-null, or Cyp1b1-null mice were exposed to PAHs benzo[a]pyrene (BP) or 3-methylcholanthrene (MC), and the effects of omega-3 fatty acid on PAH-mediated lung carcinogenesis and tumorigenesis were studied. The major findings were as follows: (i) omega-3 fatty acids significantly decreased PAH-DNA adducts in the lungs of each of the genotypes studied; (ii) decreases in PAH-DNA adduct levels by EPA/DHA was in part due to inhibition of CYP1B1; (iii) inhibition of soluble epoxide hydrolase (sEH) enhanced the EPA/DHA-mediated prevention of pulmonary carcinogenesis; and (iv) EPA/DHA attenuated PAH-mediated carcinogenesis in part by epigenetic mechanisms. Taken together, our results suggest that omega-3 fatty acids have the potential to be developed as cancer chemo-preventive agents in people.


Fatty Acids, Omega-3 , Polycyclic Aromatic Hydrocarbons , Humans , Adult , Mice , Animals , Fatty Acids, Omega-3/pharmacology , DNA Adducts , Carcinogenesis , Cell Transformation, Neoplastic , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology
5.
Nutrients ; 16(7)2024 Mar 27.
Article En | MEDLINE | ID: mdl-38612996

Managing atherosclerotic cardiovascular disease (ASCVD) often involves a combination of lifestyle modifications and medications aiming to decrease the risk of cardiovascular outcomes, such as myocardial infarction and stroke. The aim of this article is to discuss possible omega-3 (n-3) fatty acid-statin interactions in the prevention and treatment of ASCVD and to provide evidence to consider for clinical practice, highlighting novel insights in this field. Statins and n-3 fatty acids (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) are commonly used to control cardiovascular risk factors in order to treat ASCVD. Statins are an important lipid-lowering therapy, primarily targeting low-density lipoprotein cholesterol (LDL-C) levels, while n-3 fatty acids address triglyceride (TG) concentrations. Both statins and n-3 fatty acids have pleiotropic actions which overlap, including improving endothelial function, modulation of inflammation, and stabilizing atherosclerotic plaques. Thus, both statins and n-3 fatty acids potentially mitigate the residual cardiovascular risk that remains beyond lipid lowering, such as persistent inflammation. EPA and DHA are both substrates for the synthesis of so-called specialized pro-resolving mediators (SPMs), a relatively recently recognized feature of their ability to combat inflammation. Interestingly, statins seem to have the ability to promote the production of some SPMs, suggesting a largely unrecognized interaction between statins and n-3 fatty acids with relevance to the control of inflammation. Although n-3 fatty acids are the major substrates for the production of SPMs, these signaling molecules may have additional therapeutic benefits beyond those provided by the precursor n-3 fatty acids themselves. In this article, we discuss the accumulating evidence that supports SPMs as a novel therapeutic tool and the possible statin-n-3 fatty acid interactions relevant to the prevention and treatment of ASCVD.


Atherosclerosis , Cardiovascular Diseases , Fatty Acids, Omega-3 , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Fatty Acids , Inflammation
6.
Nat Commun ; 15(1): 3068, 2024 Apr 09.
Article En | MEDLINE | ID: mdl-38594269

Polyunsaturated fatty acids (PUFAs), which cannot be synthesized by animals and must be supplied from the diet, have been strongly associated with human health. However, the mechanisms for their accretion remain poorly understood. Here, we show that LDL receptor-related protein 5 (LRP5), but not its homolog LRP6, selectively transports unesterified PUFAs into a number of cell types. The LDLa ligand-binding repeats of LRP5 directly bind to PUFAs and are required and sufficient for PUFA transport. In contrast to the known PUFA transporters Mfsd2a, CD36 and FATP2, LRP5 transports unesterified PUFAs via internalization to intracellular compartments including lysosomes, and n-3 PUFAs depend on this transport mechanism to inhibit mTORC1. This LRP5-mediated PUFA transport mechanism suppresses extracellular trap formation in neutrophils and protects mice from myocardial injury during ischemia-reperfusion. Thus, this study reveals a biologically important mechanism for unesterified PUFA transport to intracellular compartments.


Fatty Acids, Omega-3 , Fatty Acids, Unsaturated , Animals , Humans , Mice , Diet , Fatty Acids/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Unsaturated/metabolism , Receptors, LDL
7.
Expert Opin Drug Discov ; 19(5): 617-629, 2024 May.
Article En | MEDLINE | ID: mdl-38595031

INTRODUCTION: ω-3 Polyunsaturated fatty acids (PUFAs) have a range of health benefits, including anticancer activity, and are converted to lipid mediators that could be adapted into pharmacological strategies. However, the stability of these mediators must be improved, and they may require formulation to achieve optimal tissue concentrations. AREAS COVERED: Herein, the author reviews the literature around chemical stabilization and formulation of ω-3 PUFA mediators and their application in anticancer drug discovery. EXPERT OPINION: Aryl-urea bioisosteres of ω-3 PUFA epoxides that killed cancer cells targeted the mitochondrion by a novel dual mechanism: as protonophoric uncouplers and as inhibitors of electron transport complex III that activated ER-stress and disrupted mitochondrial integrity. In contrast, aryl-ureas that contain electron-donating substituents prevented cancer cell migration. Thus, aryl-ureas represent a novel class of agents with tunable anticancer properties. Stabilized analogues of other ω-3 PUFA-derived mediators could also be adapted into anticancer strategies. Indeed, a cocktail of agents that simultaneously promote cell killing, inhibit metastasis and angiogenesis, and that attenuate the pro-inflammatory microenvironment is a novel future anticancer strategy. Such regimen may enhance anticancer drug efficacy, minimize the development of anticancer drug resistance and enhance outcomes.


Antineoplastic Agents , Drug Discovery , Fatty Acids, Omega-3 , Neoplasms , Humans , Fatty Acids, Omega-3/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery/methods , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Mitochondria/drug effects , Mitochondria/metabolism
8.
Biomolecules ; 14(4)2024 Apr 06.
Article En | MEDLINE | ID: mdl-38672464

Krill oil is extracted from krill, a small crustacean in the Antarctic Ocean. It has received growing attention because of krill oil's unique properties and diverse health benefits. Recent experimental and clinical studies suggest that it has potential therapeutic benefits in preventing the development of a range of chronic conditions, including inflammatory bowel disease (IBD). Krill oil is enriched with long-chain n-3 polyunsaturated fatty acids, especially eicosapentaenoic and docosahexaenoic acids, and the potent antioxidant astaxanthin, contributing to its therapeutic properties. The possible underlying mechanisms of krill oil's health benefits include anti-inflammatory and antioxidant actions, maintaining intestinal barrier functions, and modulating gut microbiota. This review aims to provide an overview of the beneficial effects of krill oil and its bioactive components on intestinal inflammation and to discuss the findings on the molecular mechanisms associated with the role of krill oil in IBD prevention and treatment.


Euphausiacea , Inflammatory Bowel Diseases , Euphausiacea/chemistry , Animals , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Humans , Gastrointestinal Microbiome/drug effects , Oils/chemistry , Oils/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-3/chemistry
9.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 107-112, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38678615

Parkinson's disease (PD) is defined as a progressive neurodegenerative disease in middle-aged and elderly people. The therapeutic effect of ω-3 PUFAs in several neurodegenerative diseases has been well recognized. Nevertheless, whether nutrition supplementing ω-3 PUFAs exerts a neuroprotective role in PD remains elusive. Bioinformatics revealed 2D chemical structural formula of three components. Mice received indicated treatment with saline, MPTP or ω-3 PUFAs according to grouping. Behavioral function of mice was measured through motor tests such as rearing, akinesia, and rotarod tests. OFT test measured anxiety-like behaviors of mice. Western blotting and TUNEL staining measured dopaminergic fibers and neurons of mice. Western blotting measured inflammation and apoptosis-related protein levels in mouse tissue. FACS measured iTreg cell proportion in colon and brain tissues of mice. ω-3 PUFAs repaired MPTP-stimulated motor function damage in PD mice. ω-3 PUFAs mitigated MPTP-stimulated comorbid anxiety in PD mice. ω-3 PUFAs relieved MPTP-stimulated deficits of dopaminergic fibers and neurons in PD mice. ω-3 PUFAs repressed MPTP-stimulated inflammation and apoptosis pathway activation in PD mice. ω-3 PUFAs repaired MPTP-stimulated immune function damage in PD mice. ω-3 PUFAs exert a protective role in PD mice through alleviating motor function impairment and neuroinflammation by increasing intestinal inducible Treg cells, which may provide a new direction for seeking targeted therapy plans for PD in humans.


Disease Models, Animal , Fatty Acids, Omega-3 , Mice, Inbred C57BL , Parkinson Disease , T-Lymphocytes, Regulatory , Animals , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Intestines/drug effects , Intestines/pathology , Behavior, Animal/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Inflammation/pathology , Inflammation/drug therapy , Inflammation/metabolism
10.
Nutrients ; 16(8)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38674874

The present study aimed to investigate the differential effects of n-3 and n-6 polyunsaturated fatty acids (PUFAs) on placental and embryonic development. Pregnant mice were assigned to five groups: healthy control (HC), diabetes mellitus control (DMC), diabetes + low-dose n-3 PUFA (Ln-3), diabetes + high-dose n-3 PUFA (Hn-3), and diabetes + n-6 PUFA (n-6). On E12.5d, the Hn-3 group, but not the n-6 group, had a higher placenta weight. The weight ratio of embryo to placenta in the n-6 group was significantly lower than in the Hn-3 group but higher than in the DMC group. The Hn-3 group had significantly higher protein levels of VEGF, IGF-1, and IGFBP3, while the n-6 group had lower VEGF than the DMC group. Compared with the DMC group, embryonic Cer-16:0 was significantly higher in the Hn-3 group, while embryonic PC (36:6), PC (38:7), and PE (40:7) were significantly lower in the n-6 group. The embryo and placenta weights were positively correlated with placental VEGF, IGFBP3, and embryonic Cer-16:0, and they were negatively correlated with embryonic PC (36:6) and PE (40:7). The weight ratio of embryo to placenta was negatively correlated with embryonic PC (36:6). In addition, embryonic Cer-16:0 was positively correlated with placental VEGF and IGFBP3. In conclusion, n-3 PUFA and n-6 PUFA improved placental and embryonic growth through different mechanisms.


Embryonic Development , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Placenta , Animals , Pregnancy , Female , Fatty Acids, Omega-3/pharmacology , Placenta/metabolism , Placenta/drug effects , Fatty Acids, Omega-6/pharmacology , Mice , Embryonic Development/drug effects , Diabetes Mellitus, Experimental , Insulin-Like Growth Factor Binding Protein 3/metabolism , Vascular Endothelial Growth Factor A/metabolism , Pregnancy in Diabetics/metabolism , Insulin-Like Growth Factor I/metabolism , Organ Size/drug effects
11.
Nutrition ; 123: 112413, 2024 Jul.
Article En | MEDLINE | ID: mdl-38518540

OBJECTIVES: We assessed the joint effects of omega (n)-3 fatty acid supplementation and dietary fish intake on systemic lipid mediators of inflammation among adults. METHODS: Within VITAL, a double-blind randomized controlled trial, adults were randomized to ω-3 fatty acids (460 mg EPA + 380 mg DHA/d) or placebo. We selected participants who reported low (<1 serving/mo) baseline dietary fish intake and matched them by age, sex, race, and trial arm to participants with self-reported highest fish intake (≥3.9 servings/wk). Baseline and 1-y plasma samples were tested for 9 ω-3 fatty acid-derived lipid mediators. Multivariable linear models assessed lipid mediator changes and joint effects of ω-3 fatty acid supplementation and dietary fish intake. RESULTS: Forty-eight participants with low baseline fish intake were matched to 48 with high fish intake. Mean age was 64.6 (±7.26), 50% were female, and 85% non-Hispanic white. One-year lipid mediator changes in expected directions were observed in those receiving ω-3 fatty acids versus placebo: reductions in proinflammatory mediators, PGD2, 5-HETE, and 12-HETE; increases in proresolving mediators, EPA and DHA. Larger 1-y lipid biomarker changes were seen in those with low baseline fish intake randomized to active ω-3 fatty acids for DHA, EPA, PGD2, Resolvin D1, and Resolvin D4 were observed, although no significant multiplicative interactions were detected. DISCUSSION: Beneficial changes in circulating proresolving and proinflammatory mediators were found with 1-y of ω-3 fatty acid supplementation versus placebo for all participants, with a trend toward larger effects among those with low baseline fish intake, although interactions were not significant.


Dietary Supplements , Docosahexaenoic Acids , Fatty Acids, Omega-3 , Fishes , Inflammation , Seafood , Humans , Female , Male , Middle Aged , Double-Blind Method , Inflammation/blood , Animals , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Aged , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/blood , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/administration & dosage , Diet/methods
12.
Am J Clin Nutr ; 119(5): 1175-1186, 2024 May.
Article En | MEDLINE | ID: mdl-38484976

BACKGROUND: Extracellular vesicles (EVs) are proposed to play a role in the development of cardiovascular diseases (CVDs) and are considered emerging markers of CVDs. n-3 PUFAs are abundant in oily fish and fish oil and are reported to reduce CVD risk, but there has been little research to date examining the effects of n-3 PUFAs on the generation and function of EVs. OBJECTIVES: We aimed to investigate the effects of fish oil supplementation on the number, generation, and function of EVs in subjects with moderate risk of CVDs. METHODS: A total of 40 participants with moderate risk of CVDs were supplemented with capsules containing either fish oil (1.9 g/d n-3 PUFAs) or control oil (high-oleic safflower oil) for 12 wk in a randomized, double-blind, placebo-controlled crossover intervention study. The effects of fish oil supplementation on conventional CVD and thrombogenic risk markers were measured, along with the number and fatty acid composition of circulating and platelet-derived EVs (PDEVs). PDEV proteome profiles were evaluated, and their impact on coagulation was assessed using assays including fibrin clot formation, thrombin generation, fibrinolysis, and ex vivo thrombus formation. RESULTS: n-3 PUFAs decreased the numbers of circulating EVs by 27%, doubled their n-3 PUFA content, and reduced their capacity to support thrombin generation by >20% in subjects at moderate risk of CVDs. EVs derived from n-3 PUFA-enriched platelets in vitro also resulted in lower thrombin generation, but did not alter thrombus formation in a whole blood ex vivo assay. CONCLUSIONS: Dietary n-3 PUFAs alter the number, composition, and function of EVs, reducing their coagulatory activity. This study provides clear evidence that EVs support thrombin generation and that this EV-dependent thrombin generation is reduced by n-3 PUFAs, which has implications for prevention and treatment of thrombosis. CLINICAL TRIAL REGISTRY: This trial was registered at clinicaltrials.gov as NCT03203512.


Blood Coagulation , Blood Platelets , Cross-Over Studies , Extracellular Vesicles , Fatty Acids, Omega-3 , Humans , Extracellular Vesicles/metabolism , Fatty Acids, Omega-3/pharmacology , Male , Female , Middle Aged , Double-Blind Method , Blood Coagulation/drug effects , Blood Platelets/metabolism , Blood Platelets/drug effects , Dietary Supplements , Cardiovascular Diseases/prevention & control , Adult , Fish Oils/pharmacology , Fish Oils/administration & dosage , Aged , Fatty Acids/metabolism
13.
Psychogeriatrics ; 24(3): 701-718, 2024 May.
Article En | MEDLINE | ID: mdl-38528391

Curcumin and omega-3 polyunsaturated fatty acids (ω-3 PUFA) are multifunctional compounds which play an important role in Alzheimer's disease (AD) and little has been addressed about the role of these two compounds together in the progression of the disease. There is evidence of the beneficial effect of combined administration of ω-3 PUFA and other dietary supplements such as vitamins and polyphenols in the prevention of AD, although much remains to be understood about their possible complementary or synergistic activity. Therefore, the objective of this work is to review the research focused on studying the effect and mechanisms of action of curcumin, ω-3 PUFA, and the combination of these nutraceutical compounds, particularly on AD, and to integrate the possible ways in which these compounds can potentiate their effect. The most important pathophysiologies that manifest in AD will be addressed, in order to have a better understanding of the mechanisms of action through which these bioactive compounds exert a neuroprotective effect.


Alzheimer Disease , Curcumin , Dietary Supplements , Fatty Acids, Omega-3 , Neuroprotective Agents , Curcumin/therapeutic use , Curcumin/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Drug Synergism
14.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article En | MEDLINE | ID: mdl-38542331

Colorectal cancer (CRC) is a major life-threatening disease, being the third most common cancer and a leading cause of death worldwide. Enhanced adiposity, particularly visceral fat, is a major risk factor for CRC, and obesity-associated alterations in metabolic, inflammatory and immune profiles in visceral adipose tissue (VAT) strongly contribute to promoting or sustaining intestinal carcinogenesis. The role of diet and nutrition in obesity and CRC has been extensively demonstrated, and AT represents the main place where diet-induced signals are integrated. Among the factors introduced with diet and processed or enriched in AT, ω3/ω6 polyunsaturated fatty acids (PUFAs) are endowed with pro- or anti-inflammatory properties and have been shown to exert either promoting or protective roles in CRC. In this study, we investigated the impact of ex vivo exposure to the ω3 and ω6 PUFAs docosahexaenoic and arachidonic acids on VAT adipocyte whole transcription in healthy lean, obese and CRC-affected individuals. High-throughput sequencing of protein-coding and long non-coding RNAs allowed us to identify specific pathways and regulatory circuits controlled by PUFAs and highlighted an impaired responsiveness of obese and CRC-affected individuals as compared to the strong response observed in healthy lean subjects. This further supports the role of healthy diets and balanced ω3/ω6 PUFA intake in the primary prevention of obesity and cancer.


Colorectal Neoplasms , Fatty Acids, Omega-3 , RNA, Long Noncoding , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Unsaturated , Adipocytes/metabolism , Obesity/genetics , Obesity/metabolism , Colorectal Neoplasms/genetics
15.
Org Biomol Chem ; 22(14): 2877-2890, 2024 04 03.
Article En | MEDLINE | ID: mdl-38525805

Oxidative stress and carbonyl stress resulting from the toxicity of small aldehydes are part of the detrimental mechanisms leading to neuronal cell loss involved in the progression of neurodegenerative diseases such as Alzheimer's disease. Polyunsaturated alkylated lipophenols represent a new class of hybrid molecules that combine the health benefits of anti-inflammatory omega-3 fatty acids with the anti-carbonyl and oxidative stress (anti-COS) properties of (poly)phenols in a single pharmacological entity. To investigate the therapeutic potential of quercetin-3-docosahexaenoic acid-7-isopropyl lipophenol in neurodegenerative diseases, three synthetic pathways using chemical or chemo-enzymatic strategies were developed to access milligram or gram scale quantities of this alkyl lipophenol. The protective effect of quercetin-3-DHA-7-iPr against cytotoxic concentrations of acrolein (a carbonyl stressor) was assessed in human SHSY-5Y neuroblastoma cells to underscore its ability to alleviate harmful mechanisms associated with carbonyl stress in the context of neurodegenerative diseases.


Fatty Acids, Omega-3 , Neurodegenerative Diseases , Humans , Quercetin/pharmacology , Oxidative Stress , Fatty Acids, Omega-3/pharmacology , Docosahexaenoic Acids/pharmacology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism
16.
Methods Mol Biol ; 2761: 209-229, 2024.
Article En | MEDLINE | ID: mdl-38427239

Omega-3 fatty acids play a seminal role in maintaining the structural and functional integrity of the nervous system. These specialized molecules function as precursors for many lipid-based biological messengers. Also, studies suggest the role of these fatty acids in regulating healthy sleep cycles, cognitive ability, brain development, etc. Dietary intake of essential poly unsaturated fatty acids (PUFA) such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are foundational to the optimal working of the nervous system. Besides regulating health, these biomolecules have great therapeutic value in treating several diseases, particularly nervous system diseases and disorders. Many recent studies conclusively demonstrated the beneficial effects of Omega-3 fatty acids in treating depression, neuropsychiatric disorders, neurodegenerative disorders, neurochemical disorders, and many other illnesses associated with the nervous system. This chapter summates the multifaceted role of poly unsaturated fatty acids, especially Omega-3 fatty acids (EPA and DHA), in the neuronal health and functioning. The importance of dietary intake of these essential fatty acids, their recommended dosages, bioavailability, the mechanism of their action, and therapeutic values are extensively discussed.


Fatty Acids, Omega-3 , Fatty Acids, Omega-3/pharmacology , Eicosapentaenoic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Fatty Acids, Unsaturated , Fatty Acids , Brain
17.
Nutrients ; 16(5)2024 Feb 24.
Article En | MEDLINE | ID: mdl-38474762

INTRODUCTION: chronic low-grade inflammation, or inflammaging, emerges as a crucial element in the aging process and is associated with cardiovascular and neurological diseases, sarcopenia, and malnutrition. Evidence suggests that omega-3 fatty acids present a potential therapeutic agent in the prevention and treatment of inflammatory diseases, mitigating oxidative stress, and improving muscle mass, attributes that are particularly relevant in the context of aging. The objective of the present study was to evaluate the effectiveness of supplementation with omega-3 fish oil in improving the immune response and oxidative stress in knockout mice for interleukin IL-10 (IL-10-/-). MATERIAL AND METHODS: female C57BL/6 wild-type (WT) and interleukin IL-10 knockout (IL-10-/-) mice were fed during 90 days with a standard diet (control groups), or they were fed/supplemented with 10% of the omega-3 polyunsaturated fatty acid diet (omega-3 groups). Muscle, liver, intestinal, and mesenteric lymph node tissue were collected for analysis. RESULTS: the IL-10-/-+O3 group showed greater weight gain compared to the WT+O3 (p = 0.001) group. The IL-10-/-+O3 group exhibited a higher frequency of regulatory T cells than the IL-10-/- group (p = 0.001). It was found that animals in the IL-10-/-+O3 group had lower levels of steatosis when compared to the IL-10-/- group (p = 0.017). There was even greater vitamin E activity in the WT group compared to the IL-10-/-+O3 group (p = 0.001) and WT+O3 compared to IL-10-/-+O3 (p = 0.002), and when analyzing the marker of oxidative stress, MDA, an increase in lipid peroxidation was found in the IL-10-/-+O3 group when compared to the IL-10-/- group (p = 0.03). Muscle tissue histology showed decreased muscle fibers in the IL-10-/-+O3, IL-10-/-, and WT+O3 groups. CONCLUSION: the findings show a decrease in inflammation, an increase in oxidative stress markers, and a decrease in antioxidant markers in the IL-10-/-+O3 group, suggesting that supplementation with omega-3 fish oil might be a potential intervention for inflammaging that characterizes the aging process and age-related diseases.


Fatty Acids, Omega-3 , Female , Mice , Animals , Fatty Acids, Omega-3/pharmacology , Antioxidants/pharmacology , T-Lymphocytes, Regulatory/metabolism , Mice, Knockout , Interleukin-10/metabolism , Mice, Inbred C57BL , Fish Oils/pharmacology , Oxidative Stress , Dietary Supplements , Liver/metabolism , Inflammation/metabolism
18.
J Nutr Biochem ; 127: 109603, 2024 May.
Article En | MEDLINE | ID: mdl-38373507

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.


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
19.
Neurosci Lett ; 824: 137688, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38360146

This research was done to examine the combination of citalopram, an antidepressant drug, and omega-3 in a mice model of depression. Mice received citalopram (1 and 2 mg/kg) or omega-3 (10 and 20 mg/kg) daily over 30 days. Then, they were exposed to acute and chronic restraint stress to assess the possible increasing effect of omega-3 on the antidepressant and anxiolytic effects of citalopram. Elevated plus-maze (EPM) and forced swimming test (FST) were used to assess anxiety and depression symptoms in non-restraint stress (NRS), acute restraint stress (ARS), and chronic restraint stress (CRS) mice. The results indicated that induction of acute and chronic restraint stress reduced %OAT (Open arm time) and %OAE (Open arm entrance) in the EPM test but enhanced immobility time in the FST, showing anxiogenic- and depressive-like effects. These stresses reduced the stability of pyramidal neurons in the prefrontal cortex (PFC) and hippocampus. Aone and combination administration with citalopram and omega-3 induced anxiolytic- and antidepressant-like effects in NRS, ARS, and CRS mice. This combination usage increased the stability of pyramidal neurons in the PFC and hippocampus. These results suggested an interaction between citalopram and omega-3 upon the induction of anxiolytic- and antidepressant-like effects as well as augmentation of the ratio of pyramidal live to dark neurons in the PFC and hippocampus of the ARS and CRS mice.


Anti-Anxiety Agents , Fatty Acids, Omega-3 , Mice , Animals , Citalopram/pharmacology , Anti-Anxiety Agents/pharmacology , Depression/drug therapy , Fatty Acids, Omega-3/pharmacology , Behavior, Animal , Anxiety/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain , Pyramidal Cells
20.
Mar Drugs ; 22(2)2024 Feb 06.
Article En | MEDLINE | ID: mdl-38393051

The products of oleaginous microbes, primarily lipids, have gained tremendous attention for their health benefits in food-based applications as supplements. However, this emerging biotechnology also offers a neuroprotective treatment/management potential for various diseases that are seldom discussed. Essential fatty acids, such as DHA, are known to make up the majority of brain phospholipid membranes and are integral to cognitive function, which forms an important defense against Alzheimer's disease. Omega-3 polyunsaturated fatty acids have also been shown to reduce recurrent epilepsy seizures and have been used in brain cancer therapies. The ratio of omega-3 to omega-6 PUFAs is essential in maintaining physiological function. Furthermore, lipids have also been employed as an effective vehicle to deliver drugs for the treatment of diseases. Lipid nanoparticle technology, used in pharmaceuticals and cosmeceuticals, has recently emerged as a biocompatible, biodegradable, low-toxicity, and high-stability means for drug delivery to address the drawbacks associated with traditional medicine delivery methods. This review aims to highlight the dual benefit that lipids offer in maintaining good health for disease prevention and in the treatment of neurological diseases.


Epilepsy , Fatty Acids, Omega-3 , Humans , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Dietary Supplements , Brain , Phospholipids/therapeutic use , Epilepsy/drug therapy
...