Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nutrients ; 15(14)2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37513526

ABSTRACT

Rationale and Methods: Skeletal muscle derangements, potentially including mitochondrial dysfunction with altered mitochondrial dynamics and high reactive oxygen species (ROS) generation, may lead to protein catabolism and muscle wasting, resulting in low exercise capacity and reduced survival in chronic heart failure (CHF). We hypothesized that 8-week n-3-PUFA isocaloric partial dietary replacement (Fat = 5.5% total cal; EPA + DHA = 27% total fat) normalizes gastrocnemius muscle (GM) mitochondrial dynamics regulators, mitochondrial and tissue pro-oxidative changes, and catabolic derangements, resulting in preserved GM mass in rodent CHF [Myocardial infarction (MI)-induced CHF by coronary artery ligation, left-ventricular ejection fraction <50%]. Results: Compared to control animals (Sham), CHF had a higher GM mitochondrial fission-fusion protein ratio, with low ATP and high ROS production, pro-inflammatory changes, and low insulin signalling. n-3-PUFA normalized all mitochondrial derangements and the pro-oxidative state (oxidized to total glutathione ratio), associated with normalized GM cytokine profile, and enhanced muscle-anabolic insulin signalling and prevention of CHF-induced GM weight loss (all p < 0.05 vs. CHF and p = NS vs. S). Conclusions:n-3-PUFA isocaloric partial dietary replacement for 8 weeks normalizes CHF-induced derangements of muscle mitochondrial dynamics regulators, ROS production and function. n-3-PUFA mitochondrial effects result in preserved skeletal muscle mass, with potential to improve major patient outcomes in clinical settings.


Subject(s)
Fatty Acids, Omega-3 , Heart Failure , Insulins , Mice , Animals , Fatty Acids, Omega-3/metabolism , Reactive Oxygen Species/metabolism , Stroke Volume , Ventricular Function, Left , Muscle, Skeletal/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Heart Failure/drug therapy , Diet , Insulins/metabolism
2.
Metabolism ; 133: 155242, 2022 08.
Article in English | MEDLINE | ID: mdl-35750236

ABSTRACT

INTRODUCTION AND METHODS: Skeletal muscle mitochondrial dysfunction may cause tissue oxidative stress and consequent catabolism in chronic kidney disease (CKD), contributing to patient mortality. We investigated in 5/6-nephrectomized (Nx) rats the impact of n3-polyunsaturated fatty-acids (n3-PUFA) isocaloric partial dietary replacement on gastrocnemius muscle (Gm) mitochondrial master-regulators, ATP production, ROS generation and related muscle-catabolic derangements. RESULTS: Nx had low Gm mitochondrial nuclear respiratory factor-2 and peroxisome proliferator-activated receptor gamma coactivator-1alpha, low ATP production and higher mitochondrial fission-fusion protein ratio with ROS overproduction. n3-PUFA normalized all mitochondrial derangements and pro-oxidative tissue redox state (oxydized to total glutathione ratio). n3-PUFA also normalized Nx-induced muscle-catabolic proinflammatory cytokines, insulin resistance and low muscle weight. Human uremic serum reproduced mitochondrial derangements in C2C12 myotubes, while n3-PUFA coincubation prevented all effects. n3-PUFA also enhanced muscle mitophagy in-vivo and siRNA-mediated autophagy inhibition selectively blocked n3-PUFA-induced normalization of C2C12 mitochondrial ROS production. CONCLUSIONS: In conclusion, dietary n3-PUFA normalize mitochondrial master-regulators, ATP production and dynamics in experimental CKD. These effects occur directly in muscle cells and they normalize ROS production through enhanced mitophagy. Dietary n3-PUFA mitochondrial effects result in normalized catabolic derangements and protection from muscle wasting, with potential positive impact on patient survival.


Subject(s)
Fatty Acids, Omega-3 , Renal Insufficiency, Chronic , Adenosine Triphosphate/metabolism , Animals , Dietary Fats/pharmacology , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Mitochondria/metabolism , Mitophagy , Muscle, Skeletal/metabolism , Muscular Atrophy , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/metabolism
3.
Nutrients ; 12(7)2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32708527

ABSTRACT

Deterioration of muscle strength during cancer results in functional limitation, poor quality of life and reduced survival. The indirect effects on muscle strength of nutritional interventions based on protein and amino acid derivatives targeted at improving muscle mass are poorly documented. A scoping review was performed to examine the available evidence on the effects of proteins, amino acids and their derivatives on muscle strength in adult cancer patients. Pubmed and Scopus databases were searched to identify research articles published in the last 10 years. Fourteen studies met the inclusion criteria, showing that changes in muscle strength following protein or amino acid supplementation are generally concordant with those in muscle mass in cancer patients. Administration of both energy and proteins in the presence of reduced oral intakes results in more robust effects on both muscle strength and mass. It is not clear whether this is due to the correction of the energy deficit or to an interaction between proteins and other macronutrients. The optimal mixture, type, and dose of amino acid/protein supplementation alone or in combination with other anabolic strategies should be determined to provide the best nutritional approach in cancer.


Subject(s)
Amino Acids/administration & dosage , Diet, High-Protein , Dietary Proteins/administration & dosage , Dietary Supplements , Muscle Strength , Muscle, Skeletal/metabolism , Neoplasms/complications , Nutritional Physiological Phenomena/physiology , Sarcopenia/etiology , Sarcopenia/therapy , Exercise Therapy , Female , Humans , Male , Muscle, Skeletal/physiopathology , Quality of Life , Sarcopenia/physiopathology
4.
Int J Mol Sci ; 19(1)2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29324650

ABSTRACT

Elderly and patients affected by chronic diseases face a high risk of muscle loss and impaired physical function. Omega 3 fatty acids (FA) attenuate inflammation and age-associated muscle loss, prevent systemic insulin resistance and improve plasma lipids, potentially impacting on sarcopenia. This paper aims to review recent randomized clinical studies assessing the effects a chronic omega 3 FA supplementation on inflammatory and metabolic profile during conditions characterized by sarcopenia (aging, insulin resistance, type 2 diabetes, chronic renal failure). A comprehensive search of three online databases was performed to identify eligible trials published between 2012 and 2017. A total of 36 studies met inclusion criteria. Omega 3 FA yielded mixed results on plasma triglycerides in the elderly and no effects in renal patients. No changes in systemic insulin resistance were observed. Inflammation markers did not benefit from omega 3 FA in insulin resistant and in renal subjects while decreasing in obese and elderly. Muscle related parameters improved in elderly and in renal patients. In conclusion, in aging- and in chronic disease-associated sarcopenia omega 3 FA are promising independently of associated anabolic stimuli or of anti-inflammatory effects. The evidence for improved glucose metabolism in insulin resistant and in chronic inflammatory states is less solid.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Insulin Resistance , Sarcopenia/drug therapy , Anti-Inflammatory Agents/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Humans , Lipid Metabolism , Oxidative Stress , Randomized Controlled Trials as Topic , Sarcopenia/metabolism
5.
Nutrients ; 9(8)2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28820443

ABSTRACT

BACKGROUND: Endothelial dysfunction is a key vascular alteration in chronic kidney disease (CKD). Omega 3 (n-3) polyunsaturated fatty acids (PUFA) reduce vascular oxidative stress and inflammation. We investigated whether n-3 PUFA could reverse endothelial dysfunction in CKD by improving endothelial nitric oxide synthase (eNOS) function and oxidative stress. METHODS: 5/6 nephrectomized male Wistar rats (CKD; n = 10) and sham operated animals (SHAM; n = 10) were treated for 6 weeks with standard diet. An additional group of CKD rats were fed an n-3 PUFA enriched diet (CKD + PUFA; n = 10). We then measured endothelium-dependent (EDD) and -independent vasodilation, markers of endothelial function and of oxidative stress in thoracic aortas. RESULTS: Compared to SHAM, in CKD aortas EDD and eNOS expression were reduced (p < 0.05) and 3-nitrotyrosine levels were increased, while expression of NADPH oxidase subunits NOX4 and p22phox was similar. In-vitro incubation with Tiron failed to reverse endothelial dysfunction in CKD. In CKD + PUFA, EDD improved (p < 0.05) compared with CKD rats, while blockade of eNOS by L-NAME worsened EDD. These effects were accompanied by increased (p < 0.05) eNOS and reduced (p < 0.05) expression of NOX4 and 3-nitrotyrosine levels. CONCLUSION: Collectively, these findings indicate that n-3 PUFA improve endothelial dysfunction by restoring NO bioavailability in CKD.


Subject(s)
Endothelium, Vascular/physiopathology , Fatty Acids, Omega-3/administration & dosage , Kidney Failure, Chronic/drug therapy , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Biomarkers/blood , Male , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide/blood , Nitric Oxide Synthase Type III/genetics , Rats , Rats, Wistar , Tyrosine/analogs & derivatives , Tyrosine/blood
6.
J Cardiovasc Pharmacol Ther ; 19(1): 114-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24038017

ABSTRACT

BACKGROUND: Menopause is associated with increased arterial stiffness, an independent marker of cardiovascular risk. Omega-3 polyunsaturated fatty acids (N3-PUFAs) are thought to have multiple cardiovascular benefits, including prevention of arterial stiffness. We investigated whether treatment with N3-PUFA prevents increase in arterial stiffness in ovariectomized rats, an animal model of experimental menopause. METHODS: A total of 43 Wistar rats, 2 months old, were divided into 3 groups, control, sham surgery, normal diet (CTRL, n = 15); ovariectomy, normal diet (OVX, n = 14); and ovariectomy with N3-PUFA supplementation (0.8 g/kg/d in daily gavages administration; OVX + O3, n = 14). Two months after surgery, carotid-femoral pulse wave velocity (PWV) and arterial blood pressure (BP) were measured by carotid and femoral cannulation. Aortic morphometric measurements were performed after dissection. RESULTS: Ovariectomy caused a significant increase in BP (P < .05), PWV (P < .0001), and elastic modulus (P = .001) compared to CTRL. After ovariectomy, N3-PUFA supplementation completely prevented increase in arterial stiffness (P < .0001 vs OVX) and BP (P < .05 vs OVX) and resulted in a significant increase in body weight and aortic thickness. CONCLUSIONS: In an experimental model of menopause, N3-PUFA supplementation prevents arterial stiffening and other vascular changes induced by ovariectomy. These results represent a therapeutic benefit of N3-PUFAs in prevention of postmenopausal cardiovascular disease.


Subject(s)
Cardiovascular Diseases/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Menopause , Animals , Aorta/drug effects , Aorta/metabolism , Arterial Pressure/drug effects , Body Weight , Cardiovascular Diseases/etiology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Disease Models, Animal , Fatty Acids, Omega-3/administration & dosage , Female , Femoral Artery/drug effects , Femoral Artery/pathology , Ovariectomy , Pulse Wave Analysis , Rats , Rats, Inbred WKY , Risk Factors , Vascular Stiffness/drug effects
7.
J Nutr Biochem ; 24(1): 371-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23159066

ABSTRACT

Menopause is associated with endothelial dysfunction and oxidative stress. In this condition, reduced n-3 polyunsaturated fatty acids (n-3 PUFAs) contribute to cardiovascular disease. We investigated whether treatment with n-3 PUFA reverses endothelial dysfunction and oxidative stress in experimental menopause. Thirty female rats underwent either sham-surgery or bilateral ovariectomy or bilateral ovariectomy+oral n-3 PUFA (0.8 g kg(-1) day(-1) for 2 months). Ovariectomy caused endothelial dysfunction to acetylcholine, which was reversed by superoxide scavenger Tiron. Erythrocyte membrane lipid composition was characterized by reduced n-3 PUFA total content and omega-3 index, and by concomitant increase in n-6:n-3 PUFA ratio. Ovariectomy-related oxidative stress, demonstrated by both enhanced superoxide production and 3-nitrotyrosine expression in aorta, was associated with increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit NOX-4 protein expression. Endothelial nitric oxide synthase (eNOS) functional inhibition by l-NG-nitroarginine methyl ester, protein expression and activity did not change. In ovariectomized rats, treatment with n-3 PUFA increased n-3 PUFA total content and omega-3 index and decreased n-6:n-3 PUFA ratio in erythrocyte membrane, reversed vascular oxidative stress, endothelial dysfunction, aortic 3-nitrotyrosine and markedly lowered NOX-4 protein expression; eNOS protein expression also increased, paralleled by reversal of inhibitory binding to Caveolin-1, while ex-vivo functional inhibition and NOS synthesis were unchanged. These findings demonstrate in vivo a therapeutic benefit of n-3 PUFA on menopause-associated endothelial dysfunction by reversal of alterations in membrane lipid composition induced by ovariectomy and by reduction of vascular oxidative stress. In this setting they also identify NOX-4 as a potential target to reduce oxidative stress-mediated vascular complications.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Fatty Acids, Omega-3/pharmacology , Menopause/drug effects , Oxidative Stress/drug effects , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Biomarkers/blood , Body Weight/drug effects , Caveolin 1/metabolism , Disease Models, Animal , Female , Free Radical Scavengers/pharmacology , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Menopause/physiology , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Ovariectomy , Rats , Rats, Wistar , Superoxides/metabolism , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL