Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cardiovasc Res ; 119(18): 2884-2901, 2024 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-38252923

RESUMO

Omega-3 fatty acids (O3FAs) possess beneficial properties for cardiovascular (CV) health and elevated O3FA levels are associated with lower incident risk for CV disease (CVD.) Yet, treatment of at-risk patients with various O3FA formulations has produced disparate results in large, well-controlled and well-conducted clinical trials. Prescription formulations and fish oil supplements containing low-dose mixtures of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have routinely failed to prevent CV events in primary and secondary prevention settings when added to contemporary care, as shown most recently in the STRENGTH and OMEMI trials. However, as observed in JELIS, REDUCE-IT, and RESPECT-EPA, EPA-only formulations significantly reduce CVD events in high-risk patients. The CV mechanism of action of EPA, while certainly multifaceted, does not depend solely on reductions of circulating lipids, including triglycerides (TG) and LDL, and event reduction appears related to achieved EPA levels suggesting that the particular chemical and biological properties of EPA, as compared to DHA and other O3FAs, may contribute to its distinct clinical efficacy. In vitro and in vivo studies have shown different effects of EPA compared with DHA alone or EPA/DHA combination treatments, on atherosclerotic plaque morphology, LDL and membrane oxidation, cholesterol distribution, membrane lipid dynamics, glucose homeostasis, endothelial function, and downstream lipid metabolite function. These findings indicate that prescription-grade, EPA-only formulations provide greater benefit than other O3FAs formulations tested. This review summarizes the clinical findings associated with various O3FA formulations, their efficacy in treating CV disease, and their underlying mechanisms of action.


Assuntos
Doenças Cardiovasculares , Ácidos Graxos Ômega-3 , Humanos , Ácidos Graxos Ômega-3/efeitos adversos , Ácido Eicosapentaenoico/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Colesterol , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/prevenção & controle , Doenças Cardiovasculares/tratamento farmacológico
2.
Nutr Metab Cardiovasc Dis ; 33(1): 11-21, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36319578

RESUMO

BACKGROUND: Evidence from clinical trial studies suggests that docosahexaenoic acids (DHA) may have greater potential effects on improving cardiovascular risk factors than eicosapentaenoic acid (EPA). However, this evidence has not yet been meta-analyzed and quantified. The aim of this study was to evaluate and compare the effect of DHA and EPA monotherapy on cardiovascular risk factors based on paired and network meta-analysis. METHODS: Relevant articles published up to January 2022 were systematically retrieved from relevant databases. We included all Randomized Controlled Trials (RCTs) on adults that directly compared the effects of DHA with EPA and RCTs of indirect comparisons (DHA and EPA monotherapy compared to control groups). Data were pooled by pairwise and network meta-analysis and expressed as mean differences (MDs) with 95% CIs. The study protocol was registered with PROSPERO (Registration ID: CRD42022328630). RESULTS: Network meta-analysis of comparisons of DHA and EPA suggested significant comparable effects only on LDL-C (MD EPA versus DHA = -8.51 mg/L; 95% CI: -16.67; -0.35). However, the Network meta-analysis not show a significant effect for other risk factors. Furthermore, pairwise meta-analysis of direct comparisons of DHA and EPA showed significant difference in their effects on plasma glucose (MD EPA versus DHA = -0.31 mg/L; 95% CI: -0.60, -0.02), Insulin (MD EPA versus DHA = -2.14 mg/L; 95% CI: -3.26, -1.02), but the results were not significant for risk factors. CONCLUSION: Our findings suggest that both EPA and DHA act similarly on the markers under study, with slight changes in plasma glucose, insulin, and LDL-C.


Assuntos
Ácido Eicosapentaenoico , Insulinas , Adulto , Humanos , Ácido Eicosapentaenoico/efeitos adversos , Metanálise em Rede , LDL-Colesterol , Glicemia , Ensaios Clínicos Controlados Aleatórios como Assunto , Ácidos Docosa-Hexaenoicos/efeitos adversos , Suplementos Nutricionais
3.
Regul Toxicol Pharmacol ; 127: 105050, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34610366

RESUMO

Omega-3 (ω3) fatty acids are a family of polyunsaturated fats. Two of the ω3 long-chain polyunsaturated fatty acids (LC-PUFA), eicosapentaenoic acid (EPA, ω3, 20:5Δ5,8,11,14,17) and docosahexaenoic acid (DHA, ω3, 22:6Δ4,7,10,13,16,19) are sourced primarily from fish. Higher consumption, limited fishing quotas and other environmental factors (e.g., heavy metals) have warranted a need for alternative sources. Nuseed offers a genetically engineered canola (Brassica napus) event,1 DHA canola (OECD Unique Identifier NS-B5ØØ27-4), which has been modified to introduce a pathway for production of the ω3 LC-PUFAs DHA and EPA from oleic acid (OA) in the seed oil. To accomplish this, genes were sourced from marine microalgae and common yeast then incorporated into canola to produce DHA canola, one of the first land-based production systems for ω 3 PUFAs. Safety was evaluated in part by conducting a repeated dose 28-day toxicity study and a dietary 13-week toxicity study using CD® IGS [Crl:CD(SD)] rats. In the 28-day study, conventional and DHA canola oil were administered orally (via gavage); no treatment-related adverse effects were observed. The 13-week toxicity study was subsequently conducted where DHA canola oil and meal were administered by dietary admixture. No adverse effects were noted in clinical observations, clinical pathology, or histopathology. These studies support the food and feed safety of DHA canola oil and meal.


Assuntos
Ácidos Docosa-Hexaenoicos/administração & dosagem , Óleo de Brassica napus/administração & dosagem , Animais , Peso Corporal , Brassica napus , Ácidos Docosa-Hexaenoicos/efeitos adversos , Feminino , Masculino , Microalgas/genética , Distribuição Aleatória , Óleo de Brassica napus/efeitos adversos , Ratos
4.
JAMA Netw Open ; 4(10): e2128771, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34648010

RESUMO

Importance: Supplementing preterm infants with long-chain polyunsaturated fatty acids (LC-PUFA) has been inconsistent in reducing the severity and incidence of retinopathy of prematurity (ROP). Furthermore, few studies have measured the long-term serum lipid levels after supplementation. Objective: To assess whether ROP severity is associated with serum levels of LC-PUFA, especially docosahexaenoic acid (DHA) and arachidonic acid (AA), during the first 28 postnatal days. Design, Setting, and Participants: This cohort study analyzed the Mega Donna Mega study, a randomized clinical trial that provided enteral fatty acid supplementation at 3 neonatal intensive care units in Sweden. Infants included in this cohort study were born at a gestational age of less than 28 weeks between December 20, 2016, and August 6, 2019. Main Outcomes and Measures: Severity of ROP was classified as no ROP, mild or moderate ROP (stage 1-2), or severe ROP (stage 3 and type 1). Serum phospholipid fatty acids were measured through gas chromatography-mass spectrometry. Ordinal logistic regression, with a description of unadjusted odds ratio (OR) as well as gestational age- and birth weight-adjusted ORs and 95% CIs, was used. Areas under the curve were used to calculate mean daily levels of fatty acids during postnatal days 1 to 28. Blood samples were obtained at the postnatal ages of 1, 3, 7, 14, and 28 days. Results: A total of 175 infants were included in analysis. Of these infants, 99 were boys (56.6%); the median (IQR) gestational age was 25 weeks 5 days (24 weeks 3 days to 26 weeks 6 days), and the median (IQR) birth weight was 785 (650-945) grams. A higher DHA proportion was seen in infants with no ROP compared with those with mild or moderate ROP or severe ROP (OR per 0.5-molar percentage increase, 0.49 [95% CI, 0.36-0.68]; gestational age- and birth weight-adjusted OR, 0.66 [95% CI, 0.46-0.93]). The corresponding adjusted OR for AA levels per 1-molar percentage increase was 0.83 (95% CI, 0.66-1.05). The association between DHA levels and ROP severity appeared only in infants with sufficient AA levels, suggesting that a mean daily minimum level of 7.8 to 8.3 molar percentage of AA was necessary for a detectable association between DHA level and less severe ROP. Conclusions and Relevance: This cohort study found that higher mean daily serum levels of DHA during the first 28 postnatal days were associated with less severe ROP even after adjustment for known risk factors, but only in infants with sufficiently high AA levels. Further studies are needed to identify LC-PUFA supplementation strategies that may prevent ROP and other morbidities.


Assuntos
Ácido Araquidônico/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Retinopatia da Prematuridade/etiologia , Ácido Araquidônico/uso terapêutico , Estudos de Coortes , Ácidos Docosa-Hexaenoicos/uso terapêutico , Feminino , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro/metabolismo , Recém-Nascido Prematuro/fisiologia , Modelos Logísticos , Masculino , Razão de Chances , Retinopatia da Prematuridade/epidemiologia , Suécia
5.
Nutr Hosp ; 38(5): 1101-1112, 2021 Oct 13.
Artigo em Espanhol | MEDLINE | ID: mdl-34465121

RESUMO

INTRODUCTION: Long-chain polyunsaturated fatty acids (LC-PUFAs) are critical for infant growth and development, particularly arachidonic acid (ARA, C20:4n-6) and docosahexaenoic acid (DHA, C22:6n-3). ARA and DHA are components of cell membrane phospholipids and play an important role in cell division, differentiation, and signaling; and DHA is the n-3 fatty acid predominant in the developing brain and retina. During the third trimester of pregnancy, LC-PUFAs increase substantially in fetal circulation, and a "biomagnification" process in the fetal brain is observed. Moreover, LC-PUFAs are precursors of eicosanoids and metabolites, which modulate the intensity and duration of the immune response. LC-PUFA synthesis implies complex desaturation and elongation processes on their principal precursors, linoleic acid (LA) (18:3 n-6) (series n-6) and α-linolenic acid (LNA) (20:3 n-3) (series n-3), where fatty acid desaturases (FADS) and elongases (ELOVL) are competing. It is important to notice that during the first months of life, as a consequence of low enzymatic activity, LC-PUFA synthesis from LA and LNA is reduced, especially in those infants carrying variations in the FADS and ELOVL genes, which are involved in LC-PUFA synthesis, and so they are unable to supply their own DHA and ARA needs. Homozygote infants for FADS haplotype A (97 % of the Latinoamerican population) show low levels of ARA (only 43 %) and DHA (only 24 %) when compared to those carrying haplotype D (more prevalent in Europe, Africa and Asia). Human milk is the only source of LA, LNA, ARA, and DHA for the neonate and infant till complementary feeding (CF) is introduced. Infants fed with infant formulas must receive enough amounts of LA, LNA, ARA, and DHA to cover their nutritional requirements. The new guidelines by the European Food Safety Authority (EFSA) (2016) recommend that infant formulas and follow-on formulas must contain 20-50 mg of DHA/100 kcal (0.5-1 % of total fatty acids, which is higher than in human milk and the majority of infant formulas in the market), and it is not necessary to add ARA. This new regulation, which is already applicable since February 2020, has resulted in profound controversy because there is no scientific evidence about its appropriateness and safety for healthy children. Then, different international expert groups have revised the research already published about the effects of ARA and DHA addition to infant formulas, and discussed different emerging questions from this European directive. The expert group led from the University of Granada (Spain) recommends the addition of ARA in similar or higher concentrations than those of DHA, at least equal to those present in human milk (0.3 % of total fatty acids), although preferably 0.5 % and up to around 0.64 % of total fatty acids, since new studies confirm the optimal intake of ARA and DHA during the different developmental stages. This recommendation could be of particular importance for infants carrying the haplotype A of FADS.


INTRODUCCIÓN: Los ácidos grasos poliinsaturados de cadena larga (AGPI-CL) son críticos para el crecimiento y desarrollo infantil, en particular los ácidos araquidónico (ARA, C20:4n-6) y docosahexaenoico (DHA, C22:6n-3). El ARA y el DHA son componentes de los fosfolípidos de las membranas celulares y desempeñan importantes funciones en la división, diferenciación y señalización celular, siendo el DHA el ácido graso de la serie n-3 predominante en el cerebro y la retina en desarrollo. Durante el tercer trimestre de la gestación, los AGPI-CL aumentan de forma sustancial en la circulación fetal, observándose un proceso de "biomagnificación" en el cerebro fetal. Además, los AGPI-CL son precursores de los eicosanoides y metabolitos implicados en la modulación de la intensidad y duración de la respuesta inmunitaria. La síntesis de AGPI-CL implica un complejo proceso de desaturación y elongación desde los precursores principales, el ácido linoleico (18:3 n-6) (LA) (serie n-6) y el ácido α-linolénico (20:3 n-3) (LNA) (serie n-3), por los cuales compiten las enzimas desaturasas (FADS) y elongasas (ELOVL). Es importante indicar que en los primeros meses de vida, como consecuencia de la baja actividad enzimática, la síntesis de AGPI-CL a partir de LA y LNA es reducida, especialmente en los niños con variaciones en los genes que codifican las FADS y ELOVL involucradas en la síntesis de AGPI-CL y que, por tanto, son incapaces de cubrir por sí mismos sus necesidades de ARA y DHA. Los homocigotos para el haplotipo A de las FADS (97 % de la población latinoamericana) muestran niveles de ARA y DHA de tan solo un 43 % y un 24 %, respectivamente, inferiores a los de los individuos con haplotipo D (más frecuente en Europa, África y Asia). La leche humana constituye la única fuente de LA, LNA, ARA y DHA para el recién nacido y el lactante hasta la introducción de la alimentación complementaria (AC). Los niños alimentados con fórmulas infantiles deben recibir las cantidades de LA, LNA, ARA y DHA suficientes para cubrir los requerimientos nutricionales. La nueva normativa de la Autoridad Europea de Seguridad Alimentaria (EFSA) (2016) indica que las fórmulas infantiles de inicio y continuación deben contener entre 20 y 50 mg de DHA/100 kcal (0,5-1 % del total de ácidos grasos: más elevado que en la leche humana y en la mayoría de fórmulas infantiles comercializadas) sin la necesidad de incluir también ARA. Esta nueva regulación, que está vigente desde febrero de 2020, ha despertado una gran controversia, al no existir evidencia científica acerca de su pertinencia y seguridad para los niños sanos. Por ello, diferentes grupos de expertos internacionales han revisado la investigación publicada acerca del ARA y el DHA, y discutido diferentes cuestiones emergentes a partir de esta nueva directiva Europea. El grupo de expertos, liderado desde la Universidad de Granada (España), recomienda la adición de ARA en concentraciones iguales o mayores que las de DHA, alcanzando al menos el contenido presente en la leche humana (0,3 % del total de ácidos grasos), aunque preferiblemente un 0,5 % y hasta alrededor del 0,64 % del total de AG, hasta que nuevos estudios confirmen la ingesta óptima de ARA y DHA durante las distintas etapas del desarrollo. Esta recomendación podría ser de especial importancia para los niños portadores del haplotipo A de las FADS.


Assuntos
Ácidos Araquidônicos/farmacologia , Suplementos Nutricionais/normas , Ácidos Docosa-Hexaenoicos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Alimentos Infantis/normas , Ácidos Araquidônicos/administração & dosagem , Ácidos Araquidônicos/efeitos adversos , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácidos Graxos Insaturados/administração & dosagem , Ácidos Graxos Insaturados/efeitos adversos , Feminino , Humanos , Lactente , Alimentos Infantis/efeitos adversos , Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Recém-Nascido , Masculino , Leite Humano/metabolismo , Leite Humano/fisiologia
6.
Nutrients ; 13(7)2021 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-34371870

RESUMO

Kidney transplant recipients are at high risk of progressive bone loss and low-energy fractures in the years following transplantation. Marine n-3 polyunsaturated fatty acids (n-3 PUFA) supplementation may have beneficial effects on bone strength. The Omega-3 fatty acids in Renal Transplantation (ORENTRA) trial was an investigator initiated, randomized, placebo-controlled trial investigating the effects of marine n-3 PUFA supplementation after kidney transplantation. Effects of supplementation on bone mineral density (BMD) and calcium metabolism were pre-defined secondary endpoints. Adult kidney transplant recipients (n = 132) were randomized to 2.6 g marine n-3 PUFA supplement or olive oil (control) from 8 to 52 weeks post-transplant. Dual energy X-ray absorptiometry was performed to assess changes in bone mineral density of hip, spine, and forearm, as well as trabecular bone score (TBS) of the lumbar spine. Student's t test was used to assess between-group differences. There were no differences in ΔBMD between the two groups (intervention vs. control) at lumbar spine (-0.020 ± 0.08 vs. -0.007 ± 0.07 g/cm², p = 0.34), total hip (0.001 ± 0.03 vs. -0.005 ± 0.04, p = 0.38), or other skeletal sites in the intention-to-treat analyses. There was no difference in the change in TBS score (0.001 ± 0.096 vs. 0.009 ± 0.102, p = 0.62). Finally, no effect on biochemical parameters of mineral metabolism was seen. Results were similar when analyzed per protocol. In conclusion, we found no significant effect of 44 weeks of supplementation with 2.6 g of marine n-3 PUFA on BMD in kidney transplant recipients.


Assuntos
Densidade Óssea/efeitos dos fármacos , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Transplante de Rim , Osteoporose/prevenção & controle , Fraturas por Osteoporose/prevenção & controle , Absorciometria de Fóton , Adulto , Idoso , Biomarcadores/sangue , Cálcio/sangue , Dinamarca , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Método Duplo-Cego , Combinação de Medicamentos , Ácido Eicosapentaenoico/efeitos adversos , Feminino , Humanos , Transplante de Rim/efeitos adversos , Masculino , Pessoa de Meia-Idade , Osteoporose/etiologia , Osteoporose/fisiopatologia , Fraturas por Osteoporose/etiologia , Fraturas por Osteoporose/fisiopatologia , Fatores de Tempo , Resultado do Tratamento
7.
Nutrients ; 13(4)2021 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920232

RESUMO

The purpose of this study is evaluate the efficacy and safety of medicinal products containing the original Age-Related Eye Disease group (AREDS) formulation at doses approved in Europe (EU, control group; n = 59) with a product that adds DHA, lutein, zeaxanthin, resveratrol and hydroxytyrosol to the formula (intervention group; n = 50). This was a multicenter, randomized, observer-blinded trial conducted in patients aged 50 years or older diagnosed with unilateral exudative Age related Macular Degeneration AMD. At month 12, the intervention did not have a significant differential effect on visual acuity compared with the control group, with an estimated treatment difference in Early Treatment Diabetic Retinopathy Study (ETDRS) of -1.63 (95% CI -0.83 to 4.09; p = 0.192). The intervention exhibited a significant and, in most cases, relevant effect in terms of a reduction in some inflammatory cytokines and a greater improvement in the fatty acid profile and serum lutein and zeaxantin concentration. In patients with unilateral wet AMD, the addition of lutein, zeaxanthin, resveratrol, hydroxytyrosol and DHA to the AREDS EU recommended doses in the short-term did not have a differential effect on visual acuity compared to a standard AREDS EU formula but, in addition to improving the fatty acid profile and increasing carotenoid serum levels, may provide a beneficial effect in improving the proinflammatory and proangiogenic profile of patients with AMD.


Assuntos
Suplementos Nutricionais/efeitos adversos , Degeneração Macular/dietoterapia , Nutrientes/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/efeitos adversos , Feminino , Humanos , Luteína/administração & dosagem , Luteína/efeitos adversos , Degeneração Macular/sangue , Degeneração Macular/diagnóstico , Masculino , Pessoa de Meia-Idade , Nutrientes/efeitos adversos , Álcool Feniletílico/administração & dosagem , Álcool Feniletílico/efeitos adversos , Álcool Feniletílico/análogos & derivados , Resveratrol/administração & dosagem , Resveratrol/efeitos adversos , Resultado do Tratamento , Acuidade Visual , Xantofilas/administração & dosagem , Zeaxantinas/administração & dosagem , Zeaxantinas/efeitos adversos
8.
Atherosclerosis ; 322: 15-23, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33706079

RESUMO

Omega-3 fatty acids have emerged as a new option for controlling the residual risk for cardiovascular disease (CVD) in the statin era after a clinical trial (REDUCE-IT) reported positive results with icosapent ethyl (IPE) in patients receiving maximally tolerated statin therapy. However, another trial which used high dose eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) combination (STRENGTH) has failed. Together, these results raise clinically important questions. Are effects of omega-3 fatty acids neutral or beneficial in patients on statin therapy, or perhaps even harmful? The current contradictory results could be attributed to different types of omega-3 fatty acids (only EPA or combination of EPA + DHA), doses (higher vs. lower dose) of omega-3 fatty acids or different comparators (corn oil or mineral oil), as well as the underlying severity of the CVD risk or use of statins. Together with these issues, we will discuss different biological and clinical effects of various types of omega-3 fatty acids and then interpret different results of past and current clinical studies and propose practical suggestions, which could be applied in patient management.


Assuntos
Doenças Cardiovasculares , Ácidos Graxos Ômega-3 , Inibidores de Hidroximetilglutaril-CoA Redutases , Hipertrigliceridemia , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/prevenção & controle , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácido Eicosapentaenoico/efeitos adversos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos
9.
Curr Probl Cardiol ; 46(3): 100718, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33097299

RESUMO

Omega-3-fatty-acids are now increasingly being used for potential beneficial anti-inflammatory effect in the treatment and management of cardiovascular disease. Eicosapentaenoic acid and Docosahexaenoic acid are 2 essential omega-3 fatty acids found predominately in fish and fish oil supplements. Despite the increased use of fish oil products for both primary and secondary prevention of cardiovascular morbidity and mortality, the available literature evidence are controversial. We searched through PubMed for studies that have investigated the impact of omega-3 fatty acids on coronary heart disease and mortality. Our systemic review suggests that most studies, which are mostly observational, have found there to be a potential benefit of omega-3 fatty acids on coronary heart disease whereas some other studies have found conflicting results. More randomized controlled studies are warranted with adequate sample size to clearly establish the risk and benefits of omega-3 fatty acids on cardiovascular disease.


Assuntos
Arritmias Cardíacas , Doenças Cardiovasculares , Ácidos Graxos Ômega-3 , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/prevenção & controle , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/prevenção & controle , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácido Eicosapentaenoico/administração & dosagem , Ácido Eicosapentaenoico/efeitos adversos , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/efeitos adversos , Óleos de Peixe/uso terapêutico , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Prevenção Secundária
10.
Cochrane Database Syst Rev ; 12: CD011679, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33320335

RESUMO

BACKGROUND: Souvenaid is a dietary supplement with a patented composition (Fortasyn Connect™)which is intended to be used by people with Alzheimer's disease (AD). It has been designed to support the formation and function of synapses in the brain, which are thought to be strongly correlated with cognitive function. If effective, it might improve symptoms of Alzheimer's disease and also prevent the progression from prodromal Alzheimer's disease to dementia. We sought in this review to examine the evidence for this proposition. OBJECTIVES: To assess the effects of Souvenaid on incidence of dementia, cognition, functional performance, and safety in people with Alzheimer's disease. SEARCH METHODS: We searched ALOIS, i.e. the specialised register of the Cochrane Dementia and Cognitive Improvement Group, MEDLINE (Ovid SP), Embase (Ovid SP), PsycINFO (Ovid SP), Web of Science (ISI Web of Science), Cinahl (EBSCOhost), Lilacs (BIREME), and clinical trials registries up to 24 June 2020. We also reviewed citations of reference lists of landmark papers, reviews, and included studies for additional studies and assessed their suitability for inclusion in the review. SELECTION CRITERIA: We included randomised, placebo-controlled trials which evaluated Souvenaid in people diagnosed with mild cognitive impairment (MCI) due to AD (also termed prodromal AD) or with dementia due to AD, and with a treatment duration of at least 16 weeks. DATA COLLECTION AND ANALYSIS: Our primary outcome measures were incidence of dementia, global and specific cognitive function, functional performance, combined cognitive-functional outcomes and adverse events. We selected studies, extracted data, assessed the quality of trials and intended to conduct meta-analyses according to the Cochrane Handbook for Systematic Reviews of Interventions. We rated the quality of the evidence using the GRADE approach. We present all outcomes grouped by stage of AD. MAIN RESULTS: We included three randomised, placebo-controlled trials investigating Souvenaid in 1097 community-dwelling participants with Alzheimer's disease. One study each included participants with prodromal AD, mild AD dementia and mild-to-moderate AD dementia. We rated the risks of bias of all trials as low. One study (in prodromal AD) was funded by European grants. The other two studies were funded by the manufacturer of Souvenaid. One trial investigated the incidence of dementia in people with prodromal AD at baseline, and found little to no difference between the Souvenaid group and the placebo group after 24 months (RR 1.09, 95% CI 0.82 to 1.43; 1 trial, 311 participants; moderate quality of evidence). In prodromal AD, and in mild and mild-to-moderate Alzheimer's disease dementia, Souvenaid probably results in little or no difference in global or specific cognitive functions (moderate quality of evidence). Everyday function, or the ability to perform activities of daily living, were measured in mild and mild-to-moderate AD dementia. Neither study found evidence of a difference between the groups after 24 weeks of treatment (moderate quality of evidence). Two studies investigated combined cognitive-functional outcomes with the Clinical Dementia Rating Sum of Boxes and observed conflicting results. Souvenaid probably results in slight improvement, which is below estimates of meaningful change, in participants with prodromal Alzheimer's disease after 24 months (moderate quality of evidence), but probably has little to no effect in mild-to-moderate Alzheimer's disease dementia after 24 weeks (moderate quality of evidence). Adverse effects observed were low in all trials, and the available data were insufficient to determine any connection with Souvenaid. AUTHORS' CONCLUSIONS: Two years of treatment with Souvenaid probably does not reduce the risk of progression to dementia in people with prodromal AD. There is no convincing evidence that Souvenaid affects other outcomes important to people with AD in the prodromal stage or mild-to-moderate stages of dementia. Conflicting evidence on combined cognitive-functional outcomes in prodromal AD and mild AD dementia warrants further investigation. Adverse effects of Souvenaid seem to be uncommon, but the evidence synthesised in this review does not permit us to make a definitive statement on the long-term tolerability of Souvenaid. The effects of Souvenaid in more severe AD dementia or in people with AD at risk of nutritional deficiencies remain unclear.


Assuntos
Doença de Alzheimer/dietoterapia , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ácido Eicosapentaenoico/uso terapêutico , Fosfolipídeos/uso terapêutico , Viés , Cognição , Demência/prevenção & controle , Suplementos Nutricionais/efeitos adversos , Progressão da Doença , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácidos Docosa-Hexaenoicos/química , Ácido Eicosapentaenoico/efeitos adversos , Ácido Eicosapentaenoico/química , Humanos , Fosfolipídeos/efeitos adversos , Fosfolipídeos/química , Placebos/uso terapêutico , Sintomas Prodrômicos , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Tempo
11.
Nutr Metab Cardiovasc Dis ; 30(10): 1795-1799, 2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-32723580

RESUMO

Type 2 Diabetes mellitus is associated with aging and shortened telomere length. Telomerase replaces lost telomeric repeats at the ends of chromosomes and is necessary for the replicative immortality of cells. Aspirin and the n3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are commonly used therapies in people with type 2 diabetes for reducing cardiovascular disease events, though their relation to telomerase activity is not well studied. We explored the effects of aspirin, EPA + DHA, and the combined effects of aspirin and EPA + DHA treatment on telomerase activity in 30 adults with diabetes mellitus. EPA and DHA ingestion alone increased telomerase activity then a decrease occurred with the addition of aspirin consumption. Crude (F-stat = 2.09, p = 0.13) and adjusted (F-stat = 2.20, p = 0.14) analyses of this decrease showed signs of a trend. These results suggest that aspirin has an adverse effect on aging in diabetics who have relatively high EPA and DHA ingestion.


Assuntos
Aspirina/administração & dosagem , Diabetes Mellitus Tipo 2/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Telomerase/metabolismo , Homeostase do Telômero/efeitos dos fármacos , Adulto , Idoso , Idoso de 80 Anos ou mais , Aspirina/efeitos adversos , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/enzimologia , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácido Eicosapentaenoico/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , New York , Resultado do Tratamento
12.
Cochrane Database Syst Rev ; 6(6): CD008428, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32573764

RESUMO

BACKGROUND: Retinitis pigmentosa (RP) comprises a group of hereditary eye diseases characterized by progressive degeneration of retinal photoreceptors. It results in severe visual loss that may lead to blindness. Symptoms may become manifest during childhood or adulthood which include poor night vision (nyctalopia) and constriction of peripheral vision (visual field loss). Visual field loss is progressive and affects central vision later in the disease course. The worldwide prevalence of RP is approximately 1 in 4000, with 100,000 individuals affected in the USA. At this time, there is no proven therapy for RP. OBJECTIVES: The objective of this review was to synthesize the best available evidence regarding the effectiveness and safety of vitamin A and fish oils (docosahexaenoic acid (DHA)) in preventing the progression of RP. SEARCH METHODS: We searched the Cochrane Central Register of Controlled Trials (CENTRAL), which contains the Cochrane Eyes and Vision Trials Register (2020, Issue 2); Ovid MEDLINE; Embase.com; PubMed; Latin American and Caribbean Health Sciences Literature Database (LILACS); ClinicalTrials.gov; the World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP); and OpenGrey. We did not use any date or language restrictions in the electronic searches for trials. We last searched the electronic databases on 7 February 2020. SELECTION CRITERIA: We included randomized controlled trials that enrolled participants of any age diagnosed with any degree of severity or type of RP, and evaluated the effectiveness of vitamin A, fish oils (DHA), or both compared to placebo, vitamins (other than vitamin A), or no therapy, as a treatment for RP. We excluded cluster-randomized trials and cross-over trials. DATA COLLECTION AND ANALYSIS: We prespecified the following outcomes: mean change from baseline visual field, mean change from baseline electroretinogram (ERG) amplitudes, and anatomic changes as measured by optical coherence tomography (OCT), at one-year follow-up, and mean change in visual acuity, at five-year follow-up. Two review authors independently extracted data and evaluated risk of bias for all included trials. We also contacted study investigators for further information when necessary. MAIN RESULTS: In addition to three trials from the previous version of this review, we included a total of four trials with 944 participants aged 4 to 55 years. Two trials included only participants with X-linked RP and the other two included participants with RP of all forms of genetic predisposition. Two trials evaluated the effect of DHA alone; one trial evaluated vitamin A alone; and one trial evaluated DHA and vitamin A versus vitamin A alone. Two trials recruited participants from the USA, and the other two recruited from the USA and Canada. All trials were at low risk of bias for most domains. We did not perform meta-analysis due to clinical heterogeneity. Four trials assessed visual field sensitivity. Investigators found no evidence of a difference in mean values between the groups. However, one trial found that the annual rate of change of visual field sensitivity over four years favored the DHA group in foveal (-0.02 ± 0.55 (standard error (SE)) dB versus -0.47 ± 0.03 dB, P = 0.039), macular (-0.42 ± 0.05 dB versus -0.85 ± 0.03 dB, P = 0.031), peripheral (-0.39 ± 0.02 versus -0.86 ± 0.02 dB, P < 0.001), and total visual field sensitivity (-0.39 ± 0.02 versus -0.86 ± 0.02 dB, P < 0.001). The certainty of the evidence was very low. The four trials evaluated visual acuity (LogMAR scale) at a follow-up of four to six years. In one trial (208 participants), investigators found no evidence of a difference between the two groups, as both groups lost 0.7 letters of the Early Treatment Diabetic Retinopathy Study (ETDRS) visual acuity per year. In another trial (41 participants), DHA showed no evidence of effect on visual acuity (mean difference -0.01 logMAR units (95% confidence interval -0.14 to 0.12; one letter difference between the two groups; very low-certainty evidence). In the third trial (60 participants), annual change in mean number of letters correct was -0.8 (DHA) and 1.4 letters (placebo), with no evidence of between-group difference. In the fourth trial (572 participants), which evaluated (vitamin A + vitamin E trace) compared with (vitamin A trace + vitamin E trace), decline in ETDRS visual acuity was 1.1 versus 0.9 letters per year, respectively. All four trials reported electroretinography (ERG). Investigators of two trials found no evidence of a difference between the DHA and placebo group in yearly rates of change in 31 Hz cone ERG amplitude (mean ± SE) (-0.028 ± 0.001 log µV versus -0.022 ± 0.002 log µV; P = 0.30); rod ERG amplitude (mean ± SE) (-0.010 ± 0.001 log µV versus -0.023 ± 0.001 log µV; P = 0.27); and maximal ERG amplitude (mean ± SE) (-0.042 ± 0.001 log µV versus -0.036 ± 0.001 log µV; P = 0.65). In another trial, a slight difference (6.1% versus 7.1%) in decline of ERG per year favored vitamin A (P = 0.01). The certainty of the evidence was very low. One trial (51 participants) that assessed optical coherence tomography found no evidence of a difference in ellipsoid zone constriction (P = 0.87) over two years, with very low-certainty evidence. The other three trials did not report this outcome. Only one trial reported adverse events, which found that 27/60 participants experienced 42 treatment-related emergent adverse events (22 in DHA group, 20 in placebo group). The certainty of evidence was very low. The rest of the trials reported no adverse events, and no study reported any evidence of benefit of vitamin supplementation on the progression of visual acuity loss. AUTHORS' CONCLUSIONS: Based on the results of four studies, it is uncertain if there is a benefit of treatment with vitamin A or DHA, or both for people with RP. Future trials should also take into account the changes observed in ERG amplitudes and other outcome measures from trials included in this review.


Assuntos
Ácidos Docosa-Hexaenoicos/uso terapêutico , Retinose Pigmentar/terapia , Vitamina A/uso terapêutico , Vitaminas/uso terapêutico , Adolescente , Adulto , Criança , Pré-Escolar , Terapia Combinada/métodos , Progressão da Doença , Ácidos Docosa-Hexaenoicos/efeitos adversos , Eletrorretinografia , Feminino , Óleos de Peixe/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto , Retinose Pigmentar/genética , Acuidade Visual , Campos Visuais/fisiologia , Vitamina A/efeitos adversos , Vitaminas/efeitos adversos , Adulto Jovem
13.
Yakugaku Zasshi ; 140(5): 723-728, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32378676

RESUMO

The widespread use of health foods, including supplements, is now common among patients. This is because many health foods are being claimed to be beneficial. If patients use medicines and health foods concurrently, the interaction between the two might lead to adverse events. Additionally, it is reported that pharmacists do not generally care about health food use in their patients, because they also lack sufficient knowledge about health foods. On the contrary, there are some licenses to be a health food advisor in Japan, and the generic name of these licenses is "advisory staff". Pharmacists who have this license are specialists in both medicines and health foods, and thus, they might pay more attention to the concurrent use of medicines and health foods compared to those who do not have the advisory staff license. To address this issue, we conducted a study with an online questionnaire about health food consultation, and 87 pharmacists with advisory staff license participated. Only 36.8% of participants were found to always ask their patients about health food use. However, 92.0% of them had experience of consultation about the simultaneous use of medicines and health foods, and 17.2% of them recognized adverse events by knowing about the concurrent use. Patients who experienced adverse events have used either eicosapentaenoic acid/docosahexaenoic acid supplement with epadel or Ginkgo biloba extract with warfarin. Therefore, an active interview with pharmacists is important to avoid such adverse events in patients.


Assuntos
Suplementos Nutricionais , Interações Medicamentosas , Interações Alimento-Droga , Alimentos Especializados , Alimento Funcional , Licenciamento , Farmacêuticos , Encaminhamento e Consulta , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácido Eicosapentaenoico/efeitos adversos , Alimentos Especializados/efeitos adversos , Alimento Funcional/efeitos adversos , Ginkgo biloba , Humanos , Japão , Conhecimento , Extratos Vegetais/efeitos adversos , Encaminhamento e Consulta/estatística & dados numéricos , Inquéritos e Questionários , Varfarina/efeitos adversos
14.
Nutr Cancer ; 72(3): 439-450, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31290697

RESUMO

Background: This pilot, double-blind, comparator-controlled trial evaluated the safety and tolerability of an oral targeted medical nutrition (TMN) supplement for the management of cachexia in patients with non-small-cell lung cancer (NSCLC).Methods: Patients receiving first-line chemotherapy for NSCLC with weight loss or low BMI were randomized 1:1 to receive juice-based TMN (∼200 kcal; 10 g whey protein; ≥2.0 g eicosapentaenoic acid/docosahexaenoic acid in fish oil; and 10 µg 25-hydroxy-vitamin D3) or a milk-based isocaloric comparator twice daily for 12 weeks (ClinicalTrials.gov: NCT02515032). Primary endpoints included number/type of adverse events and changes in vital signs/laboratory parameters. Secondary endpoints included measures of clinical relevance. Survival was an exploratory endpoint.Results: The TMN group (n = 26; mean 64.4 years) experienced fewer adverse events (64 vs. 87) than the comparator group (n = 29; mean 66.0 years), including fewer cases of neutropenia (0 vs. 4). Compliance was slightly lower in the TMN (58.5%) vs. comparator group (73.6%). There were no statistically significant between-group differences in efficacy endpoints. Fewer (4 vs. 10) patients who received TMN than comparator had died by 1-year post baseline.Conclusions: TMN was well tolerated. Trends for improved clinical outcomes with TMN identified in this study warrant further investigation.


Assuntos
Caquexia/dietoterapia , Carcinoma Pulmonar de Células não Pequenas/complicações , Suplementos Nutricionais/estatística & dados numéricos , Neoplasias Pulmonares/complicações , Idoso , Antineoplásicos/uso terapêutico , Peso Corporal/efeitos dos fármacos , Caquexia/complicações , Calcifediol/administração & dosagem , Calcifediol/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/efeitos adversos , Método Duplo-Cego , Ácido Eicosapentaenoico/administração & dosagem , Ácido Eicosapentaenoico/efeitos adversos , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Neutropenia/complicações , Projetos Piloto , Resultado do Tratamento , Redução de Peso , Proteínas do Soro do Leite/administração & dosagem , Proteínas do Soro do Leite/efeitos adversos
15.
Nutr Metab Cardiovasc Dis ; 30(3): 441-447, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-31831363

RESUMO

BACKGROUND AND AIMS: Omega-3 polyunsaturated fatty acids (PUFAs) are natural peroxisome proliferator-activated receptor gamma (PPAR-γ) ligands. Activated PPAR-γ protects the cardiovascular system against atherosclerotic lesion formation and exerts its anti-inflammatory role by suppressing cytokines induced by nuclear factor kappa-B (NF-κB) in endothelial cells (ECs), and it is hypothesized that apoptosis and cell cycle arrest induced by PPAR-γ ligands may be mediated by the p53-dependent pathway. The aim of our study was to investigate the effects of docosahexaenoic acid (DHA)-enriched fish oil supplement on PPAR-γ activity and mRNA expression levels of p53 and NF-κB. METHODS AND RESULTS: Fifty patients with type 2 diabetes mellitus (T2DM) aged 30-70 years were randomly assigned to receive either 2400 mg/d DHA-rich fish oil or placebo for 8 weeks. Metabolic parameters were assessed at baseline and at the end of the intervention. PPAR-γ activity in the peripheral blood mononuclear cells (PBMCs) was measured using ELISA-based PPAR-γ Transcription Factor Assay Kit, and the gene expression levels of p53 and NF-κB were assessed using real-time quantitative reverse transcription polymerase chain reaction (RT-PCR). On the basis of our finding, 8 weeks of treatment with DHA-rich fish oil increased PPAR-γ activity in PBMCs of subjects with T2DM (p < 0.01) compared to that in placebo (p = 0.4). Between-group comparisons of mean PPAR-γ activity changes showed significant differences (p = 0.03), whereas mRNA expression levels of the p53 and NF-κB genes did not show significant differences between studied groups (p = 0.2 and p = 0.5, respectively). CONCLUSION: Our findings indicated that short-term DHA-rich fish oil supplementation may modulate PPAR-γ activity in PBMCs.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Leucócitos Mononucleares/efeitos dos fármacos , NF-kappa B/sangue , PPAR gama/sangue , Proteína Supressora de Tumor p53/metabolismo , Adulto , Idoso , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Irã (Geográfico) , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , Fatores de Tempo , Resultado do Tratamento , Proteína Supressora de Tumor p53/genética
16.
JAMA ; 322(19): 1899-1909, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31703120

RESUMO

Importance: Chronic kidney disease (CKD) is a common complication of type 2 diabetes that can lead to end-stage kidney disease and is associated with high cardiovascular risk. Few treatments are available to prevent CKD in type 2 diabetes. Objective: To test whether supplementation with vitamin D3 or omega-3 fatty acids prevents development or progression of CKD in type 2 diabetes. Design, Setting, and Participants: Randomized clinical trial with a 2 × 2 factorial design conducted among 1312 adults with type 2 diabetes recruited between November 2011 and March 2014 from all 50 US states as an ancillary study to the Vitamin D and Omega-3 Trial (VITAL), coordinated by a single center in Massachusetts. Follow-up was completed in December 2017. Interventions: Participants were randomized to receive vitamin D3 (2000 IU/d) and omega-3 fatty acids (eicosapentaenoic acid and docosahexaenoic acid; 1 g/d) (n = 370), vitamin D3 and placebo (n = 333), placebo and omega-3 fatty acids (n = 289), or 2 placebos (n = 320) for 5 years. Main Outcomes and Measures: The primary outcome was change in glomerular filtration rate estimated from serum creatinine and cystatin C (eGFR) from baseline to year 5. Results: Among 1312 participants randomized (mean age, 67.6 years; 46% women; 31% of racial or ethnic minority), 934 (71%) completed the study. Baseline mean eGFR was 85.8 (SD, 22.1) mL/min/1.73 m2. Mean change in eGFR from baseline to year 5 was -12.3 (95% CI, -13.4 to -11.2) mL/min/1.73 m2 with vitamin D3 vs -13.1 (95% CI, -14.2 to -11.9) mL/min/1.73 m2 with placebo (difference, 0.9 [95% CI, -0.7 to 2.5] mL/min/1.73 m2). Mean change in eGFR was -12.2 (95% CI, -13.3 to -11.1) mL/min/1.73 m2 with omega-3 fatty acids vs -13.1 (95% CI, -14.2 to -12.0) mL/min/1.73 m2 with placebo (difference, 0.9 [95% CI, -0.7 to 2.6] mL/min/1.73 m2). There was no significant interaction between the 2 interventions. Kidney stones occurred among 58 participants (n = 32 receiving vitamin D3 and n = 26 receiving placebo) and gastrointestinal bleeding among 45 (n = 28 receiving omega-3 fatty acids and n = 17 receiving placebo). Conclusions and Relevance: Among adults with type 2 diabetes, supplementation with vitamin D3 or omega-3 fatty acids, compared with placebo, resulted in no significant difference in change in eGFR at 5 years. The findings do not support the use of vitamin D or omega-3 fatty acid supplementation for preserving kidney function in patients with type 2 diabetes. Trial Registration: ClinicalTrials.gov Identifier: NCT01684722.


Assuntos
Colecalciferol/uso terapêutico , Diabetes Mellitus Tipo 2/complicações , Ácidos Graxos Ômega-3/uso terapêutico , Insuficiência Renal Crônica/prevenção & controle , Vitaminas/uso terapêutico , Idoso , Colecalciferol/efeitos adversos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/etnologia , Progressão da Doença , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácidos Docosa-Hexaenoicos/uso terapêutico , Método Duplo-Cego , Quimioterapia Combinada/métodos , Ácido Eicosapentaenoico/efeitos adversos , Ácido Eicosapentaenoico/uso terapêutico , Ácidos Graxos Ômega-3/efeitos adversos , Feminino , Hemorragia Gastrointestinal/induzido quimicamente , Taxa de Filtração Glomerular/efeitos dos fármacos , Taxa de Filtração Glomerular/fisiologia , Humanos , Rim/efeitos dos fármacos , Rim/fisiologia , Cálculos Renais/induzido quimicamente , Masculino , Adesão à Medicação/estatística & dados numéricos , Pessoa de Meia-Idade , Placebos/uso terapêutico , Insuficiência Renal Crônica/etnologia , Insuficiência Renal Crônica/etiologia , Fatores de Tempo , Estados Unidos , Vitaminas/efeitos adversos
17.
Nutrients ; 11(12)2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31757095

RESUMO

Resting heart rate (rHR) and heart rate variability (HRV) are non-invasive measurements that predict the risk of sudden cardiac death (SCD). Marine n-3 polyunsaturated fatty acid (PUFA) supplementation may decrease rHR, increase HRV, and reduce the risk of SCD. To date, no studies have investigated the effect of marine n-3 PUFA on HRV in renal transplant recipients. In a randomized controlled trial, 132 renal transplant recipients were randomized to receive either three 1 g capsules of marine n-3 PUFA, each containing 460 mg/g EPA and 380 mg/g DHA, or control (olive oil) for 44 weeks. HRV was calculated in the time and frequency domains during a conventional cardiovascular reflex test (response to standing, deep breathing, and Valsalva maneuver) and during 2 min of resting in the supine position. There was no significant effect of marine n-3 PUFA supplementation on time-domain HRV compared with controls. rHR decreased 3.1 bpm (± 13.1) for patients receiving marine n-3 PUFA compared to 0.8 (± 11.0) in controls (p = 0.28). In the frequency domain HRV analyses, there was a significant change in response to standing in both high and low frequency measures, 2.9 (p = 0.04, 95% CI (1.1;8)) and 2.7 (p = 0.04, 95% CI (1.1;6.5)), respectively. In conclusion, 44 weeks of supplemental marine n-3 PUFAs in renal transplant recipients significantly improved the cardiac autonomic function, assessed by measuring HRV during conventional cardiovascular reflex tests.


Assuntos
Sistema Nervoso Autônomo/efeitos dos fármacos , Morte Súbita Cardíaca/prevenção & controle , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Frequência Cardíaca/efeitos dos fármacos , Coração/inervação , Transplante de Rim , Transplantados , Adulto , Idoso , Sistema Nervoso Autônomo/fisiopatologia , Morte Súbita Cardíaca/etiologia , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Método Duplo-Cego , Combinação de Medicamentos , Ácido Eicosapentaenoico/efeitos adversos , Feminino , Humanos , Transplante de Rim/efeitos adversos , Transplante de Rim/mortalidade , Masculino , Pessoa de Meia-Idade , Noruega , Fatores de Tempo , Resultado do Tratamento
18.
Nutrients ; 11(7)2019 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-31284588

RESUMO

Evidence suggests a role of long chain polyunsaturated fatty acids (LC-PUFA), in which animal foods are especially rich, in optimal neural development. The LC-PUFAs docosahexaenoic acid (DHA) and arachidonic acid, found in high concentrations in the brain and retina, have potential beneficial effects on cognition, and motor and visual functions. Phenylketonuria (PKU) is the most common inborn error of amino acid metabolism. The treatment of PKU consists of a phenylalanine-free diet, which limits the intake of natural proteins of high biological value. In this systematic review, we summarize the available evidence supporting a role for LC-PUFA supplementation as an effective means of increasing LC-PUFA levels and improving visual and neurocognitive functions in PKU patients. Data from controlled trials of children and adults (up to 47 years of age) were obtained by searching the MEDLINE and SCOPUS databases following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. For each selected study, the risk of bias was assessed applying the methodology of the Cochrane Collaboration. The findings indicate that DHA supplementation in PKU patients from 2 weeks to 47 years of age improves DHA status and decreases visual evoked potential P100 wave latency in PKU children from 1 to 11 years old. Neurocognitive data are inconclusive.


Assuntos
Ácido Araquidônico/administração & dosagem , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Fenilcetonúrias/dietoterapia , Adolescente , Adulto , Ácido Araquidônico/efeitos adversos , Criança , Pré-Escolar , Cognição , Dieta com Restrição de Proteínas , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Potencial Evocado Motor , Potenciais Evocados Visuais , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Fenilcetonúrias/diagnóstico , Fenilcetonúrias/fisiopatologia , Fenilcetonúrias/psicologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento , Adulto Jovem
19.
Saudi J Kidney Dis Transpl ; 30(3): 634-639, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31249227

RESUMO

Atherosclerosis, cardiovascular diseases (CVDs), and inflammation are important problems in chronic kidney disease (CKD) patients on chronic hemodialysis (HD). Prevention of CKD patients of CVD can lower the mortality rate in them. Omega 3 may help in the treatment of inflammation and lipid abnormalities in end-stage renal disease (ESRD) patients on chronic HD. This study aimed to evaluate the effect of high-dose Omega 3 on lipid profile and inflammatory markers in ESRD children on chronic HD. This clinical trial was conducted on 49 ESRD children on chronic HD selected from pediatric HD unit of Menoufia University Hospital. The study was conducted for a period of three months. The selected children were supplemented with 2000-mg Omega 3 tablets daily; lipid profile and inflammatory markers were assessed at the beginning and at the end of the study. Supplementation with high-dose Omega 3 resulted in a highly statistically significant decrease in total cholesterol, triglyceride, low-density lipoprotein, interleukin-6, C- reactive protein levels and a highly statistically significant increase in high-density lipoprotein (P <0.001). The differences between hemoglobin, platelets, iron profile, parathyroid hormone, albumin, phosphorus, calcium, potassium, sodium, and efficiency of dialysis before and after Omega 3 supplementation were not statistically significant (P >0.05). Supplementation with highdose Omega 3 caused significant improvement in lipid profile and inflammatory markers of children on chronic HD.


Assuntos
Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Mediadores da Inflamação/sangue , Lipídeos/sangue , Diálise Renal , Insuficiência Renal Crônica/terapia , Adolescente , Fatores Etários , Biomarcadores/sangue , Criança , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Egito , Ácido Eicosapentaenoico/efeitos adversos , Feminino , Humanos , Masculino , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/diagnóstico , Fatores de Tempo , Resultado do Tratamento
20.
PLoS One ; 14(6): e0217794, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31158258

RESUMO

An Algal Oil Containing EPA and DHA (AOCED) at ~50% was developed as a sustainable source of omega-3 fatty acids. AOCED was incorporated into extruded dry foods for dogs at 0, 0.75%, 1.5% and 3.0% levels (equivalent to 0, 7.5, 15 and 30 g/kg diet) on dry matter basis at the expense of chicken fat and fed to healthy female Beagle dogs starting at mating and throughout gestation and lactation. The offspring were fed their maternal corresponding diets for 26 weeks after weaning. AOCED-enriched diets were well tolerated by dogs in both generations and did not affect their overall health, physiological parameters, food consumption, body weights and body weight gains. There were no changes in hematology, clinical chemistry, and coagulation parameters in both generations of dogs fed the AOCED diets when compared to those in the control group. Plasma levels of DHA and EPA increased significantly and generally dose-dependently in both generations. The study demonstrated the safety of AOCED in dogs during gestation, lactation, and growth periods at dietary levels up to 3.0wt%, equivalent to 30 g/kg diet. AOCED's bioavailability as a source of DHA and EPA in dogs was demonstrated by the increased plasma concentrations of these nutritional lipids.


Assuntos
Ração Animal/efeitos adversos , Ácidos Docosa-Hexaenoicos/efeitos adversos , Ácido Eicosapentaenoico/efeitos adversos , Comportamento Alimentar , Lactação , Animais , Animais Recém-Nascidos , Peso Corporal , Dieta/veterinária , Ácidos Docosa-Hexaenoicos/sangue , Cães , Ácido Eicosapentaenoico/sangue , Feminino , Masculino , Óleos , Gravidez , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA