Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
J Diabetes Complications ; 38(4): 108722, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38503000

RESUMO

BACKGROUNDS: Non-alcoholic fatty liver disease (NAFLD) is a common condition affecting >25 % of the population worldwide. This disorder ranges in severity from simple steatosis (fat accumulation) to severe steatohepatitis (inflammation), fibrosis and, at its end-stage, liver cancer. A number of studies have identified overexpression of several key genes that are critical in the initiation and progression of NAFLD. MiRNAs are potential therapeutic agents that can regulate several genes simultaneously. Therefore, we transfected cell lines with two key miRNAs involved in targeting NAFLD-related genes. METHODS: The suppression effects of the investigated miRNAs (miR-124 and miR-16) and genes (TNF, TLR4, SCD, FASN, SREBF2, and TGFß-1) from our previous study were investigated by real-time PCR in Huh7 and HepG2 cells treated with oleic acid. Oil red O staining and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay were utilized to assess cell lipid accumulation and cytotoxic effects of the miRNAs, respectively. The pro-oxidant-antioxidant balance (PAB) assay was undertaken for miR-16 and miR-124 after cell transfection. RESULTS: Following transfection of miRNAs into HepG2, oil red O staining showed miR-124 and miR-16 reduced oleic acid-induced lipid accumulation by 35.2 % and 28.6 % respectively (p < 0.05). In Huh7, miR-124 and miR-16 reduced accumulation by 23.5 % and 31.3 % respectively (p < 0.05) but without impacting anti-oxidant activity. Real-time PCR in HepG2 revealed miR-124 decreased expression of TNF by 0.13-fold, TLR4 by 0.12-fold and SREBF2 by 0.127-fold (p < 0.05). miR-16 decreased TLR4 by 0.66-fold and FASN by 0.3-fold (p < 0.05). In Huh7, miR-124 decreased TNF by 0.12-fold and FASN by 0.09-fold (p < 0.05). miR-16 decreased SCD by 0.28-fold and FASN by 0.64-fold (p < 0.05). MTT assays showed, in HepG2, viability was decreased 24.7 % by miR-124 and decreased 33 % by miR-16 at 72 h (p < 0.05). In Huh7, miR-124 decreased viability 42 % at 48 h and 29.33 % at 72 h (p < 0.05), while miR-16 decreased viability by 32.3 % (p < 0.05). CONCLUSION: These results demonstrate the ability of miR-124 and miR-16 to significantly reduce lipid accumulation and expression of key pathogenic genes associated with NAFLD through direct targeting. Though this requires further in vivo investigation.


Assuntos
Compostos Azo , MicroRNAs , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico , Receptor 4 Toll-Like , Metabolismo dos Lipídeos/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fígado/metabolismo
2.
Nutrients ; 15(20)2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37892524

RESUMO

Acylethanolamides (NAEs) are bioactive lipids derived from diet fatty acids that modulate important homeostatic functions, including appetite, fatty acid synthesis, mitochondrial respiration, inflammation, and nociception. Among the naturally circulating NAEs, the pharmacology of those derived from either arachidonic acid (Anandamide), oleic acid (OEA), and palmitic acid (PEA) have been extensively characterized in diet-induced obesity. For the present work, we extended those studies to linoleoylethanolamide (LEA), one of the most abundant NAEs found not only in plasma and body tissues but also in foods such as cereals. In our initial study, circulating concentrations of LEA were found to be elevated in overweight humans (body mass index (BMI, Kg/m2) > 25) recruited from a representative population from the south of Spain, together with AEA and the endocannabinoid 2-Arachidonoyl glycerol (2-AG). In this population, LEA concentrations correlated with the circulating levels of cholesterol and triglycerides. In order to gain insight into the pharmacology of LEA, we administered it for 14 days (10 mg/kg i.p. daily) to obese male Sprague Dawley rats receiving a cafeteria diet or a standard chow diet for 12 consecutive weeks. LEA treatment resulted in weight loss and a reduction in circulating triglycerides, cholesterol, and inflammatory markers such as Il-6 and Tnf-alpha. In addition, LEA reduced plasma transaminases and enhanced acetyl-CoA-oxidase (Acox) and Uncoupling protein-2 (Ucp2) expression in the liver of the HFD-fed animals. Although the liver steatosis induced by the HFD was not reversed by LEA, the overall data suggest that LEA contributes to the homeostatic signals set in place in response to diet-induced obesity, potentially contributing with OEA to improve lipid metabolism after high fat intake. The anti-inflammatory response associated with its administration suggests its potential for use as a nutrient supplement in non-alcoholic steatohepatitis.


Assuntos
Dislipidemias , Hepatopatia Gordurosa não Alcoólica , Ratos , Humanos , Animais , Masculino , Ratos Sprague-Dawley , Obesidade/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Dieta Hiperlipídica/efeitos adversos , Aumento de Peso , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Triglicerídeos , Colesterol/metabolismo , Dislipidemias/metabolismo , Ácido Oleico/uso terapêutico
3.
Sci Rep ; 13(1): 17986, 2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37863994

RESUMO

Whilst the presence of 2 subphenotypes among the heterogenous Acute Respiratory Distress Syndrome (ARDS) population is becoming clinically accepted, subphenotype-specific treatment efficacy has yet to be prospectively tested. We investigated anti-inflammatory treatment in different ARDS models in sheep, previously shown similarities to human ARDS subphenotypes, in a preclinical, randomized, blinded study. Thirty anesthetized sheep were studied up to 48 h and randomized into: (a) OA: oleic acid (n = 15) and (b) OA-LPS: oleic acid and subsequent lipopolysaccharide (n = 15) to achieve a PaO2/FiO2 ratio of < 150 mmHg. Then, animals were randomly allocated to receive treatment with methylprednisolone or erythromycin or none. Assessed outcomes were oxygenation, pulmonary mechanics, hemodynamics and survival. All animals reached ARDS. Treatment with methylprednisolone, but not erythromycin, provided the highest therapeutic benefit in Ph2 animals, leading to a significant increase in PaO2/FiO2 ratio by reducing pulmonary edema, dead space ventilation and shunt fraction. Animals treated with methylprednisolone displayed a higher survival up to 48 h than all others. In animals treated with erythromycin, there was no treatment benefit regarding assessed physiological parameters and survival in both phenotypes. Treatment with methylprednisolone improves oxygenation and survival, more so in ovine phenotype 2 which resembles the human hyperinflammatory subphenotype.


Assuntos
Ácido Oleico , Síndrome do Desconforto Respiratório , Animais , Humanos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Eritromicina/uso terapêutico , Metilprednisolona/farmacologia , Metilprednisolona/uso terapêutico , Ácido Oleico/uso terapêutico , Respiração , Ovinos
4.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2687-2699, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37126194

RESUMO

The pharmacology of urolithin C (UroC) on non-alcoholic fatty liver disease (NAFLD) is largely undetermined. We sought to investigate the potential for NAFLD improvement by administration of UroC and the underlying mechanisms. We verified the therapeutic effect of UroC on choline-deficient amino acid-defined high fat diet (CDAHFD) induced NAFLD mice via evaluating NAFLD activity score (NAS), AST, ALT, hepatic phosphorylated AMPK, and 4-hydroxynonenal. Oleic acid-induced AML12 cell was appraised by oil red staining and western blotting to explore the effect and mechanism of UroC in vitro. Transcriptional regulation of UroC was explored by liver RNA sequencing, gut microbiota composition was explored by 16SrRNA sequencing, and colorectal tight junctional proteins were detected by western blotting and immunohistochemistry. The detrimental effects of CDAHFD included the increased liver index, AST, ALT, hepatic 4-hydroxynonenal, impaired intestinal mucosal barrier, and most importantly, pathological damage in liver. Oral administration of UroC largely protected against these harmful alterations. Remarkably, both RNA sequencing and western blotting results indicated an activation in hepatic AMPK signaling pathway which was thought to inhibit ferroptosis response to UroC in vivo, while no change were found in AMPK-ferroptosis axis response to UroC in oleic acid-induced AML12 cells, hinted an indispensable linkage between UroC and hepatic AMPK, presumably the gut-liver axis. Furthermore, UroC could neither alleviate lipid deposition nor inhibit ferroptosis in vitro. The 16SrRNA showed UroC partially counteracted the dysbiosis induced by CDAHFD. Specifically, UroC reversed the elevated proportion of Firmicutes/Bacteroidota and enhanced the level of Parabacteroides goldsteinii and Lactobacillus vaginalis, which played a beneficial role in metabolic disorders. Oral administration of Urolithin C protected against the detrimental impact of CDAHFD via regulating AMPK-ferroptosis axis, maintaining intestinal mucosal barrier and counteracting gut dysbiosis.


Assuntos
Ferroptose , Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Disbiose/metabolismo , Disbiose/patologia , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico , Fígado , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL
5.
Nutrients ; 15(1)2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36615882

RESUMO

In 2010, the Mediterranean diet was recognized by UNESCO as an Intangible Cultural Heritage of Humanity. Olive oil is the most characteristic food of this diet due to its high nutraceutical value. The positive effects of olive oil have often been attributed to its minor components; however, its oleic acid (OA) content (70-80%) is responsible for its many health properties. OA is an effective biomolecule, although the mechanism by which OA mediates beneficial physiological effects is not fully understood. OA influences cell membrane fluidity, receptors, intracellular signaling pathways, and gene expression. OA may directly regulate both the synthesis and activities of antioxidant enzymes. The anti-inflammatory effect may be related to the inhibition of proinflammatory cytokines and the activation of anti-inflammatory ones. The best-characterized mechanism highlights OA as a natural activator of sirtuin 1 (SIRT1). Oleoylethanolamide (OEA), derived from OA, is an endogenous ligand of the peroxisome proliferator-activated receptor alpha (PPARα) nuclear receptor. OEA regulates dietary fat intake and energy homeostasis and has therefore been suggested to be a potential therapeutic agent for the treatment of obesity. OEA has anti-inflammatory and antioxidant effects. The beneficial effects of olive oil may be related to the actions of OEA. New evidence suggests that oleic acid may influence epigenetic mechanisms, opening a new avenue in the exploration of therapies based on these mechanisms. OA can exert beneficial anti-inflammatory effects by regulating microRNA expression. In this review, we examine the cellular reactions and intracellular processes triggered by OA in T cells, macrophages, and neutrophils in order to better understand the immune modulation exerted by OA.


Assuntos
Dieta Mediterrânea , Ácido Oleico , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico , Azeite de Oliva/farmacologia , Ácidos Oleicos/farmacologia , Anti-Inflamatórios/farmacologia
6.
J Nucl Med ; 64(3): 423-429, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36109184

RESUMO

Today, there is a lack of clinically available imaging techniques to detect and quantify specific immune cell populations. Neutrophils are one of the first immune cells at the site of inflammation, and they secrete the serine protease neutrophil elastase (NE), which is crucial in the fight against pathogens. However, the prolonged lifespan of neutrophils increases the risk that patients will develop severe complications, such as acute respiratory distress syndrome (ARDS). Here, we evaluated the novel radiolabeled NE inhibitor 11C-GW457427 in a pig model of ARDS, for detection and quantification of neutrophil activity in the lungs. Methods: ARDS was induced by intravenous administration of oleic acid to 5 farm pigs, and 4 were considered healthy controls. The severity of ARDS was monitored by clinical parameters of lung function and plasma biomarkers. Each pig was studied with 11C-GW457427 and PET/CT, before and after pretreatment with the NE inhibitor GW311616 to determine in vivo binding specificity. PET image data were analyzed as SUVs and correlated with immunohistochemical staining for NE in biopsies. Results: The binding of 11C-GW457427 was increased in pig lungs with induced ARDS (median SUVmean, 1.91; interquartile range [IQR], 1.67-2.55) compared with healthy control pigs (P < 0.05 and P = 0.03, respectively; median SUVmean, 1.04; IQR, 0.66-1.47). The binding was especially strong in lung regions with high levels of NE and ongoing inflammation, as verified by immunohistochemistry. The binding was successfully blocked by pretreatment of an NE inhibitor drug, which demonstrated the in vivo specificity of 11C-GW457427 (P < 0.05 and P = 0.04, respectively; median SUVmean, 0.60; IQR, 0.58-0.77). The binding in neutrophil-rich tissues such as bone marrow (P < 0.05 and P = 0.04, respectively; baseline median SUVmean, 5.01; IQR, 4.48-5.49; block median SUVmean, 1.57; IQR, 0.95-1.85) and spleen (median SUVmean, 2.14; IQR, 1.19-2.36) was also high in all pigs. Conclusion: 11C-GW457427 binds to NE in a porcine model of oleic acid-induced lung inflammation in vivo, with a specific increase in regional lung, bone marrow, and spleen SUV. 11C-GW457427 is a promising tool for localizing, tracking, and quantifying neutrophil-facilitated inflammation in clinical diagnostics and drug development.


Assuntos
Elastase de Leucócito , Síndrome do Desconforto Respiratório , Animais , Suínos , Elastase de Leucócito/uso terapêutico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/efeitos adversos , Ácido Oleico/uso terapêutico , Síndrome do Desconforto Respiratório/diagnóstico por imagem , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/etiologia , Inflamação/complicações , Neutrófilos
7.
J Nat Med ; 77(1): 73-86, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36115008

RESUMO

Excessive intrahepatocellular lipid accumulation or steatosis is caused by abnormal lipid metabolism and a common character of nonalcoholic fatty liver disease (NAFLD), which may progress into cirrhosis and hepatocellular cancer. Andrographolide (Andro) is the primary active ingredient extracted from Andrographis paniculata, showing a protective role against dietary steatosis with the mechanism not fully understood. In this study, we showed that administration of Andro (50, 100, and 200 mg/kg/day for 8 weeks, respectively) attenuated obesity and metabolic syndrome in high-fat diet (HFD)-fed mice with improved glucose tolerance, insulin sensitivity, and reduced hyperinsulinemia, hyperglycemia, and hyperlipidemia. HFD-fed mice presented hepatic steatosis, which was significantly prevented by Andro. In vitro, Andro decreased the intracellular lipid droplets in oleic acid-treated LO2 cells. The selected RT-PCR array revealed a robust expression suppression of the fatty acid transport proteins (FATPs) by Andro treatment. Most importantly, we found that Andro consistently reduced the expression of FATP2 in both the oleic acid-treated LO2 cells and liver tissues of HFD-fed mice. Overexpression of FATP2 abolished the lipid-lowering effect of Andro in oleic acid-treated LO2 cells. Andro treatment also reduced the fatty acid uptake in oleic acid-treated LO2 cells, which was blunted by FATP2 overexpression. Collectively, our findings reveal a novel mechanism underlying the anti-steatosis effect of Andro by suppressing FATP2-mediated fatty acid uptake, suggesting the potential therapeutic application of Andro in the treatment of NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Coenzima A Ligases/metabolismo , Coenzima A Ligases/farmacologia , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Ácidos Graxos/uso terapêutico , Metabolismo dos Lipídeos , Fígado , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico
8.
ACS Chem Neurosci ; 13(8): 1263-1272, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35385250

RESUMO

Parkinson's disease (PD) is the second most common devastating neurodegenerative disorder. Presently used therapies for PD have severe side effects and are limited to only temporary improvement. Therefore, a new therapeutic approach to treat PD urgently needs to be developed. α-Lactalbumin, the most abundant milk protein in camel milk, has been attributed to various medicinal properties. This study intended to investigate the neuroprotective efficacy of the camel α-lactalbumin and oleic acid (CLOA) complex. One mechanism postulated to underlie neuroprotection by the CLOA complex is the induction of silent information regulatory protein (SIRT1). SIRT1 is known to be involved in several pathological and physiological processes, and it has been suggested that SIRT1 plays a protective role in PD. Oxidative stress, inflammation, mitochondrial dysfunction, and apoptosis are involved in PD pathogenesis. Our results revealed that SIRT1 inhibits oxidative stress by maintaining HIF-1α in a deacetylated state. SIRT1 upregulates the expression of FOXO3a and HSF-1, thus inhibiting apoptosis and maintaining the homeostasis of cellular proteins. Increased SIRT1 expression reduces the levels of TNF-α, IL-6, and IL-8, which in turn inhibits neuroinflammation. In addition to SIRT1, the CLOA complex also enhances the expression of survivin and leptin and promotes the survival of neuroblastoma cells. Altogether, our results suggest that the CLOA complex might be a novel therapeutic molecule that could ameliorate neuronal cell damage in PD.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Camelus/metabolismo , Lactalbumina/metabolismo , Lactalbumina/farmacologia , Lactalbumina/uso terapêutico , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico , Estresse Oxidativo , Doença de Parkinson/tratamento farmacológico , Rotenona , Sirtuína 1/metabolismo , Sirtuína 1/farmacologia , Sirtuína 1/uso terapêutico
9.
Nutrients ; 13(8)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34444748

RESUMO

Obesity is currently a major epidemic in the developed world. However, we lack a wide range of effective pharmacological treatments and therapies against obesity, and those approved are not devoid of adverse effects. Dietary components such as palmitoleic acid have been proposed to improve metabolic disbalance in obesity, although the mechanisms involved are not well understood. Both palmitoleic acid (POA) and oleic acid (OA) can be transformed in N-acylethanolamines (NAEs), mediating the effects of dietary POA and OA. To test this hypothesis, here, we study the effects on food intake and body weight gain of palmitoleylethanolamide (POEA) and the OA-derived NAE analogue, oleoylethanolamide (OEA), in Sprague-Dawley rats with a hypercaloric cafeteria diet (HFD). Plasma biochemical metabolites, inflammatory mediators, and lipogenesis-associated liver protein expression were also measured. The results indicate that POEA is able to improve health status in diet-induced obesity, decreasing weight, liver steatosis, inflammation, and dyslipemia. The action of POEA was found to be almost identical to that of OEA, which is an activator of the nuclear peroxisome proliferator receptor alpha (PPARα), and it is structurally related to POEA. These results suggest that the dietary administration of either POA or POEA might be considered as nutritional intervention as complementary treatment for complicated obesity in humans.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Ácidos Graxos Monoinsaturados/uso terapêutico , Obesidade/tratamento farmacológico , Ácidos Oleicos/farmacologia , Ácidos Oleicos/uso terapêutico , Animais , Peso Corporal , Citocinas , Dieta , Endocanabinoides , Etanolaminas , Ácidos Graxos , Fígado Gorduroso/metabolismo , Humanos , Resistência à Insulina , Lipogênese , Fígado/metabolismo , Masculino , Ácido Oleico/uso terapêutico , Ratos , Ratos Sprague-Dawley
11.
Nutrients ; 12(6)2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570937

RESUMO

Multifactorial pathogenesis of non-alcoholic steatohepatitis (NASH) disease, a wide-spread liver pathology associated with metabolic alterations triggered by hepatic steatosis, should be hit by multitarget therapeutics. We tested a multicomponent food supplement mixture (AP-NHm), whose components have anti-dislipidemic, antioxidant and anti-inflammatory effects, on in vitro and in vivo models of NASH. In vitro, hepatic cells cultures were treated for 24 h with 0.5 mM oleic acid (OA): in the co-treatment set cells were co-treated with AP-NH mixtures (AP-NHm, 1:3:10 ratio) and in the post-injury set AP-NHm was added for 48 h after OA damage. In vivo, C57BL/6 mice were fed with high-fat diet (HFD) for 12 weeks, inducing NASH at 7th week, and treated with AP-NHm at two dosages (1:3 ratio) in co-treatment or post-injury protocols, while a control group was fed with a standard diet. In in vitro co-treatment protocol, alterations of redox balance, proinflammatory cytokines release and glucose uptake were restored in a dose-dependent manner, at highest dosages also in post-injury regimen. In both regimens, pathologic dyslipidemias were also ameliorated by AP-NHm. In vivo, high-dose-AP-NHm-co-treated-HFD mice dose-dependently gained less body weight, were protected from dyslipidemia, and showed a lower liver weight. Dose-dependently, AP-NHm treatment lowered hepatic LDL, HDL, triglycerides levels and oxidative damage; co-treatment regimen was anti-inflammatory, reducing TNF-α and IL-8 levels. Hepatic lipidic infiltration significantly decreased in co-treated and post-injury-AP-NHm-HFD animals. The multitarget approach with AP-NHm was effective in preventing and reducing NASH-related pathologic features, warranting for the clinical development of this compound.


Assuntos
Ácido Clorogênico/uso terapêutico , Colina/uso terapêutico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ácido Oleico/uso terapêutico , Extratos Vegetais/uso terapêutico , alfa-Tocoferol/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Células Cultivadas , Café , Suplementos Nutricionais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tanacetum parthenium
12.
J Vis Exp ; (159)2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32538907

RESUMO

The treatment of ARDS continues to pose major challenges for intensive care physicians in the 21st century with mortality rates still reaching up to 50% in severe cases. Further research efforts are needed to better understand the complex pathophysiology of this disease. There are different well-established animal models to induce acute lung injury but none has been able to adequately mimic the complex pathomechanisms of ARDS. The most crucial factor for the development of this condition is the damage to the alveolar capillary unit. The combination of two well-established lung injury models allow us to mimic in more detail the underlying pathomechanism. Bronchoalveolar lavage (BAL) leads to surfactant depletion as well as alveolar collapse. The repeated instillation of fluid volumes causes subsequent hypoxemia. Surfactant depletion is a key factor of ARDS in humans. BAL is often combined with other lung injury approaches, but not with a second hit followed by oleic acid injection (OAI) yet. Oleic acid injection leads to severely impaired gas exchange, a deterioration of lung mechanics and disruption of the alveolo-capillary barrier. The OAI mimics most of the expected effects of ARDS consisting of extended inflammation of lung tissue with an increase of alveolar leakage and gas exchange impairment. A disadvantage of the combination of different models is the difficulty to determine the influence to the lung injury caused by BAL alone, OAI alone or both together. The model presented in this report represents the combination of BAL and OAI as a new double-hit lung injury model. This new model is easy to implement and an alternative to study different therapeutic approaches in ARDS in the future.


Assuntos
Lavagem Broncoalveolar/métodos , Injeções/métodos , Ácido Oleico/uso terapêutico , Síndrome do Desconforto Respiratório/diagnóstico , Animais , Modelos Animais de Doenças , Humanos , Ácido Oleico/farmacologia , Síndrome do Desconforto Respiratório/fisiopatologia , Suínos
13.
J Endocrinol ; 246(2): 175-187, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32485676

RESUMO

Maternal diabetes impairs fetal development and increases the risk of metabolic diseases in the offspring. Previously, we demonstrated that maternal dietary supplementation with 6% of olive oil prevents diabetes-induced embryo and fetal defects, in part, through the activation of peroxisome proliferator-activated receptors (PPARs). In this study, we examined the effects of this diet on neonatal and adult pancreatic development in male and female offspring of mothers affected with pre-gestational diabetes. A mild diabetic model was developed by injecting neonatal rats with streptozotocin (90 mg/kg). During pregnancy, these dams were fed a chow diet supplemented or not with 6% olive oil. Offspring pancreata was examined at day 2 and 5 months of age by immunohistochemistry followed by morphometric analysis to determine number of islets, α and ß cell clusters and ß-cell mass. At 5 months, male offspring of diabetic mothers had reduced ß-cell mass that was prevented by maternal supplementation with olive oil. PPARα and PPARγ were localized mainly in α cells and PPARß/δ in both α and ß cells. Although Pparß/δ and Pparγ RNA expression showed reduction in 5-month-old male offspring of diabetic rats, Pparß/δ expression returned to control levels after olive-oil supplementation. Interestingly, in vitro exposure to oleic acid (major component of olive oil) and natural PPAR agonists such as LTB4, CPC and 15dPGJ2 also significantly increased expression of all Ppars in αTC1-6 cells. However, only oleic acid and 15dPGJ2 increased insulin and Pdx-1 expression in INS-1E cells suggesting a protective role in ß-cells. Olive oil may be considered a dietary supplement to improve islet function in offspring of affected mothers with pre-gestational diabetes.


Assuntos
Diabetes Mellitus Experimental/dietoterapia , Diabetes Gestacional/dietoterapia , Azeite de Oliva/uso terapêutico , Animais , Suplementos Nutricionais , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Leucotrieno B4/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Ácido Oleico/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Gravidez , Ratos , Estreptozocina/toxicidade , Transativadores/genética , Transativadores/metabolismo
14.
Nutrients ; 12(6)2020 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-32466599

RESUMO

The increasing access to antihypertensive medications has improved longevity and quality of life in hypertensive patients. Nevertheless, hypertension still remains a major risk factor for stroke and myocardial infarction, suggesting the need to implement management of pre- and hypertensive patients. In addition to antihypertensive medications, lifestyle changes, including healthier dietary patterns, such as the Dietary Approaches to Stop Hypertension (DASH) and the Mediterranean diet, have been shown to favorably affect blood pressure and are now recommended as integrative tools in hypertension management. An analysis of the effects of nutritional components of the Mediterranean diet(s) on blood pressure has therefore become mandatory. After a literature review of the impact of Mediterranean diet(s) on cardiovascular risk factors, we here analyze the effects of olive oil and its major components on blood pressure in healthy and cardiovascular disease individuals and examine underlying mechanisms of action. Both experimental and human studies agree in showing anti-hypertensive effects of olive oil. We conclude that due to its high oleic acid and antioxidant polyphenol content, the consumption of olive oil may be advised as the optimal fat choice in the management protocols for hypertension in both healthy and cardiovascular disease patients.


Assuntos
Anti-Hipertensivos/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Hipertensão/epidemiologia , Azeite de Oliva/uso terapêutico , Antioxidantes/uso terapêutico , Bases de Dados Factuais , Dieta Mediterrânea , Humanos , Ácido Oleico/uso terapêutico , Polifenóis , Qualidade de Vida , Fatores de Risco , Acidente Vascular Cerebral/tratamento farmacológico
15.
J Ethnopharmacol ; 254: 112708, 2020 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-32142866

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Popularly used in India and sub-Hymalaian region, Moringa oleifera (Moringaceae) is associated with healing properties demonstrated in its use as treatment of acute and chronic skin diseases. Our study aimed at investigating the effects of M. oleifera seed oil (MOSO) in animal models for inflammatory and hyperproliferative skin conditions. MATERIALS AND METHODS: MOSO was analyzed using gas chromatography/mass spectrometry. The anti-inflammatory and anti-hyperproliferative effects of treatment with either MOSO or oleic acid (OA), its main constituent, was evaluated. Acute and chronic inflammation was induced by applying 12-O-Tetradecanoylphorbol-13-acetate (TPA) and acute inflammation with either Arachidonic Acid (AA) or Phenol onto the ear of Swiss mice. Systemic activity and the influence of glucocorticoid receptors (GC) was also evaluated. RESULTS: Topical application of MOSO and OA inhibited ear edema caused by TPA, and Phenol. Only MOSO inhibited ear edema induced by AA. Neutrophil migration was also inhibited by treatment with MOSO. Topical application of MOSO, but not OA, significantly reduced chronic skin inflammation and epidermal hypertrophy induced by multiple TPA applications. Pre-treatment with GC antagonist mifepristone reversed the anti-inflammatory effect of MOSO and OA on the TPA model. Repeated administration of MOSO show a similar effect to dexamethasone on thymus weight, though MOSO did not present any influence on skin thickness, as well as in the weight of the spleen, adrenal gland and lymph node. CONCLUSION: The results suggest that MOSO is effective as a treatment for skin diseases that rely on keratinocyte hyperproliferation. OA is also effective in acute inflammation. Both MOSO and OA depend on GC activation for anti-inflammatory effect but do not exhibit the same adverse effects seen in topical treatment with dexamethasone. We hereby evidence the use of MOSO as a topical anti-inflammatory agent in inflammatory skin diseases, thus, expanding its therapeutic potential.


Assuntos
Anti-Inflamatórios/uso terapêutico , Dermatite de Contato/tratamento farmacológico , Moringa oleifera , Ácido Oleico/uso terapêutico , Óleos de Plantas/uso terapêutico , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Atrofia/tratamento farmacológico , Atrofia/metabolismo , Proliferação de Células/efeitos dos fármacos , Dermatite de Contato/metabolismo , Edema/tratamento farmacológico , Edema/metabolismo , Feminino , Irritantes , Queratinócitos/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Camundongos , Receptores de Glucocorticoides/metabolismo , Sementes , Pele/efeitos dos fármacos , Pele/patologia , Baço/efeitos dos fármacos , Acetato de Tetradecanoilforbol , Timo/efeitos dos fármacos
16.
Lipids Health Dis ; 19(1): 28, 2020 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-32093685

RESUMO

BACKGROUND: Diets based on meat products are not recommended in the case of ulcerative colitis (UC). The objective here is to test if some traditional cured meat products, as acorn-fed ham (high levels of oleic acid), may be useful for controlling inflammatory diseases as UC in animal models, which could represent a new dietary complementary intervention in the prevention of this inflammatory disease in humans. METHODS: Two rat cohorts have been used: conventional vegetable rat feed and acorn-fed ham. UC was induced with DSS in drinking water ad libitum for 1 week. Short-chain fatty acids (SCFAs) and 16S rRNA metagenomics from bacterial populations were analyzed in cecum samples. Colon samples were analyzed for histological parameters. RESULTS: Acorn-fed ham diet induced changes in gut microbiota composition, with pronounced enrichments in anti-inflammatory bacterial genera (Alistipes, Blautia, Dorea, Parabacteroides). The animals with this diet showed a strong reduction in most parameters associated to ulcerative colitis: disease activity index, macroscopic score of colitis, epitelium alteration in colon mucosa, inflammatory cell density in colon, myeloperoxidase titers in colon, proinflammatory cytokines (IL-17, IFN-γ). Also, acorn-fed ham diet animals showed increased total antioxidant activity an oleic acid levels in plasma, as well as higher short-chain fatty acid concentrations in cecum (isobutyric, isovaleric and valeric). CONCLUSIONS: In the acorn-fed ham cohort, as a result of the dietary intake of oleic acid and low intake of omega-6 fatty acids, a strong preventive effect against UC symptoms was observed.


Assuntos
Ração Animal , Anti-Inflamatórios/uso terapêutico , Colite Ulcerativa/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Ácido Oleico/uso terapêutico , Animais , Anti-Inflamatórios/química , Colite Ulcerativa/microbiologia , Colo/microbiologia , Citocinas/sangue , Modelos Animais de Doenças , Cromatografia Gasosa-Espectrometria de Massas , Mucosa Intestinal/microbiologia , Masculino , Ácido Oleico/química , Filogenia , RNA Ribossômico 16S/metabolismo , Ratos , Ratos Endogâmicos F344
17.
Hypertension ; 75(3): 680-692, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31928110

RESUMO

Plasma metabolic profiles were compared between patients with hypertension with and without left ventricular hypertrophy and significantly decreased oleic acid (OA) levels were observed in the peripheral blood of patients with hypertension with left ventricular hypertrophy. We sought to determine the effect and underlying mechanisms of OA on cardiac remodeling. In vitro studies with isolated neonatal mouse cardiomyocytes and cardiac fibroblasts revealed that OA significantly attenuated Ang II (angiotensin II)-induced cardiomyocyte growth and cardiac fibroblast collagen expression. In vivo, cardiac function, hypertrophic growth of cardiomyocytes, and fibrosis were analyzed after an Ang II (1000 ng/kg/minute) pump was implanted for 14 days. We found that OA could significantly prevent Ang II-induced cardiac remodeling in mice. RNA sequencing served as a gene expression roadmap highlighting gene expression changes in the hearts of Ang II-induced mice and OA-treated mice. The results revealed that FGF23 (fibroblast growth factor 23) expression was significantly upregulated in mouse hearts in response to Ang II infusion, which was significantly suppressed in the hearts of OA-treated mice. Furthermore, overexpression of FGF23 in the heart by injection of an AAV-9 vector aggravated Ang II-induced cardiac remodeling and impaired the protective effect of OA on cardiac remodeling. Further study found that OA could suppress Ang II-induced FGF23 expression by inhibiting the translocation of Nurr1 (nuclear receptor-related 1 protein) from the cytoplasm to the nucleus. Our findings suggest a novel role of OA in preventing Ang II-induced cardiac remodeling via suppression of FGF23 expression.


Assuntos
Angiotensina II/farmacologia , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Hipertrofia Ventricular Esquerda/sangue , Ácido Oleico/fisiologia , Remodelação Ventricular/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Colágeno/biossíntese , Dependovirus/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Ontologia Genética , Vetores Genéticos , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Ácido Oleico/sangue , Ácido Oleico/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Organismos Livres de Patógenos Específicos , Remodelação Ventricular/efeitos dos fármacos
18.
Int Wound J ; 17(2): 485-490, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31876118

RESUMO

Wound healing, especially diabetic ones, is a relevant clinical problem, so it is not surprising that surgical procedures are often needed. To overcome invasive procedures, several strategies with drugs or natural compound are used. Recently, in an experimental study, we described an increase in keratinocyte proliferation after their exposition to quercetin plus oleic acid. In the present clinical study, we evaluated both the clinical efficacy and the safety of nano-hydrogel embedded with quercetin and oleic acid in the treatment of lower limb skin wound in patients with diabetes mellitus (DM). Fifty-six DM patients (28 men and 28 women, mean age 61.7 ± 9.2 years) unsuccessfully treated with mechanical compression were enrolled and randomised to receive an add on treatment with hyaluronic acid (0.2%) or nano-hydrogel embedded with quercetin and oleic acid. The treatment with nano-hydrogel embedded with quercetin and oleic acid significantly (P < .01) reduced the wound healing time, in comparison to hyaluronic acid (0.2%) without developing of adverse drug reactions, suggesting that this formulation could be used in the management of wound healing even if other clinical trials must be performed in order to validate this observation.


Assuntos
Pé Diabético/terapia , Hidrogéis/uso terapêutico , Ácido Oleico/uso terapêutico , Quercetina/uso terapêutico , Cicatrização , Idoso , Antioxidantes/uso terapêutico , Combinação de Medicamentos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Estudos Prospectivos
19.
Sci Rep ; 9(1): 10732, 2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31341184

RESUMO

Oleic acid (OA) is released from brain phospholipids after cerebral ischaemia; however, its role in ischaemic injury remains unknown. We hypothesised that OA has neuroprotective effects after cerebral ischaemia, which may be exerted through peroxisome proliferator-activated receptor gamma (PPAR-γ) activation, since OA is an endogenous ligand of PPAR-γ. The effects of OA administration were evaluated in rodent models of middle cerebral artery occlusion (MCAO), photothrombosis, and four-vessel occlusion (4-VO). We determined the time window of therapeutic opportunity and examined the ability of the PPAR-γ antagonist GW9662 to reverse OA's protective effects after MCAO. We found that OA administration decreased the MCAO-induced infarct volume and functional deficits, photothrombosis-induced infarct volume, and 4-VO-induced hippocampal neuronal death. Additionally, OA was highly efficacious when administered up to 3 h after MCAO. Pre-treatment with GW9662 abolished the inhibitory effects of OA on the infarct volume and immunoreactivity of key inflammatory mediators in the ischaemic cortex. Our results indicate that OA has neuroprotective effects against transient and permanent focal cerebral ischaemia, as well as global cerebral ischaemia. It may have therapeutic value for the ischaemic stroke treatment with a clinically feasible therapeutic window. The OA-mediated neuroprotection might be attributable to its anti-inflammatory actions through PPAR-γ activation.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Ácido Oleico/uso terapêutico , Animais , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Ratos Wistar
20.
Bioorg Chem ; 84: 478-492, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30579158

RESUMO

A new series of octadec-9-enoic acid schiff base entities (S1-S30) were designed and synthesized targeting peroxisome proliferator activated receptor-gamma for agonist action. Molinspiration software (online) was used to estimate drug like molecular properties of the metabolites. Docking disquisition on co-crystallized protein of PPAR-γ (PDB ID 1FM9) was carried out which showed S21, S10 and S7 as best situated in the vital sites of receptor having docking scores -9.19, -8.68 and -8.64 respectively. Free binding energy measured using model of Maestro 9.0 and was in range of from -40.01 and -80.54 kcal/mol, significant when compared with pioglitazone (-51.58 Kcal/mol). Seven best docked derivatives were assessed for in-vivo oral glucose tolerance on normal rats and anti-hyperglycaemic activity by streptozotocin induced diabetes model. S21 unveiled to be the best measured analogue among all the synthesized entities. Encouraging outcomes motivates fatty acids for further development of more effective and safer compounds.


Assuntos
Hipoglicemiantes/síntese química , Simulação de Acoplamento Molecular , Ácido Oleico/química , Bases de Schiff/química , Animais , Sítios de Ligação , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Desenho de Fármacos , Feminino , Teste de Tolerância a Glucose , Meia-Vida , Humanos , Hipoglicemiantes/metabolismo , Hipoglicemiantes/uso terapêutico , Masculino , Ácido Oleico/metabolismo , Ácido Oleico/uso terapêutico , PPAR gama/química , PPAR gama/metabolismo , Ratos , Ratos Wistar , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...