Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Thyroid Res ; 2021: 7664641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34567510

RESUMO

Graves's disease and thyroiditis induce hyperthyroidism, the causes of which remain unclear, although they are involved with genetic and environmental factors. We aimed to evaluate polygenetic variants for hyperthyroidism risk and their interaction with metabolic parameters and nutritional intakes in an urban hospital-based cohort. A genome-wide association study (GWAS) of participants with (cases; n = 842) and without (controls, n = 38,799) hyperthyroidism was used to identify and select genetic variants. In clinical and lifestyle interaction with PRS, 312 participants cured of hyperthyroidism were excluded. Single nucleotide polymorphisms (SNPs) associated with gene-gene interactions were selected by hyperthyroidism generalized multifactor dimensionality reduction. Polygenic risk scores (PRSs) were generated by summing the numbers of selected SNP risk alleles. The best gene-gene interaction model included tumor-necrosis factor (TNF)_rs1800610, mucin 22 (MUC22)_rs1304322089, tribbles pseudokinase 2 (TRIB2)_rs1881145, cytotoxic T-lymphocyte-associated antigen 4 (CTLA4)_rs231775, lipoma-preferred partner (LPP)_rs6780858, and human leukocyte antigen (HLA)-J_ rs767861647. The PRS of the best model was positively associated with hyperthyroidism risk by 1.939-fold (1.317-2.854) after adjusting for covariates. PRSs interacted with age, metabolic syndrome, and dietary inflammatory index (DII), while hyperthyroidism risk interacted with energy, calcium, seaweed, milk, and coffee intake (P < 0.05). The PRS impact on hyperthyroidism risk was observed in younger (<55 years) participants and adults without metabolic syndrome. PRSs were positively associated with hyperthyroidism risk in participants with low dietary intakes of energy (OR = 2.74), calcium (OR = 2.84), seaweed (OR = 3.43), milk (OR = 2.91), coffee (OR = 2.44), and DII (OR = 3.45). In conclusion, adults with high PRS involved in inflammation and immunity had a high hyperthyroidism risk exacerbated under low intakes of energy, calcium, seaweed, milk, or coffee. These results can be applied to personalized nutrition in a clinical setting.

2.
Clin Exp Nephrol ; 24(11): 1015-1024, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32700113

RESUMO

BACKGROUND: Intake of Na-to-K ratio (I-Na/K), urinary Na-to-K ratio (U-Na/K), and estimated glomerular filtration rate (GFR) have been reported to be risk factors of metabolic syndrome (MetS), but results are inconsistent. We examined the hypothesis that U-Na/K, GFR, and a preference for salty foods are associated with MetS risk and the hypothesis in 8540 adults aged over 40 years without chronic kidney disease. METHODS: Participants were categorized using a U-Na/K of < 2.1 (low-U-Na/k) and a GFR of < 60 mL/min (low-GFR). A GFR of 60-90 mL/min was considered as a normal level, since it is a normal or marginal disease state. Correlations and associations were determined using Pearson's correlation coefficients and logistic regression analysis after adjusting for covariates related to MetS. RESULTS: U-Na/K, but not I-Na/K, was positively correlated with blood pressure (r2 = 0.20, P < 0.0001). The GFR was negatively correlated with age, gender, HOMA-B, and MetS (r2 = - 0.14 to - 0.595, P < 0.0001), and positively correlated with education, current smoking, and alcohol intake (r2 = 0.21 to 0.40, P < 0.0001). MetS risk had a positive association with the following combinations with low-U-Na/K + low-GFR, high-U-Na/K + high-GFR, and high-U-Na/K +low-GFR by 1.830-, 3.182-, and 3.696-fold, respectively, as compared with low-U-Na/K + high-GFR. Risks of the MetS components (abdominal obesity, hypertriglyceridemia, hypo-HDL-cholesterolemia, hypertension, and hyperglycemia) were similarly associated with U-Na/K and GFR, though hypertension had the strongest association. Hypertension risk had positive associations with low-U-Na/K + low-GFR, high-U-Na/K + high-GFR, and high-U-Na/K + low-GFR by 1.526-, 14.06-, and 7.079-fold, respectively, as compared with low-U-Na/K + high-GFR. CONCLUSION: MetS risk was found to be associated with U-Na/K and GFR regardless of I-Na/K. Women need to maintain a high GFR to reduce the MetS risk, especially the risk of hypertension.


Assuntos
Taxa de Filtração Glomerular , Síndrome Metabólica/epidemiologia , Potássio na Dieta , Potássio/urina , Sódio na Dieta , Sódio/urina , Adulto , Fatores Etários , Consumo de Bebidas Alcoólicas/epidemiologia , Pressão Sanguínea , Escolaridade , Feminino , Humanos , Hipercolesterolemia/epidemiologia , Hiperglicemia/epidemiologia , Hipertensão/epidemiologia , Hipertrigliceridemia/epidemiologia , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Obesidade/epidemiologia , Fatores de Risco , Fatores Sexuais , Fumar/epidemiologia
3.
Food Sci Nutr ; 8(7): 3492-3503, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32724612

RESUMO

We hypothesized that subjects with genetic variants that increase sweet taste preference would consume more sucrose-containing foods and have altered energy and glucose metabolisms, which would have interactions with lifestyles. Korean genome and epidemiology study (KoGES) was conducted to determine genetic variants and lifestyles including nutrient intakes by the Korean Center for Disease and Control during 2004-2013. Subjects were 8,842 adults aged 40-69 years in Ansan/Ansung cohorts in Korea. The associations between genetic risk scores(GRS) selected for influencing higher sweet preference and energy and glucose metabolism were examined using logistic regression after adjusting for covariates. GRS included 8 SNPs, TAS1R2_rs61761364, SLC2A5_rs11121306, SLC2A7_ rs769902, SLC2A5_rs765618, TRPM5_rs1965606, TRPV1_rs224495, TRPV1_ rs8065080, and TRPV1_rs8078502. Sweet taste preference was higher by 1.30-folds in high GRS than in low GRS (p < .0001). Consistent with sweet taste preference, carriers with high GRS had a higher intake of sucrose-containing foods by 1.25 (1.08-1.46)-fold than those with low GRS after adjusting age, gender, BMI, and energy intake. However, glucose intolerance risk was rather lower by 0.861 (0.76-0.98)-fold in high GRS than low GRS (p < .05). GRS tended to interact with mental stress to affect sucrose intake (p = .048). Only in low mental stress levels, sucrose-containing food intake was higher in high GRS than low GRS. There was an interaction of GRS with physical activity to influence glucose intolerance. Serum glucose concentrations were lower by 0.808-folds in high GRS than low GRS only in a high physical activity state. In conclusion, adults with genetically high sweet taste preference had a positive association with high sucrose-containing food intakes and improved glucose tolerance. The genetic impact on sweetness preference was associated with offset by high mental stress and lack of physical activity.

4.
Nutrition ; 79-80: 110859, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32619791

RESUMO

OBJECTIVES: Osteoporosis is associated with genetic and environmental factors. The aim of this article was to determine how the polygenic risk scores (PRS) of genetic variants that affect osteoporosis and its related signaling interact with the lifestyle of middle-aged adults. METHODS: The study examined 8845 participants from Ansan/Ansung cohorts. Osteoporosis was defined as a T-score of bone mineral density ≤-2.5 in either the wrist or tibia; 1136 participants had osteoporosis. Genome-wide association studies of individuals 40 to 65 y of age were conducted and the best gene-gene interactions from the genetic variants related to osteoporosis were selected and explored using the generalized multifactor dimensionality reduction method. PRS for the best model (PRSBM) was calculated by weighted PRS that was divided into low, medium, and high groups. RESULTS: The model that contributed the most influence on osteoporosis risk with gene-gene interactions included AKAP11_rs238340, KCNMA1_ rs628948, PUM1_rs7529390, SPTBN1_ rs6752877, and EPDR1_rs2722298. The risk for osteoporosis in the tibia was elevated by 1.71-fold in the high PRSBM group compared with the low PRSBM group. Energy and nutrient intake did not have any interaction with PRSBM and thus did not influence risk for osteoporosis. However, interestingly, only coffee and caffeine intake did interact with PRSBM and affected risk for osteoporosis. In patients with low coffee (<3 cup/wk) and caffeine(<60 mg/d) consumption, osteoporosis risk was higher in the high PRSBM group than the low PRSBM group by 2.27- and 2.29-fold, respectively. In the low coffee intake group, bone mineral density in the high PRSBM group was significantly higher than in the low PRSBM arm. CONCLUSIONS: Carriers with high PRSBM increased susceptibility to osteoporosis, especially in low coffee and caffeine intake. The results can be applied to personalized nutrition for lowering the risk for osteoporosis.


Assuntos
Estudo de Associação Genômica Ampla , Osteoporose , Proteínas de Ancoragem à Quinase A , Adulto , Densidade Óssea/genética , Humanos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Estilo de Vida , Pessoa de Meia-Idade , Proteínas de Neoplasias , Proteínas do Tecido Nervoso , Osteoporose/epidemiologia , Osteoporose/genética , Proteínas de Ligação a RNA , Fatores de Risco , Espectrina
5.
Br J Nutr ; 122(8): 919-927, 2019 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-31544728

RESUMO

Metabolic syndrome (MetS) risk is influenced by genetic and environmental factors. The present study explored genetic risk scores (GRS) of genetic variants that influence the MetS and the effect of interactions between GRS and nutrient intake on MetS risk. The genetic variants that influence MetS risk were selected by genome-wide association study after adjusting for age, sex, area of residence and BMI in 8840 middle-aged adults. GRS were calculated by summing the risk alleles of the selected SNP and divided into low (0-1), medium (2-3) and high (4-7) risk groups, and the relationships between the MetS and GRS were determined by logistic regression after adjusting covariates involved in MetS risk. We also analysed the interaction between GRS and lifestyles. Four genetic variants (APOA5_rs651821, EFCAB4B_rs4766165, ZNF259_rs2160669 and APOBEC1_rs10845640) were selected because they increased MetS risk after adjusting for covariates. Individuals with medium-GRS and high-GRS alleles had a higher MetS risk by 1·48- and 2·23-fold, respectively, compared with those with low-GRS after adjusting for covariates. The increase in MetS risk was mainly related to serum TAG and HDL-cholesterol concentrations. The GRS had an interaction with carbohydrate (CHO) and Na intakes and daily physical activities for MetS risk. In conclusion, Asian middle-aged adults with high-GRS alleles were at increased MetS risk mainly due to dyslipidaemia. High daily physical activity (≥1 h moderate activity per d) reduced the MetS risk but a low-CHO diet (<65 % of total energy intake) increased the risk in carriers with high-GRS alleles. Low Na intake (<1·6 g Na intake/4 MJ) did not decrease its risk.


Assuntos
Carboidratos da Dieta/efeitos adversos , Ingestão de Energia/genética , Exercício Físico , Síndrome Metabólica/genética , Sódio na Dieta/efeitos adversos , Adulto , Idoso , Alelos , Povo Asiático/genética , Dieta/efeitos adversos , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Estilo de Vida , Metabolismo dos Lipídeos/genética , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , República da Coreia , Fatores de Risco
6.
Exp Mol Med ; 51(5): 1-10, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113929

RESUMO

Adipose tissue inflammation is a reproducible feature of obesity and obesity-linked insulin resistance. Although sirtuin 6 (Sirt6) deficiency has previously been implicated in diet-induced obesity and systemic insulin resistance, the adipocyte-specific role of Sirt6 in the regulation of adipose tissue inflammation and systemic metabolic dysfunction in mice fed normal chow and in humans remains elusive. Here, using Adipoq-Cre-mediated adipocyte-specific Sirt6 knockout (aS6KO) mice, we explored whether adipocyte Sirt6 inhibits adipose tissue inflammation and its underlying mechanism. aS6KO mice fed normal chow gained more body weight and fat mass than wild-type mice and exhibited glucose intolerance and systemic insulin resistance. Measurement of plasma and tissue cytokines and flow cytometric analysis of adipose stromal vascular cells indicated a decrease in alternatively activated M2 macrophages in the adipose tissue of aS6KO mice. Mechanistically, Sirt6 regulated the expression of the canonical type 2 cytokine IL-4 by adipocytes in a cell autonomous manner, which in turn affects M2 macrophage polarization. Consistent with animal experimental data, the degree of obesity and insulin resistance demonstrated by the body mass index, fasting blood glucose and HbA1c correlated negatively with the expression of Sirt6 in human visceral fat tissues. Collectively, these results suggest that adipocyte Sirt6 regulates body weight gain and insulin sensitivity independent of diet, and the increased IL-4 production by Sirt6 and resultant M2 polarization of adipose tissue macrophages may attenuate proinflammatory responses in adipose tissue.


Assuntos
Resistência à Insulina/imunologia , Interleucina-4/imunologia , Macrófagos/imunologia , Sirtuínas/imunologia , Tecido Adiposo/imunologia , Adiposidade , Animais , Células Cultivadas , Humanos , Inflamação/imunologia , Macrófagos/citologia , Camundongos , Obesidade/imunologia
7.
Int J Mol Sci ; 19(1)2018 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-29342124

RESUMO

Targeting energy expenditure offers a strategy for treating obesity more effectively and safely. In previous studies, we found that the root of Atractylodes macrocephala Koidzumi (Atractylodis Rhizoma Alba, ARA) increased energy metabolism in C2C12 cells. Here, we investigated the effects of ARA on obesity and glucose intolerance by examining energy metabolism in skeletal muscle and brown fat in high-fat diet (HFD) induced obese mice. ARA decreased body weight gain, hepatic lipid levels and serum total cholesterol levels, but did not modify food intake. Fasting serum glucose, serum insulin levels and glucose intolerance were all improved in ARA treated mice. Furthermore, ARA increased peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) expression, and the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in skeletal muscle tissues, and also prevented skeletal muscle atrophy. In addition, the numbers of brown adipocytes and the expressions of PGC1α and uncoupling protein 1 (UCP1) were elevated in the brown adipose tissues of ARA treated mice. Our results show that ARA can prevent diet-induced obesity and glucose intolerance in C5BL/6 mice and suggests that the mechanism responsible is related to the promotion of energy metabolism in skeletal muscle and brown adipose tissues.


Assuntos
Atractylodes/química , Metabolismo Energético/efeitos dos fármacos , Intolerância à Glucose/metabolismo , Obesidade/metabolismo , Extratos Vegetais/farmacologia , Raízes de Plantas/química , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Modelos Animais de Doenças , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Obesidade/etiologia , Obesidade/prevenção & controle , Extratos Vegetais/química
8.
Biol Pharm Bull ; 40(3): 339-344, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28250276

RESUMO

Targeting energy expenditure provides a potential alternative strategy for achieving energy balance to combat obesity and the development of type 2 diabetes mellitus (T2DM). In the present study, we investigated whether atractylenolide III (AIII) regulates energy metabolism in skeletal muscle cells. Differentiated C2C12 myotubes were treated with AIII (10, 20, or 50 µM) or metformin (2.5 mM) for indicated times. The levels of glucose uptake, the expressions of key mitochondrial biogenesis-related factors and their target genes were measured in C2C12 myotubes. AIII significantly increased the glucose uptake levels, and significantly increased the expressions of peroxisome proliferator-activated receptor coactivator-1α (PGC1α) and mitochondrial biogenesis-related markers, such as, nuclear respiratory factor-1 (NRF-1), and mitochondrial transcription factor A (TFAM) and mitochondrial mass and total ATP contents. In addition, AIII significantly increased the phosphorylation of AMP-activated protein kinase (AMPK) and the expression of sirtuin1 (SIRT1). These results suggest that AIII may have beneficial effects on obesity and T2DM by improving energy metabolism in skeletal muscle.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético/efeitos dos fármacos , Lactonas/farmacologia , Músculo Esquelético/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Extratos Vegetais/farmacologia , Sesquiterpenos/farmacologia , Sirtuína 1/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Atractylodes/química , Glicemia/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/sangue , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Grupo de Alta Mobilidade/sangue , Camundongos , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fator 1 Nuclear Respiratório/sangue , Obesidade/metabolismo , Fosforilação
9.
Nutrients ; 9(1)2017 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-28054981

RESUMO

Radix Pueraria lobata (RP) has been reported to prevent obesity and improve glucose metabolism; however, the mechanism responsible for these effects has not been elucidated. The mechanism underlying anti-obesity effect of RP was investigated in high-fat diet (HFD) induced obese mice and skeletal muscle cells (C2C12). Five-week-old C5BL/6 mice were fed a HFD containing or not containing RP (100 or 300 mg/kg) or metformin (250 mg/kg) for 16 weeks. RP reduced body weight gain, lipid accumulation in liver, and adipocyte and blood lipid levels. In addition, RP dose-dependently improved hyperglycemia, insulinemia, and glucose tolerance, and prevented the skeletal muscle atrophy induced by HFD. Furthermore, RP increased the peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) expression and phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in skeletal muscle tissues. RP and its main component, puerarin, increased mitochondrial biogenesis and myotube hypertrophy in C2C12 cells. The present study demonstrates that RP can prevent diet-induced obesity, glucose tolerance, and skeletal muscle atrophy in mouse models of obesity. The mechanism responsible for the effect of RP appears to be related to the upregulation of energy metabolism in skeletal muscle, which at the molecular level may be associated with PGC-1α and AMPK activation.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Isoflavonas/farmacologia , Músculo Esquelético/efeitos dos fármacos , Obesidade/prevenção & controle , Extratos Vegetais/farmacologia , Pueraria/química , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Alanina Transaminase/sangue , Animais , Fármacos Antiobesidade/farmacologia , Aspartato Aminotransferases/sangue , Linhagem Celular , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Dieta Hiperlipídica/efeitos adversos , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fosforilação , Raízes de Plantas/química , Triglicerídeos/sangue
10.
Sci Rep ; 6: 30321, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27457971

RESUMO

Sirtuin 6 (Sirt6), a chromatin associated class III deacetylase, controls whole-body energy homeostasis and has a critical role in glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells. However, its underlying molecular mechanism remains poorly understood. To gain further insights, we studied the pathway by which Sirt6 regulates GSIS utilizing mice lacking Sirt6 in their ß cells (ßS6KO). Further, we overexpressed wild type or deacetylase-inactive mutant Sirt6 in isolated islets as well as in MIN6 cells. We confirmed that ßS6KO mice developed glucose intolerance with severely impaired GSIS. Gene expression analysis of knockout islets and overexpression studies demonstrated that Sirt6 deacetylates forkhead box protein O1 (FoxO1) to trigger its nuclear export and releases its transcriptional repression of key glucose sensing genes such as Pdx1 and Glut2. Ectopic overexpression of Sirt6 in knockout islets resulted in rescue of the defective insulin secretion and restoration of the expression of Pdx1 and Glut2. These results show that Sirt6 in pancreatic ß cells deacetylates FoxO1 and subsequently increases the expression of Pdx1 and Glut2 to maintain the glucose-sensing ability of pancreatic ß cells and systemic glucose tolerance.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sirtuínas/genética , Acetilação , Animais , Linhagem Celular Tumoral , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Intolerância à Glucose , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Processamento de Proteína Pós-Traducional , Transativadores/genética , Transativadores/metabolismo
11.
Mol Nutr Food Res ; 60(11): 2481-2492, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27306613

RESUMO

SCOPE: Diet-induced obesity and consequent insulin resistance are caused, in part, by macrophage polarization and accumulation in peripheral tissues. Here, we examined the effects of endogenously synthesized n-3 PUFAs on macrophage chemotaxis and polarization. METHODS AND RESULTS: Fat-1 mice and wild-type (WT) littermates were fed a 60% calorie high-fat diet (HFD) for 10 weeks. Bone marrow macrophages (BMMs) from fat-1 and WT mice were used in in vitro chemotaxis assays and macrophage polarization studies. WT mice fed a HFD exhibited glucose intolerance, insulin resistance, and lipid accumulation and macrophage infiltration in liver and adipose tissue. However, these metabolic and inflammatory phenotypes were not observed in HFD-fed fat-1 mice. In flow cytometric analysis, M1 macrophage infiltration into adipose tissue was markedly attenuated in fat-1 mice. Consistently, results from in vitro experiments indicated that n-3 PUFAs prevented adipocyte conditioned medium-mediated macrophage chemotaxis, stimulated M2 polarization, and suppressed M1 polarization. The inhibition of macrophage migration by n-3 PUFAs was associated with suppression of multiple kinases, such as IκB kinase, AKT, and focal adhesion kinase. CONCLUSION: Our results indicate that n-3 PUFAs play a crucial role in macrophage polarization and chemotaxis, and thus regulate the development of HFD-induced tissue inflammation and metabolic derangements.


Assuntos
Dieta Hiperlipídica , Macrófagos/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Gorduras na Dieta/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Insaturados/farmacologia , Inflamação/metabolismo , Resistência à Insulina , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo
12.
Nutr Res ; 35(8): 655-63, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26048342

RESUMO

Schisandra chinensis fruit (SCF) is known to have beneficial effects on metabolic diseases, including obesity, and to affect gut microbiota in in vivo studies. However, in human research, there have been a few studies in terms of its clinical roles in lipid metabolism and modulation of gut microbiota. A double-blind, placebo-controlled study with 28 obese women with SCF or placebo was conducted for 12 weeks. Anthropometry and blood and fecal sampling were performed before and after treatment. Analysis of the gut microbiota in feces was performed using denaturing gradient gel electrophoresis and quantitative polymerase chain reaction. Although the values did not differ significantly between the 2 groups, the SCF group tended to show a greater decrease in waist circumference, fat mass, fasting blood glucose, triglycerides, aspartate aminotransferase, and alanine aminotransferase than the placebo group. Clustering of the denaturing gradient gel electrophoresis fingerprints for total bacteria before and after treatment indicated more separate clustering in SCF group than placebo. In correlation analysis, Bacteroides and Bacteroidetes (both increased by SCF) showed significant negative correlation with fat mass, aspartate aminotransferase, and/or alanine aminotransferase, respectively. Ruminococcus (decreased by SCF) showed negative correlation with high-density lipoprotein cholesterol and fasting blood glucose. In conclusion, administration of SCF for 12 weeks resulted in modulation of the gut microbiota composition in Korean obese women, and significant correlations with some bacterial genera and metabolic parameters were noted. However, in general, SCF was not sufficient to induce significant changes in obesity-related parameters compared with placebo.


Assuntos
Frutas , Microbioma Gastrointestinal , Obesidade/metabolismo , Schisandra/química , Adulto , Alanina Transaminase/sangue , Povo Asiático , Aspartato Aminotransferases/sangue , Bacteroides/isolamento & purificação , Bacteroidetes/isolamento & purificação , Biomarcadores/metabolismo , Glicemia/metabolismo , Composição Corporal , Índice de Massa Corporal , HDL-Colesterol/sangue , Análise por Conglomerados , DNA Bacteriano/genética , Método Duplo-Cego , Jejum , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Genômica , Humanos , Obesidade/sangue , Polifenóis/análise , Ruminococcus/isolamento & purificação , Triglicerídeos/sangue , Circunferência da Cintura
13.
Gut ; 64(2): 260-71, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24902766

RESUMO

OBJECTIVE: Programmed death-ligand 1 (PD-L1) has been shown to negatively regulate immune responses via its interaction with PD-1 receptor. In this study, we investigated the effects of PD-L1-Fc treatment on intestinal inflammation using two murine models of inflammatory colitis induced by dextran sulfate sodium (DSS) and T-cell transfer. DESIGN: The anti-colitis effect of adenovirus expressing Fc-conjugated PD-L1 (Ad/PD-L1-Fc) and recombinant PD-L1-Fc protein was evaluated in DSS-treated wild-type and Rag-1 knockout (KO) mice. We examined differentiation of T-helper cells, frequency of innate immune cells, and cytokine production by dendritic cells (DCs) in the colon from DSS-treated mice after PD-L1-Fc administration. In Rag-1 KO mice reconstituted with CD4 CD45RB(high) T cells, we assessed the treatment effect of PD-L1-Fc protein on the development of colitis. RESULTS: Administration of Ad/PD-L1-Fc significantly ameliorated DSS-induced colitis, which was accompanied by diminished frequency of interleukin (IL)-17A-producing CD4 T cells and increased interferon-γ-producing CD4 T cells in the colon of DSS-fed mice. The anti-colitic effect of PD-L1-Fc treatment was also observed in DSS-treated Rag-1 KO mice, indicating lymphoid cell independency. PD-L1-Fc modulated cytokine production by colonic DCs and the effect was dependent on PD-1 expression. Furthermore, PD-L1-Fc protein could significantly reduce the severity of colitis in CD4 CD45RB(high) T-cell-transferred Rag-1 KO mice. CONCLUSIONS: Based on the protective effect of PD-L1-Fc against DSS-induced and T-cell-induced colitis, our results suggest that PD-1-mediated inhibitory signals have a crucial role in limiting the development of colonic inflammation. This implicates that PD-L1-Fc may provide a novel therapeutic approach to treat inflammatory bowel disease.


Assuntos
Antígeno B7-H1/uso terapêutico , Colite Ulcerativa/prevenção & controle , Fatores Imunológicos/uso terapêutico , Doença Aguda , Adenoviridae/genética , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/farmacologia , Diferenciação Celular/efeitos dos fármacos , Colite Ulcerativa/etiologia , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Colo/imunologia , Citocinas/biossíntese , Células Dendríticas/imunologia , Sulfato de Dextrana , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Vetores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imunidade Inata , Imunidade nas Mucosas , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Fatores Imunológicos/genética , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Mucosa Intestinal/imunologia , Transfusão de Linfócitos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Auxiliares-Indutores/imunologia , Células Th17/imunologia
14.
J Endocrinol ; 224(2): 109-18, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25349250

RESUMO

Inflammation is an important factor in the development of insulin resistance. SIRT1, a class 3 histone/protein deacetylase, has anti-inflammatory functions. Myeloid-specific deletion of Sirt1 promotes macrophage infiltration into insulin-sensitive organs and aggravates tissue inflammation. In this study, we investigated how SIRT1 in macrophages alters tissue inflammation in the pancreas as well as liver and adipose tissue, and further explored the role of SIRT1 in locomotion of macrophages. Myeloid-specific Sirt1-deleted mice (mS1KO) and WT littermates were fed a 60% calorie high-fat diet (HFD) for 16 weeks. Tissue inflammation and metabolic phenotypes were compared. Bone marrow macrophages (BMMs) from WT or mS1KO mice were used in in vitro chemotaxis assays and macrophage polarization studies. mS1KO mice fed a HFD exhibited glucose intolerance, reduced insulin secretion, and insulin sensitivity with a slight decrease in body weight. Consistent with these results, pancreatic islets of mS1KO mice fed a HFD displayed decreased mass with profound apoptotic cell damage and increased macrophage infiltration and inflammation. Liver and adipose tissues from mS1KO HFD mice also showed greater accumulation of macrophages and tissue inflammation. Results from in vitro experiments indicated that deletion of myeloid Sirt1 stimulated proinflammatory M1-like polarization of BMMs and augmented the adipocyte-mediated macrophage chemotaxis. The latter effect was accompanied by increased expression and acetylation of focal adhesion kinase, as well as nuclear factor kappa B. Our results indicate that myeloid SIRT1 plays a crucial role in macrophage polarization and chemotaxis, and thus regulates the development of HFD-induced pancreatic inflammation and insulin secretion, and metabolic derangements in liver and adipose tissue.


Assuntos
Movimento Celular/genética , Dieta Hiperlipídica , Resistência à Insulina/genética , Macrófagos/imunologia , Sirtuína 1/fisiologia , Tecido Adiposo/imunologia , Animais , Atrofia/imunologia , Ensaios de Migração de Macrófagos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Gorduras na Dieta/farmacologia , Células HEK293 , Humanos , Inflamação/imunologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Fígado/imunologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Sirtuína 1/genética
15.
Am J Chin Med ; 42(4): 891-904, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25004881

RESUMO

The root bark of Lycium barbarum (Lycii radicis cortex, LRC) is used as a cooling agent for fever and night sweats in East Asian traditional medicine. The inhibitory effect of LRC water extract on inflammation is unknown. In this study, the anti-inflammatory effect of LRC was investigated in lipopolysaccharide (LPS)-stimulated mouse macrophage, RAW 264.7 cells. LRC extract significantly decreased the LPS-induced production of inflammatory mediators, nitric oxide (NO), prostaglandin (PG) E2 and pro-inflammatory cytokines, interleukin (IL)-1ß and IL-6 in the cells. In addition, LRC extract inhibited the LPS-induced expression of inducible NO synthase (iNOS) and cyclooxygenase (COX)-2 mRNA and protein, and inflammatory cytokines mRNA in the cells. The action mechanism of LRC underlies the blocking of LPS-mediated p38 and Jun N-terminal kinase (JNK), mitogen-activated protein kinases (MAPKs), and the nuclear factor (NF)-κB signaling pathway. These results indicate that LRC extract inhibits the inflammatory response in activated macrophages by down-regulating the transcription levels of inflammatory mediators and blocking the MAPKs and NF-κB pathway.


Assuntos
Anti-Inflamatórios , Lipopolissacarídeos/imunologia , Lycium , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Extratos Vegetais/farmacologia , Animais , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Casca de Planta , Raízes de Plantas
16.
Exp Mol Med ; 46: e98, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24875130

RESUMO

Type 1 diabetes is an autoimmune disease caused by permanent destruction of insulin-producing pancreatic ß cells and requires lifelong exogenous insulin therapy. Recently, islet transplantation has been developed, and although there have been significant advances, this approach is not widely used clinically due to the poor survival rate of the engrafted islets. We hypothesized that improving survival of engrafted islets through ex vivo genetic engineering could be a novel strategy for successful islet transplantation. We transduced islets with adenoviruses expressing betacellulin, an epidermal growth factor receptor ligand, which promotes ß-cell growth and differentiation, and transplanted these islets under the renal capsule of streptozotocin-induced diabetic mice. Transplantation with betacellulin-transduced islets resulted in prolonged normoglycemia and improved glucose tolerance compared with those of control virus-transduced islets. In addition, increased microvascular density was evident in the implanted islets, concomitant with increased endothelial von Willebrand factor immunoreactivity. Finally, cultured islets transduced with betacellulin displayed increased proliferation, reduced apoptosis and enhanced glucose-stimulated insulin secretion in the presence of cytokines. These experiments suggest that transplantation with betacellulin-transduced islets extends islet survival and preserves functional islet mass, leading to a therapeutic benefit in type 1 diabetes.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Intolerância à Glucose/cirurgia , Células Secretoras de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Transplante das Ilhotas Pancreáticas , Animais , Apoptose , Betacelulina , Proliferação de Células , Humanos , Células Secretoras de Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Ratos
17.
Vaccine ; 32(10): 1205-12, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23928465

RESUMO

Chemokines have been known to play an important role in eliciting adaptive immune responses by, selectively attracting the innate cellular components to the site of antigen presentation. In this study, we demonstrated that all three CXCR3 ligands, CXCL9, CXCL10, and CXCL11, could act as a strong, genetic adjuvant. Among them, CXCL11 increased vaccine antigen-specific CD8 T cells, including, several cytokine secretions (IFN-γ and TNF-α) to a greater degree than the other two CXCR3 ligands. Fc-fusion of CXCL11 (CXCL11-Fc) induced similar but slightly higher CD8 T cell response, which, appeared to be antigen- (ovalbumin (OVA) vs. human papillomavirus 16 (HPV16) E7) and vaccine, type- (adenovirus vs. DNA vaccine) independent. In addition, the adjuvant effect of CXCL11-Fc was, further confirmed by suppressing tumor growth and extension of survival rates in a therapeutic tumor, model, which was correlated with enhanced antigen-specific CD8 T cell responses. Interestingly, the, enhanced antigen-specific CD8 T cell responses by co-delivery of CXCL11-Fc were associated with CD8, T cell proliferation, followed by increased total and effector memory T cell frequencies. Taken together, our findings provide a novel role of CXCL11 as a strong genetic adjuvant which might be used to, increase antigen-specific CD8 T cell immunity elicited by vaccination.


Assuntos
Adjuvantes Imunológicos/genética , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Quimiocina CXCL11/imunologia , Adenoviridae , Animais , Vacinas Anticâncer/genética , Proliferação de Células , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quimiocina CXCL11/genética , Quimiocina CXCL9/genética , Quimiocina CXCL9/imunologia , Citocinas/imunologia , Feminino , Células HEK293 , Papillomavirus Humano 16 , Humanos , Imunidade Celular , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/terapia , Proteínas E7 de Papillomavirus/imunologia
18.
Food Chem ; 135(2): 452-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22868113

RESUMO

Lipopolysaccharide (LPS) can produce endotoxic shock by triggering the systemic inflammatory response. Here, we evaluated the in vitro and in vivo protective effects of unfermented and fermented Rhizoma coptidis (RC and FRC, respectively) against LPS-insult. In general, RC suppressed the LPS-induced expression of key inflammatory mediators in RAW264.7 cells, in a dose-dependent manner. Notably, FRC at a 20 µg/ml dose in combination with the probiotic used for fermentation showed more potent in vitro anti-inflammatory activities than that exhibited by the corresponding dose of RC. Moreover, oral treatment with FRC in association with the probiotic, but not oral administration of RC, significantly attenuated blood endotoxin and C-reactive protein (CRP) levels, and gut permeability, and significantly augmented the intestinal population of Bifodobacterium spp. and Lactobacillus spp. in LPS-treated animals. Our results demonstrate the beneficial impact of fermented RC in combination with the associated probiotic in combating LPS-insult both in vitro and in vivo.


Assuntos
Anti-Inflamatórios/farmacologia , Coptis/química , Inflamação/tratamento farmacológico , Lipopolissacarídeos/imunologia , Extratos Vegetais/farmacologia , Probióticos/farmacologia , Rizoma/química , Animais , Anti-Inflamatórios/análise , Anti-Inflamatórios/metabolismo , Linhagem Celular , Coptis/microbiologia , Fermentação , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Inflamação/genética , Inflamação/imunologia , Leuconostoc/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Extratos Vegetais/análise , Extratos Vegetais/metabolismo , Probióticos/análise , Probióticos/metabolismo , Ratos , Ratos Sprague-Dawley , Rizoma/microbiologia
19.
Clin Immunol ; 144(3): 190-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22836084

RESUMO

IL-12p40 homodimer is a natural antagonist of IL-12 and IL-23, which are potent pro-inflammatory cytokines required for Th1 and Th17 immune responses, respectively. It has been reported that Th17 response is involved in inflammatory bowel disease (IBD), a chronic disorder of the digestive system with steadily increasing incidence. Here, we investigated the effects of IL-12p40 delivered via recombinant adenovirus (rAd/IL-12p40) or mesenchymal stem cells (MSC/IL-12p40) in a dextran sulfate sodium salt (DSS)-induced colitis model. Injection of rAd/IL-12p40 or MSC/IL-12p40 efficiently attenuated colitis symptoms and tissue damage, leading to an increased survival rate. Moreover, IL-12p40 delivery suppressed IL-17A, but enhanced IFN-γ production from mesenteric lymph node cells, supporting the preferential suppression of IL-23 by IL-12p40 homodimer in vitro and the suppression of Th17 responses in vivo. Our results demonstrate that IL-12p40 delivery ameliorates DSS-induced colitis by suppressing IL-17A production and inflammation in the intestinal mucosa, providing an effective new therapeutic strategy for IBDs.


Assuntos
Colite/imunologia , Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Subunidade p40 da Interleucina-12/imunologia , Interleucina-17/imunologia , Mucosa Intestinal/imunologia , Adenoviridae/imunologia , Animais , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Feminino , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-23/imunologia , Linfonodos/imunologia , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Células Th17/imunologia
20.
J Immunother ; 34(3): 297-306, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21389868

RESUMO

Programmed death 1 (PD-1) signaling through its ligands, PD-L1 and PD-L2, has been known to negatively regulate T-cell responses. In addition, PD-L1 has been shown to interact with B7-1 costimulatory molecule to inhibit T-cell responses. Extensive studies have shown that PD-1/PD-L blockade restores exhausted T cells during chronic viral infections and tumors. In this study, we evaluated the effects of soluble PD-1 (sPD-1) as a blockade of PD-1 and PD-L1 on vaccine-elicited antigen-specific T-cell responses in mice. Coadministration of sPD-1 DNA with human papilloma virus-16 E7 DNA vaccine significantly enhanced E7-specific CD8(+) T-cell responses, resulting in potent antitumor effects against E7-expressing tumors. We also found that sPD-1, codelivered with adenovirus-based vaccine, could increase antigen-specific CD8(+) T-cell responses, indicating vaccine type-independent adjuvant effect of sPD-1. In addition, the frequency and functional activity of adoptively transferred OT-I cells, particularly memory CD8(+) T cells, were augmented by coadministration of sPD-1 DNA, which was closely associated with increased T-cell proliferation and reduced T-cell apoptosis through upregulation of Bcl-xL expression during T-cell activation. Codelivery of sPD-1 DNA also enhanced maturation of dendritic cells (DCs) in vivo which was accompanied by upregulation of DC maturation markers such as major histocompatibility complex class II. Taken together, our findings show that sPD-1 potently enhances codelivered antigen-specific CD8(+) T-cell responses and in vivo maturation of DCs during activation of naive CD8(+) T cells, suggesting that an immunization strategy with sPD-1 as an adjuvant can be used to increase antigen-specific T-cell immunity elicited by vaccination.


Assuntos
Adjuvantes Imunológicos/farmacologia , Transferência Adotiva , Antígenos CD/imunologia , Antígenos CD/farmacologia , Proteínas Reguladoras de Apoptose/imunologia , Proteínas Reguladoras de Apoptose/farmacologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Vacinas de DNA , Adenoviridae/genética , Adenoviridae/imunologia , Adenoviridae/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos CD/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/uso terapêutico , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/imunologia , Papillomavirus Humano 16/metabolismo , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Papiloma/imunologia , Papiloma/metabolismo , Papiloma/terapia , Proteínas E7 de Papillomavirus/imunologia , Proteínas E7 de Papillomavirus/metabolismo , Receptor de Morte Celular Programada 1 , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA