Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Arthritis Res Ther ; 24(1): 234, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253807

RESUMEN

BACKGROUND: Abnormal proliferation of fibroblast-like synoviocytes (FLSs) in the synovial lining layer is the primary cause of synovial hyperplasia and joint destruction in rheumatoid arthritis (RA). Currently, the relationship between metabolic abnormalities and FLS proliferation is a new focus of investigation. However, little is known regarding the relationship between amino acid metabolism and RA. METHODS: The concentrations of amino acids and cytokines in the synovial fluid of RA (n = 9) and osteoarthritis (OA, n = 9) were detected by LC-MS/MS and CBA assay, respectively. The mRNA and protein expression of cationic amino acid transporter-1 (CAT-1) were determined in FLSs isolated from RA and OA patients by real-time PCR and western blotting. MTT assay, cell cycle, apoptosis, invasion, and cytokine secretion were determined in FLSs knocked down of CAT-1 using siRNA or treated with D-arginine under normoxic and hypoxic culture conditions. A mouse collagen-induced arthritis (CIA) model was applied to test the therapeutic potential of blocking the uptake of L-arginine in vivo. RESULTS: L-rginine was upregulated in the synovial fluid of RA patients and was positively correlated with the elevation of the cytokines IL-1ß, IL-6, and IL-8. Further examination demonstrated that CAT-1 was the primary transporter for L-arginine and was overexpressed on RA FLSs compared to OA FLSs. Moreover, knockdown of CAT-1 using siRNA or inhibition of L-arginine uptake using D-arginine significantly suppressed L-arginine metabolism, cell proliferation, migration, and cytokine secretion in RA FLSs under normoxic and hypoxic culture conditions in vitro but increased cell apoptosis in a dose-dependent manner. Meanwhile, in vivo assays revealed that an L-arginine-free diet or blocking the uptake of L-arginine using D-arginine suppressed arthritis progression in CIA mice. CONCLUSION: CAT-1 is upregulated and promotes FLS proliferation by taking up L-arginine, thereby promoting RA progression.


Asunto(s)
Arginina , Artritis Experimental , Artritis Reumatoide , Transportador de Aminoácidos Catiónicos 1 , Sinoviocitos , Animales , Ratones , Aminoácidos/metabolismo , Artritis Experimental/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Transportador de Aminoácidos Catiónicos 1/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cromatografía Liquida , Citocinas/metabolismo , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Ratones Endogámicos CBA , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Membrana Sinovial/metabolismo , Sinoviocitos/metabolismo , Espectrometría de Masas en Tándem
2.
Br J Nutr ; 123(2): 135-148, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31647043

RESUMEN

Oral arginine supplements are popular mainly for their presumed vasodilatory benefit. Arginine is a substrate for at least four enzymes including nitric oxide synthase (NOS) and arginase, but the impact of oral supplements on its different metabolic pathways is not clear. Deficiencies of arginine-metabolising enzymes are associated with conditions such as hyperammonaemia, endothelial dysfunction, central nervous system and muscle dysfunction, which complicate the use of oral arginine supplements. We examined the effect of l-arginine (l-Arg) and d-arginine (d-Arg), each at 500 mg/kg per d in drinking water administered for 4 weeks to separate groups of 9-week-old male Sprague-Dawley rats. We quantified the expression of enzymes and plasma, urine and organ levels of various metabolites of arginine. l-Arg significantly decreased cationic transporter-1 expression in the liver and the ileum and increased endothelial NOS expression in the aorta and the kidney and plasma nitrite levels, but did not affect the mean arterial pressure. l-Arg also decreased the expression of arginase II in the ileum, arginine:glycine amidinotransferase in the liver and the kidney and glyoxalase I in the liver, ileum and brain, but increased the expression of arginine decarboxylase and polyamines levels in the liver. d-Arg, the supposedly inert isomer, also unexpectedly affected the expression of some enzymes and metabolites. In conclusion, both l- and d-Arg significantly affected enzymes and metabolites in several pathways that use arginine as a substrate and further studies with different doses and treatment durations are planned to establish their safety or adverse effects to guide their use as oral supplements.


Asunto(s)
Arginina/administración & dosificación , Arginina/metabolismo , Suplementos Dietéticos , Administración Oral , Animales , Arginasa/efectos de los fármacos , Arginasa/metabolismo , Arginina/farmacología , Transportador de Aminoácidos Catiónicos 1/efectos de los fármacos , Transportador de Aminoácidos Catiónicos 1/metabolismo , Creatina/efectos de los fármacos , Creatina/metabolismo , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Nitratos/sangre , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa/metabolismo , Nitritos/sangre , Ratas , Ratas Sprague-Dawley
3.
Cell Physiol Biochem ; 44(5): 1749-1761, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29216634

RESUMEN

BACKGROUND/AIMS: Our previous reports suggested that dietary supplementation with lysine influenced intestinal absorption and metabolism of amino acids. In this study, we further investigated the effect of lysine restriction (30%) on feed intake and we also tested the hypothesis that gut microbiome contributed to the potential mechanism of lysine restriction-mediated feeding behavior. Here, we profiled gut microbial communities by sequencing 16S ribosomal ribonucleic acid (rRNA) genes from gut samples as well as growth performance, serum hormones, and intestinal lysine transport in a piglet model. RESULTS: Piglets preferred to the lysine restricted diet when giving three diets and the feed intake was markedly higher in the lysine-restricted group than that in the control group. Altered hormones (leptin, CCK, and ghrelin) might contribute to the feeding behavior caused by lysine restriction. Meanwhile, lysine transporting ability (SLC7A1 and SLC7A2 expression, intestinal electrophysiological changes, and amino acid pool in mesenteric vein) was decreased in response to lysine restriction. Through deep sequencing of bacterial rRNA markers, we observed that bacterial diversity was enhanced in the lysine-restricted group (Shannon H, PD, and Chao1). At the phylum level, lysine restriction enhanced gut Actinobacteria, Saccharibacteria, and Synergistetes abundances. At the family level, Moraxellaceae, Halomonadaceae, Shewanellaceae, Corynebacteriaceae, Bacillaceae, Comamonadaceae, Microbacteriaceae, Caulobacteraceae, and Synergistaceae abundances were increased in response to lysine restriction. Predictive functional profiling of microbial communities by PICRUSt also confirmed that dietary lysine restriction affected gut microbiome, which might further mediate amino acid metabolism, membrane transport, and endocrine system. CONCLUSION: Our results indicated that lysine restriction inhibited intestinal lysine transport and promoted feed intake, which might be associated with gut microbiome.


Asunto(s)
Aminoácidos/metabolismo , Dieta , Intestinos/microbiología , Lisina/deficiencia , Microbiota , Aminoácidos/sangre , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Transportador de Aminoácidos Catiônicos 2/genética , Transportador de Aminoácidos Catiônicos 2/metabolismo , Ingestión de Alimentos , Ensayo de Inmunoadsorción Enzimática , Gastrinas/sangre , Ghrelina/sangre , Leptina/sangre , Masculino , Análisis de Componente Principal , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Porcinos
4.
Sci Rep ; 7(1): 4767, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28684763

RESUMEN

Low plasma concentration of L-homoarginine is an independent predictor of cardiovascular events and total mortality. Experimental data indicate that supplementation of L-homoarginine may have protective effects. We aimed to elucidate the mechanisms involved in the cellular uptake of L-homoarginine, which are little understood, so far. Using human embryonic kidney (HEK293) cell lines stably overexpressing the human cationic amino acid transporters CAT1 [solute carrier family 7 (SLC7A1)], CAT2A (SLC7A2A) or CAT2B (SLC7A2B) we assessed the transport kinetics of L-homoarginine and interactions with the CAT substrates L-arginine and asymmetric dimethylarginine (ADMA). Significant uptake of L-homoarginine was observed for all three CATs with apparent KM-values of 175 ± 7 µM for CAT1 and 523 ± 35 µM for CAT2B. Saturation of CAT2A-mediated L-homoarginine uptake could not be reached. Uptake of L-homoarginine by any of the three CATs could be inhibited by L-arginine and ADMA. Significant inhibition of CAT1-mediated uptake of L-homoarginine by L-arginine already occurred in the physiological concentration range. Taken together these data demonstrate that L-homoarginine is a substrate of CAT1, CAT2A and CAT2B and that CAT1 is a key site with regard to physiological relevance and interactions with related substrates such as L-arginine.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Arginina/análogos & derivados , Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Homoarginina/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Transporte Biológico Activo , Transportador de Aminoácidos Catiónicos 1/genética , Expresión Génica , Células HEK293 , Humanos , Cinética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Especificidad por Sustrato , Transgenes
5.
Mol Cell Biochem ; 399(1-2): 229-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25336270

RESUMEN

L-arginine (L-Arg) uptake is mediated by members of cationic amino acid transporter (CAT) family and may coincide with the induction of nitric oxide synthases (NOS). The present study was conducted to investigate the extracellular concentrations of L-Arg regulating the CAT-1, CAT-4 and inducible NOS (iNOS) in chick intestinal epithelial cells. The cells were cultured for 4 days in Arg-free Dulbecco's modified Eagle's medium containing 10, 100, 200, 400, or 600 µM L-Arg. Cell viability, nitric oxide (NO) concentrations, uptake and metabolism of L-[3H]-Arg as well as expression of CAT-1, CAT-4, and iNOS were determined. Our results showed that L-Arg enhances cell growth with a maximal response at 10-400 µM. Addition of 100, 200, or 400 µM L-Arg increased the L-[3H]-Arg uptake, which was associated with greater conversion of L-[3H]-citrulline and NO production in comparison with 10 µM L-Arg group. Increasing extracellular concentrations of L-Arg from 10 to 400 µM dose dependently increased the levels of CAT-1 mRNA and protein, while no effect on CAT-4 mRNA abundance was found. Furthermore, supplementation of 100, 200, or 400 µM L-Arg upregulated the expression of iNOS mRNA, and the relative protein levels for iNOS in 200 and 400 µM L-Arg groups were higher than those in 10 and 100 µM L-Arg groups. Collectively, we conclude that the CAT-1 isoform plays a role in L-Arg uptake, and L-Arg-mediated elevation of NO via iNOS promotes the growth of chick intestinal epithelial cells.


Asunto(s)
Arginina/fisiología , Proteínas Aviares/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Proliferación Celular , Células Epiteliales/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Embrión de Pollo , Expresión Génica , Mucosa Intestinal/citología , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética
6.
Amino Acids ; 46(4): 883-92, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24368521

RESUMEN

This study tested the hypothesis that dietary L-arginine supplementation confers beneficial effects on growing pigs fed a mold-contaminated diet. The measured variables included: (1) the average daily weight gain and feed:gain ratio; (2) activities of total superoxide dismutase, glutathione peroxidase, diamine oxidase, as well as amino acid and D-lactate concentrations in serum; (3) intestinal morphology; (4) expression of the genes for SLC7A7 (amino acid transporter light chain, y(+L) system, family 7, member 7), SLC7A1 (cationic amino acid transporter, y(+) system, family 7, member 1), SLC1A1 (neuronal/epithelial high affinity glutamate transporter, system XAG, member 1), SLC5A1 (sodium/glucose cotransporter, family 5, member 1) in the ileum and jejunum. Mycotoxins in feedstuffs resulted in an enlarged small intestine mass, oxidative injury in tissues, and reduced growth performance in pigs. Dietary arginine supplementation enhanced (P < 0.05) expression of jejunal SLC7A7 and ileal SLC7A1, in comparison with the control and mycotoxin groups. In addition, supplementing 1% L-arginine to the mycotoxin-contaminated feed had the following beneficial effects (P < 0.05): (1) alleviating the imbalance of the antioxidant system in the body; (2) ameliorating intestinal abnormalities; and (3) attenuating whole-body growth depression, compared with the mycotoxin group without arginine treatment. Collectively, these results indicate that dietary supplementation with L-arginine exerts a protective role in pigs fed mold-contaminated foods. The findings may have important nutritional implications for humans and other mammals.


Asunto(s)
Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/genética , Suplementos Dietéticos/análisis , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Intestino Delgado/metabolismo , Micotoxinas/efectos adversos , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo , Alimentación Animal/análisis , Animales , Transportador de Aminoácidos Catiónicos 1/metabolismo , Femenino , Contaminación de Alimentos/análisis , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Masculino , Micotoxinas/análisis , Porcinos/genética
7.
Genet Mol Res ; 12(1): 115-26, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23408397

RESUMEN

Leucine (Leu) participates in the activity of cationic amino acid (aa) transporters. Also, branched-chain aa [Leu, isoleucine (Ile), and valine (Val)] share intestinal transporters for absorption. We conducted an experiment with 16 young pigs (body weight of about 16 kg) to determine whether Leu and Ile affect expression of aa transporters b(0,+) and CAT-1 in the jejunum and expression of myosin in muscle, as well as serum concentration of essential aa, and growth performance in pigs. Dietary treatments were: wheat-based diets fortified with Lys, Thr, and Met; basal diet plus 0.50% Leu; basal diet plus 0.50% Ile, and basal diet plus 0.50% Leu and 0.50% Ile. After 28 days, the pigs were sacrificed to collect blood, jejunum, and semitendinosus and longissimus muscle samples. The effects of single and combined addition of Leu and Ile were analyzed. Leu alone or combined with Ile significantly decreased daily weight gain and reduced feed conversion. Leu and Ile, alone or in combination, significantly decreased expression of b(0,+) and significantly increased CAT-1. Ile alone or combined with Leu significantly decreased myosin expression in semitendinosus and significantly decreased it in longissimus muscle. Leu alone significantly decreased Lys, Ile and Thr serum concentrations; Ile significantly decreased Thr serum concentration; combined Leu and Ile significantly decreased Thr and significantly increased Val serum concentration. We conclude that dietary levels of Leu and Ile affect growth performance, expression of aa transporters and myosin, and aa serum concentrations in pigs.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Expresión Génica/genética , Isoleucina/metabolismo , Leucina/metabolismo , Miosinas/genética , Porcinos/fisiología , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Aminoácidos/sangre , Aminoácidos/genética , Aminoácidos/metabolismo , Alimentación Animal , Animales , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Dieta , Suplementos Dietéticos , Isoleucina/genética , Yeyuno/metabolismo , Leucina/genética , Músculos/metabolismo , Miosinas/metabolismo , Porcinos/genética , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo , Valina/genética , Valina/metabolismo , Aumento de Peso
8.
Vascul Pharmacol ; 56(3-4): 150-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22239978

RESUMEN

UNLABELLED: Ginsenoside Rb1 and Rg1 are major components of Panax notoginseng (P.N.), an herb with known clinical efficacy in hypertension and myocardial ischemia in Eastern countries. This investigation is to elicit the mechanism of these components in hypertension via their effect on vascular reaction. To assess the ability of P.N. in hypertension, P.N. extracts were injected in spontaneously hypertensive rats (SHR) via the vena caudalis; Low dosages of P.N. extracts significantly lowered blood pressure in SHR. Examination with Rb1 and Rg1 revealed significant vasodilatation using mouse coronary arteries in a dose-dependent manner. Rb1- and Rg1-induced vasodilatation was blocked by pre-incubation with eNOS and PI3K inhibitors. Coronaries of eNOS-/- mice showed attenuated vasodilatation with Rb1 and Rg1. In addition, both Rb1 and Rg1 induce nitric oxide (NO) generation through increasing the phosphorylation of eNOS, activating Na+-independent l-arginine transport, and stimulating cationic amino acid transport (CAT)-1 mRNA expression in cultured endothelial cells. CONCLUSION: Ginsenoside Rb1 and Rg1 increased endothelial-dependent vessel dilatation through the activation of NO by modulating the PI3K/Akt/eNOS pathway and l-arginine transport in endothelial cells. These findings may have important implications for understanding the mechanisms of clinical efficacy of the herb P.N. when used in the regulation of blood pressure.


Asunto(s)
Antihipertensivos/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Ginsenósidos/farmacología , Hipertensión/tratamiento farmacológico , Animales , Antihipertensivos/análisis , Antihipertensivos/química , Antihipertensivos/farmacología , Transportador de Aminoácidos Catiónicos 1/antagonistas & inhibidores , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Células Cultivadas , Medicamentos Herbarios Chinos/química , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ginsenósidos/análisis , Técnicas In Vitro , Masculino , Ratones , Ratones Congénicos , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Distribución Aleatoria , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Sus scrofa , Vasodilatación/efectos de los fármacos
9.
Biol Reprod ; 83(5): 791-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20668257

RESUMEN

In vitro embryo culture systems promote development at rates lower than in vivo systems. The goal of this project was to discover transcripts that may be responsible for a decrease of embryo competency in blastocyst-stage embryos cultured in vitro. Gilts were artificially inseminated on the first day of estrus, and on Day 2, one oviduct and the tip of a uterine horn were flushed and the recovered embryos were cultured in porcine zygote medium 3 for 4 days. On Day 6, the gilts were euthanized and the contralateral horn was flushed to obtain in vivo derived embryos. Total RNA was extracted from three pools of 10 blastocysts from each treatment. First and second strand cDNA was synthesized and sequenced using Illumina sequencing. The reads generated were aligned to a custom-built database designed to represent the known porcine transcriptome. A total of 1170 database members were different between the two groups (P < 0.05), and 588 of those had at least a 2-fold difference. Eleven transcripts were subjected to real-time PCR that validated the sequencing. There was an overall decrease in inner cell mass (ICM) and trophectodermal (TE) cell numbers in embryos cultured in vitro; however, no difference in the ICM:TE ratio was found. Interestingly, the transcript SLC7A1 was higher in the in vitro cultured group. This difference disappeared after addition of arginine to the 4-day culture. Illumina sequencing and alignment to a custom transcriptome identified a large number of genes that yield clues on ways to manipulate the culture media to mimic the in vivo environment.


Asunto(s)
Blastocisto/metabolismo , Desarrollo Embrionario , Fertilización In Vitro/veterinaria , Regulación del Desarrollo de la Expresión Génica , ARN Mensajero/metabolismo , Sus scrofa/embriología , Crianza de Animales Domésticos/métodos , Animales , Arginina/metabolismo , Blastocisto/citología , Masa Celular Interna del Blastocisto/citología , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Recuento de Células/veterinaria , ADN Complementario/química , ADN Complementario/metabolismo , Bases de Datos de Ácidos Nucleicos , Técnicas de Cultivo de Embriones/veterinaria , Femenino , Fertilización In Vitro/métodos , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/veterinaria , Microquímica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de ADN/veterinaria , Sus scrofa/metabolismo , Trofoblastos/citología
10.
Hypertension ; 48(2): 278-85, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16801489

RESUMEN

Endothelial dysfunction is a hallmark of cardiovascular disease, and the l-arginine:NO pathway plays a critical role in determining endothelial function. Recent studies suggest that smoking, a well-recognized risk factor for vascular disease, may interfere with l-arginine and NO metabolism; however, this remains poorly characterized. Accordingly, we performed a series of complementary in vivo and in vitro studies to elucidate the mechanism by which cigarette smoke adversely affects endothelial function. In current smokers, plasma levels of asymmetrical dimethyl-arginine (ADMA) were 80% higher (P = 0.01) than nonsmokers, whereas citrulline (17%; P < 0.05) and N-hydroxy-l-arginine (34%; P < 0.05) were significantly lower. Exposure to 10% cigarette smoke extract (CSE) significantly affected endothelial arginine metabolism with reductions in the intracellular content of citrulline (81%), N-hydroxy-l-arginine (57%), and arginine (23%), while increasing ADMA (129%). CSE significantly inhibited (38%) arginine uptake in conjunction with a 34% reduction in expression of the arginine transporter, CAT1. In conjunction with these studies, CSE significantly reduced the activity of eNOS and NO production by endothelial cells, while stimulating the production of reactive oxygen species. In conclusion, cigarette smoke adversely affects the endothelial l-arginine NO synthase pathway, resulting in reducing NO production and elevated oxidative stress. In conjunction, exposure to cigarette smoke increases ADMA concentration, the latter being a risk factor for cardiovascular disease.


Asunto(s)
Arginina/metabolismo , Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo , Humo/efectos adversos , Fumar , Adulto , Animales , Antioxidantes/farmacología , Arginina/análogos & derivados , Arginina/sangre , Arginina/farmacocinética , Disponibilidad Biológica , Transporte Biológico/efectos de los fármacos , Transportador de Aminoácidos Catiónicos 1/metabolismo , Bovinos , Células Cultivadas , Citrulina/sangre , Cotinina/sangre , Medios de Cultivo/química , Medios de Cultivo/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Óxido Nítrico Sintasa/metabolismo , Oxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Fumar/efectos adversos , Fumar/sangre , Nicotiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA