Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nitric Oxide ; 148: 13-22, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38642795

RESUMEN

Endothelial dysfunction, underlying the vascular complications of diabetes and other cardiovascular disorders, may result from uncoupling of endothelial nitric oxide synthase (eNOS) activity due to decreased levels of tetrahydrobiopterin (BH4), a critical co-factor for eNOS. Some clinical trials attempting to deliver exogenous BH4 as a potential therapeutic strategy in vascular disease states have failed due to oxidation of BH4 in the circulation. We sought to develop a means of protecting BH4 from oxidation while delivering it to dysfunctional endothelial cells. Polymeric and solid lipid nanoparticles (NPs) loaded with BH4 were delivered by injection or oral gavage, respectively, to streptozotocin-induced diabetic rats. BH4 was measured in coronary endothelial cells and endothelium-dependent vascular reactivity was assessed in vascular rings. Lymphatic uptake of orally delivered lipid NPs was verified by sampling mesenteric lymph. BH4-loaded polymeric NPs maintained nitric oxide production by cultured endothelial cells under conditions of oxidative stress. BH4-loaded NPs, delivered via injection or ingestion, increased coronary endothelial BH4 concentration and improved endothelium-dependent vasorelaxation in diabetic rats. Pharmacodynamics assessment indicated peak concentration of solid lipid NPs in the systemic bloodstream 6 hours after ingestion, with disappearance noted by 48 hours. These studies support the feasibility of utilizing NPs to deliver BH4 to dysfunctional endothelial cells to increase nitric oxide bioavailability. BH4-loaded NPs could provide an innovative tool to restore redox balance in blood vessels and modulate eNOS-mediated vascular function to reverse or retard vascular disease in diabetes.


Asunto(s)
Biopterinas , Diabetes Mellitus Experimental , Endotelio Vascular , Nanopartículas , Animales , Biopterinas/análogos & derivados , Biopterinas/farmacología , Biopterinas/administración & dosificación , Biopterinas/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Masculino , Nanopartículas/química , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ratas , Ratas Sprague-Dawley , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo
2.
Int J Mol Sci ; 24(6)2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36982254

RESUMEN

Sustained tissue hypoxia is associated with many pathophysiological conditions, including chronic inflammation, chronic wounds, slow-healing fractures, microvascular complications of diabetes, and metastatic spread of tumors. This extended deficiency of oxygen (O2) in the tissue sets creates a microenvironment that supports inflammation and initiates cell survival paradigms. Elevating tissue carbon dioxide levels (CO2) pushes the tissue environment toward "thrive mode," bringing increased blood flow, added O2, reduced inflammation, and enhanced angiogenesis. This review presents the science supporting the clinical benefits observed with the administration of therapeutic CO2. It also presents the current knowledge regarding the cellular and molecular mechanisms responsible for the biological effects of CO2 therapy. The most notable findings of the review include (a) CO2 activates angiogenesis not mediated by hypoxia-inducible factor 1a, (b) CO2 is strongly anti-inflammatory, (c) CO2 inhibits tumor growth and metastasis, and (d) CO2 can stimulate the same pathways as exercise and thereby, acts as a critical mediator in the biological response of skeletal muscle to tissue hypoxia.


Asunto(s)
Dióxido de Carbono , Hipoxia , Humanos , Dióxido de Carbono/metabolismo , Oxígeno/metabolismo , Ejercicio Físico , Homeostasis
3.
Amino Acids ; 54(2): 193-204, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34741684

RESUMEN

Dietary supplementation with 0.4 or 0.8% L-arginine (Arg) to gilts between days 14 and 25 of gestation enhances embryonic survival and vascular development in placentae; however, the underlying mechanisms are largely unknown. This study tested the hypothesis that Arg supplementation stimulated placental expression of mRNAs and proteins that enhance angiogenesis, including endothelial nitric oxide synthase (eNOS), vascular endothelial growth factor (VEGF), placental growth factor (PGF), GTP cyclohydrolase-I (GTP-CH1), ornithine decarboxylase (ODC1), and vascular endothelial growth factor receptors 1 and 2 (VEGFR1 and VEGFR2). Beginning on the day of breeding, gilts were fed daily 2 kg of a corn-soybean meal-based diet supplemented with 0.0 (control), 0.4, or 0.8% Arg. On day 25 of gestation, gilts were hysterectomized to obtain uteri and conceptuses for histochemical and biochemical analyses. eNOS and VEGFR1 proteins were localized to endothelial cells of maternal uterine blood vessels and to the uterine luminal epithelium, respectively. Compared with the control, dietary supplementation with 0.4 or 0.8% Arg increased (P < 0.05) the amounts of nitrite plus nitrate (NOx; oxidation products of NO) and polyamines in allantoic and amniotic fluids, concentrations of NOx, tetrahydrobiopterin (BH4, an essential cofactor for all NOS isoforms) and polyamines in placentae, as well as placental protein abundances of GTP-CH1 (the key enzyme for BH4 production) and ODC1 (the key enzyme for polyamine synthesis). Placental  mRNA levels for GTP-CH1, eNOS, PGF, VEGF, and VEGFR2 increased in response to both 0.4% and 0.8% Arg supplementation. Collectively, these results indicate that dietary Arg supplementation to gilts between days 14 and 25 of pregnancy promotes placental angiogenesis by increasing the expression of mRNAs and proteins for angiogenic factors as well as NO and polyamine syntheses.


Asunto(s)
Proteínas Angiogénicas , Placenta , Proteínas Angiogénicas/metabolismo , Animales , Arginina/metabolismo , Arginina/farmacología , Suplementos Dietéticos , Células Endoteliales/metabolismo , Femenino , Placenta/metabolismo , Factor de Crecimiento Placentario/metabolismo , Poliaminas/metabolismo , Embarazo , Sus scrofa/metabolismo , Porcinos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(24): 11590-11595, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138695

RESUMEN

Exposure to fine particulate matter (PM) during pregnancy is associated with high risks of birth defects/fatality and adverse long-term postnatal health. However, limited mechanistic data are available to assess the detailed impacts of prenatal PM exposure. Here we evaluate fine PM exposure during pregnancy on prenatal/postnatal organogenesis in offspring and in predisposing metabolic syndrome for adult life. Between days 0 and 18 of gestation, two groups of adult female rats (n = 10 for each) were placed in a dual-exposure chamber device, one with clean ambient air (∼3 µg·m-3) and the other with ambient air in the presence of 100 to 200 µg·m-3 of ultrafine aerosols of ammonium sulfate. At birth (postnatal day 0, PND0), four males and four females were selected randomly from each litter to be nursed by dams, whereas tissues were collected from the remaining pups. At PND21, tissues were collected from two males and two females, whereas the remaining pups were fed either a high- or low-fat diet until PND105, when tissues were obtained for biochemical and physiological analyses. Maternal exposure to fine PM increased stillbirths; reduced gestation length and birth weight; increased concentrations of glucose and free fatty acids in plasma; enhanced lipid accumulation in the liver; and decreased endothelium-dependent relaxation of aorta. This lead to altered organogenesis and predisposed progeny to long-term metabolic defects in an age-, organ-, and sex-specific manner. Our results highlight the necessity to develop therapeutic strategies to remedy adverse health effects of maternal PM exposure on conceptus/postnatal growth and development.


Asunto(s)
Exposición Materna/efectos adversos , Síndrome Metabólico/inducido químicamente , Organogénesis/efectos de los fármacos , Material Particulado/efectos adversos , Efectos Tardíos de la Exposición Prenatal/patología , Contaminación del Aire/efectos adversos , Animales , Peso al Nacer/efectos de los fármacos , Susceptibilidad a Enfermedades/sangre , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/patología , Exposición a Riesgos Ambientales/efectos adversos , Ácidos Grasos/sangre , Femenino , Glucosa/metabolismo , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Organogénesis/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Adv Exp Med Biol ; 1332: 167-187, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34251644

RESUMEN

As a functional amino acid (AA), L-arginine (Arg) serves not only as a building block of protein but also as an essential substrate for the synthesis of nitric oxide (NO), creatine, polyamines, homoarginine, and agmatine in mammals (including humans). NO (a major vasodilator) increases blood flow to tissues. Arg and its metabolites play important roles in metabolism and physiology. Arg is required to maintain the urea cycle in the active state to detoxify ammonia. This AA also activates cellular mechanistic target of rapamycin (MTOR) and focal adhesion kinase cell signaling pathways in mammals, thereby stimulating protein synthesis, inhibiting autophagy and proteolysis, enhancing cell migration and wound healing, promoting spermatogenesis and sperm quality, improving conceptus survival and growth, and augmenting the production of milk proteins. Although Arg is formed de novo from glutamine/glutamate and proline in humans, these synthetic pathways do not provide sufficient Arg in infants or adults. Thus, humans and other animals do have dietary needs of Arg for optimal growth, development, lactation, and fertility. Much evidence shows that oral administration of Arg within the physiological range can confer health benefits to both men and women by increasing NO synthesis and thus blood flow in tissues (e.g., skeletal muscle and the corpora cavernosa of the penis). NO is a vasodilator, a neurotransmitter, a regulator of nutrient metabolism, and a killer of bacteria, fungi, parasites, and viruses [including coronaviruses, such as SARS-CoV and SARS-CoV-2 (the virus causing COVID-19). Thus, Arg supplementation can enhance immunity, anti-infectious, and anti-oxidative responses, fertility, wound healing, ammonia detoxification, nutrient digestion and absorption, lean tissue mass, and brown adipose tissue development; ameliorate metabolic syndromes (including dyslipidemia, obesity, diabetes, and hypertension); and treat individuals with erectile dysfunction, sickle cell disease, muscular dystrophy, and pre-eclampsia.


Asunto(s)
COVID-19 , Óxido Nítrico , Animales , Arginina/metabolismo , Femenino , Humanos , Masculino , Embarazo , Biosíntesis de Proteínas , SARS-CoV-2
6.
Adv Exp Med Biol ; 1332: 211-227, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34251646

RESUMEN

Measuring usual dietary intake in freely living humans is difficult to accomplish. As a part of our recent study, a food frequency questionnaire was completed by healthy adult men and women at days 0 and 90 of the study. Data from the food questionnaire were analyzed with a nutrient analysis program ( www.Harvardsffq.date ). Healthy men and women consumed protein as 19-20% and 17-19% of their total energy intakes, respectively, with animal protein representing about 75 and 70% of their total protein intakes, respectively. The intake of each nutritionally essential amino acid (EAA) by the persons exceeded that recommended for healthy adults with a minimal physical activity. In all individuals, the dietary intake of leucine was the highest, followed by lysine, valine, and isoleucine in descending order, and the ingestion of amino acids that are synthesizable de novo in animal cells (AASAs) was about 20% greater than that of total EAAs. The intake of each AASA met those recommended for healthy adults with a minimal physical activity. Intakes of some AASAs (alanine, arginine, aspartate, glutamate, and glycine) from a typical diet providing 90-110 g food protein/day does not meet the requirements of adults with an intensive physical activity. Within the male or female group, there were not significant differences in the dietary intakes of all amino acids between days 0 and 90 of the study, and this was also true for nearly all other essential nutrients. Our findings will help to improve amino acid nutrition and health in both the general population and exercising individuals.


Asunto(s)
Aminoácidos , Dieta , Adulto , Ingestión de Alimentos , Ingestión de Energía , Femenino , Humanos , Masculino , Nutrientes
7.
Am J Physiol Heart Circ Physiol ; 318(5): H1283-H1295, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32275470

RESUMEN

The lymphatic functions in maintaining lymph transport, and immune surveillance can be impaired by infections and inflammation, thereby causing debilitating disorders, such as lymphedema and inflammatory bowel disease. Histamine is a key inflammatory mediator known to trigger vasodilation and vessel hyperpermeability upon binding to its receptors and evoking intracellular Ca2+ ([Ca2+]i) dynamics for downstream signal transductions. However, the exact molecular mechanisms beneath the [Ca2+]i dynamics and the downstream cellular effects have not been elucidated in the lymphatic system. Here, we show that Ca2+ release-activated Ca2+ (CRAC) channels, formed by Orai1 and stromal interaction molecule 1 (STIM1) proteins, are required for the histamine-elicited Ca2+ signaling in human dermal lymphatic endothelial cells (HDLECs). Blockers or antagonists against CRAC channels, phospholipase C, and H1R receptors can all significantly diminish the histamine-evoked [Ca2+]i dynamics in lymphatic endothelial cells (LECs), while short interfering RNA-mediated knockdown of endogenous Orai1 or STIM1 also abolished the Ca2+ entry upon histamine stimulation in LECs. Furthermore, we find that histamine compromises the lymphatic endothelial barrier function by increasing the intercellular permeability and disrupting vascular endothelial-cadherin integrity, which is remarkably attenuated by CRAC channel blockers. Additionally, the upregulated expression of inflammatory cytokines, IL-6 and IL-8, after histamine stimulation was abolished by silencing Orai1 or STIM1 with RNAi in LECs. Taken together, our data demonstrated the essential role of CRAC channels in mediating the [Ca2+]i signaling and downstream endothelial barrier and inflammatory functions induced by histamine in the LECs, suggesting a promising potential to relieve histamine-triggered vascular leakage and inflammatory disorders in the lymphatics by targeting CRAC channel functions.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Señalización del Calcio , Células Endoteliales/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Calcio/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Histamina/farmacología , Humanos , Interleucina-6/genética , Interleucina-8/genética , Vasos Linfáticos/citología
8.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R590-R604, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913658

RESUMEN

Lymphatic vessels play a critical role in mounting a proper immune response by trafficking peripheral immune cells to draining lymph nodes. Mast cells (MCs) are well known for their roles in type I hypersensitivity reactions, but little is known about their secretory regulation in the lymphatic niche. MCs, as innate sensor and effector cells, reside close to mesenteric lymphatic vessels (MLVs), and their activation and ability to release histamine influences the lymphatic microenvironment in a histamine-NF-κB-dependent manner. Using an established experimental protocol involving surgical isolation of rat mesenteric tissue segments, including MLVs and surrounding perilymphatic tissues, we tested the hypothesis that perilymphatic mesenteric MCs possess histamine receptors (HRs) that bind and respond to the histamine released from these same MCs. Under various experimental conditions, including inflammatory stimulation by LPS, we measured histamine in mesenteric perilymphatic tissues, evaluated expression of histidine decarboxylase in MCs along with the degree of MC degranulation, assessed the functional status of HRs in MCs, and evaluated the ability of histamine itself to induce MC activation. Finally, we evaluated the importance of MCs and HR1 and -2 for MLV-directed trafficking of CD11b/c-positive cells during acute tissue inflammation. Our data indicate the existence of a functionally potent MC-histamine autocrine regulatory loop, the elements of which are crucially important for acute inflammation-induced trafficking of the CD11b/c-positive cells toward MLVs. This MC-histamine loop serves as a first-line cellular servo control system, playing a key role in the innate and adaptive immune response as well as NF-κB-mediated maintenance of body homeostasis.


Asunto(s)
Comunicación Autocrina/fisiología , Inflamación/metabolismo , Mastocitos/metabolismo , Mesenterio/metabolismo , Animales , Histamina/farmacología , Homeostasis/fisiología , Inflamación/fisiopatología , Vasos Linfáticos/metabolismo , Masculino , FN-kappa B/metabolismo , Ratas Sprague-Dawley
9.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G217-G227, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30475062

RESUMEN

This study aimed to establish mechanistic links between the prolonged intake of desloratadine, a common H1 receptor blocker (i.e., antihistamine), and development of obesity and metabolic syndrome. Male Sprague-Dawley rats were treated for 16 wk with desloratadine. We analyzed the dynamics of body weight gain, tissue fat accumulation/density, contractility of isolated mesenteric lymphatic vessels, and levels of blood lipids, glucose, and insulin, together with parameters of liver function. Prolonged intake of desloratadine induced development of an obesity-like phenotype and signs of metabolic syndrome. These alterations in the body included excessive weight gain, increased density of abdominal subcutaneous fat and intracapsular brown fat, high blood triglycerides with an indication of their rerouting toward portal blood, high HDL, high fasting blood glucose with normal fasting and nonfasting insulin levels (insulin resistance), high liver/body weight ratio, and liver steatosis (fatty liver). These changes were associated with dysfunction of mesenteric lymphatic vessels, specifically high lymphatic tone and resistance to flow together with diminished tonic and abolished phasic responses to increases in flow, (i.e., greatly diminished adaptive reserves to respond to postprandial increases in lymph flow). The role of nitric oxide in this flow-dependent adaptation was abolished, with remnants of these responses controlled by lymphatic vessel-derived histamine. Our current data, considered together with reports in the literature, support the notion that millions of the United States population are highly likely affected by underevaluated, lymphatic-related side effects of antihistamines and may develop obesity and metabolic syndrome due to the prolonged intake of this medication. NEW & NOTEWORTHY Prolonged intake of desloratadine induced development of obesity and metabolic syndrome associated with dysfunction of mesenteric lymphatic vessels, high lymphatic tone, and resistance to flow together with greatly diminished adaptive reserves to respond to postprandial increases in lymph flow. Data support the notion that millions of the USA population are highly likely affected by underevaluated, lymphatic-related side effects of antihistamines and may develop obesity and metabolic syndrome due to the prolonged intake of this medication.


Asunto(s)
Hígado Graso/tratamiento farmacológico , Loratadina/análogos & derivados , Vasos Linfáticos/efectos de los fármacos , Síndrome Metabólico/etiología , Obesidad/etiología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Hígado Graso/complicaciones , Resistencia a la Insulina/fisiología , Lípidos/sangre , Loratadina/farmacología , Vasos Linfáticos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Aumento de Peso/efectos de los fármacos
10.
Microcirculation ; 25(7): e12492, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30025187

RESUMEN

OBJECTIVE: Lymphatic vessel dysfunction and increased lymph leakage have been directly associated with several metabolic diseases. However, the underlying cellular mechanisms causing lymphatic dysfunction have not been determined. Aberrant insulin signaling affects the metabolic function of cells and consequently impairs tissue function. We hypothesized that insulin resistance in LECs decreases eNOS activity, disrupts barrier integrity increases permeability, and activates mitochondrial dysfunction and pro-inflammatory signaling pathways. METHODS: LECs were treated with insulin and/or glucose to determine the mechanisms leading to insulin resistance. RESULTS: Acute insulin treatment increased eNOS phosphorylation and NO production in LECs via activation of the PI3K/Akt signaling pathway. Prolonged hyperglycemia and hyperinsulinemia induced insulin resistance in LECs. Insulin-resistant LECs produced less NO due to a decrease in eNOS phosphorylation and showed a significant decrease in impedance across an LEC monolayer that was associated with disruption in the adherence junctional proteins. Additionally, insulin resistance in LECs impaired mitochondrial function by decreasing basal-, maximal-, and ATP-linked OCRs and activated NF-κB nuclear translocation coupled with increased pro-inflammatory signaling. CONCLUSION: Our data provide the first evidence that insulin resistance disrupts endothelial barrier integrity, decreases eNOS phosphorylation and mitochondrial function, and activates inflammation in LECs.


Asunto(s)
Endotelio Linfático/metabolismo , Resistencia a la Insulina , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Endotelio Linfático/patología , Glucosa/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Insulina/farmacología , Uniones Intercelulares/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos
11.
Amino Acids ; 50(9): 1215-1229, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29858688

RESUMEN

Previous studies with animals and humans have shown beneficial effects of dietary supplementation with L-arginine (Arg) on reducing white fat and improving health. At present, a long-term safe level of Arg administration to adult humans is unknown. The objective of this study was to conduct a randomized, placebo-controlled, clinical trial to evaluate the safety and tolerability of oral Arg in overweight or obese but otherwise healthy adults with a body mass index of ≥ 25 kg/m2. A total of 142 subjects completed a 7-day wash-in period using a 12 g Arg/day dose. All the remaining eligible 101 subjects who tolerated the wash-in dose (45 men and 56 women) were assigned randomly to ingest 0, 15 or 30 g Arg (as pharmaceutical-grade Arg-HCl) per day for 90 days. Arg was taken daily in at least two divided doses by mixing with a flavored beverage. At Days 0 and 90, blood pressures of study subjects were recorded, their physical examinations were performed, and their blood and 24-h urine samples were obtained to measure: (1) serum concentrations of amino acids, glucose, fatty acids, and related metabolites; and (2) renal, hepatic, endocrine and metabolic parameters. Our results indicate that the serum concentration of Arg in men or women increased (P < 0.05) progressively with increasing oral Arg doses from 0 to 30 g/day. Dietary supplementation with 30 g Arg/day reduced (P < 0.05) systolic blood pressure and serum glucose concentration in females, as well as serum concentrations of free fatty acids in both males and females. Based on physiological and biochemical variables, study subjects tolerated oral administration of 15 and 30 g Arg/day without adverse events. We conclude that a long-term safe level of dietary Arg supplementation is at least 30 g/day in adult humans.


Asunto(s)
Arginina/administración & dosificación , Suplementos Dietéticos/análisis , Adulto , Aminoácidos/sangre , Arginina/efectos adversos , Arginina/sangre , Presión Sanguínea/efectos de los fármacos , Suplementos Dietéticos/efectos adversos , Ácidos Grasos/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
12.
Int J Mol Sci ; 18(5)2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28467354

RESUMEN

This review provides a comprehensive summary of research on aging-associated alterations in lymphatic vessels and mast cells in perilymphatic tissues. Aging alters structure (by increasing the size of zones with low muscle cell investiture), ultrastructure (through loss of the glycocalyx), and proteome composition with a concomitant increase in permeability of aged lymphatic vessels. The contractile function of aged lymphatic vessels is depleted with the abolished role of nitric oxide and an increased role of lymphatic-born histamine in flow-dependent regulation of lymphatic phasic contractions and tone. In addition, aging induces oxidative stress in lymphatic vessels and facilitates the spread of pathogens from these vessels into perilymphatic tissues. Aging causes the basal activation of perilymphatic mast cells, which, in turn, restricts recruitment/activation of immune cells in perilymphatic tissues. This aging-associated basal activation of mast cells limits proper functioning of the mast cell/histamine/NF-κB axis that is essential for the regulation of lymphatic vessel transport and barrier functions as well as for both the interaction and trafficking of immune cells near and within lymphatic collecting vessels. Cumulatively, these changes play important roles in the pathogenesis of alterations in inflammation and immunity associated with aging.


Asunto(s)
Envejecimiento/fisiología , Inmunidad/inmunología , Inflamación/inmunología , Vasos Linfáticos/fisiología , Tejido Linfoide/fisiología , Animales , Histamina/metabolismo , Humanos , Mastocitos/metabolismo , Ratones , FN-kappa B/metabolismo , Ratas
13.
J Nutr ; 146(12): 2587S-2593S, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27934649

RESUMEN

l-Arginine (Arg) appears to have a beneficial effect on the regulation of nutrient metabolism to enhance lean tissue deposition and on insulin resistance in humans. The observed safe level for oral administration of Arg is ∼20 g/d, but higher levels have been tested in short-term studies without serious adverse effects; however, more data are needed in both animal models and humans to fully evaluate safety as well as efficacy. The primary objective of this review is to summarize the current knowledge of the safety, pharmacokinetics, and effectiveness of oral Arg in adults. Arg supplementation has been used safely in vulnerable populations, such as pregnant women, preterm infants, and individuals with cystic fibrosis. Several recent studies have shown beneficial effects of Arg in individuals with obesity, insulin resistance, and diabetes. Collectively, the data suggest that Arg supplementation is a safe and generally well-tolerated nutriceutical that may improve metabolic profiles in humans.


Asunto(s)
Arginina/efectos adversos , Arginina/farmacología , Administración Oral , Adulto , Arginina/farmacocinética , Humanos
14.
Amino Acids ; 48(10): 2401-10, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27180260

RESUMEN

Polyamines are essential for proliferation of endothelial cells (EC) and angiogenesis. This study was conducted to identify the metabolic source(s) of ornithine for polyamine synthesis in EC, using N(ω)-hydroxy-nor-L-arginine (Nor-NOHA, an inhibitor of arginase) and gabaculine (an inhibitor of ornithine aminotransferase; OAT). Nor-NOHA inhibited arginase with an IC50 value of 10 µM for intact EC. Nor-NOHA (0.5 mM) alone inhibited arginase activity in EC by 98 %, increased total cellular concentrations of arginine by 14 %, and decreased total cellular concentrations of ornithine, putrescine and spermidine by 17, 65 and 74 %, respectively. Arginine and glutamine contributed to 73 and 26 % of the ornithine produced by EC, respectively. Gabaculine (1 mM) alone decreased the total cellular concentrations of arginine, ornithine, putrescine, and spermidine by 14, 96, 32, and 42 %, respectively. A combination of both Nor-NOHA and gabaculine completely blocked ornithine production in EC, resulting in no detectable cellular ornithine and almost complete depletion of cellular putrescine and spermidine. Addition of 0.5 mM ornithine restored the intracellular concentrations of polyamines in EC treated with Nor-NOHA plus gabaculine, indicating that Nor-NOHA and gabaculine did not inhibit ornithine decarboxylase activity. Our results suggest that the arginase and OAT pathways are the exclusive sources of ornithine in EC when there is little extracellular ornithine and that there is intracellular compartmentalization of arginine and ornithine for endothelial synthesis of polyamines.  These novel findings may have important implications for improving placental vascular growth, wound healing, and cancer therapy.


Asunto(s)
Células Endoteliales/metabolismo , Ornitina/metabolismo , Putrescina/biosíntesis , Espermidina/biosíntesis , Animales , Arginina/análogos & derivados , Arginina/farmacología , Bovinos , Células Cultivadas , Ácidos Ciclohexanocarboxílicos/farmacología
15.
Amino Acids ; 48(7): 1541-52, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27156062

RESUMEN

L-arginine (Arg) is utilized via multiple pathways to synthesize protein and low-molecular-weight bioactive substances (e.g., nitric oxide, creatine, and polyamines) with enormous physiological importance. Furthermore, Arg regulates cell signaling pathways and gene expression to improve cardiovascular function, augment insulin sensitivity, enhance lean tissue mass, and reduce obesity in humans. Despite its versatile roles, the use of Arg as a dietary supplement is limited due to the lack of data to address concerns over its safety in humans. Data from animal studies are reviewed to assess arginine catabolism and the safety of long-term Arg supplementation. The arginase pathway was responsible for catabolism of 76-85 and 81-96 % Arg in extraintestinal tissues of pigs and rats, respectively. Dietary supplementation with Arg-HCl or the Arg base [315- and 630-mg Arg/(kg BW d) for 91 d] had no adverse effects on male or female pigs. Similarly, no safety issues were observed for male or female rats receiving supplementation with 1.8- and 3.6-g Arg/(kg BW d) for at least 91 d. Intravenous administration of Arg-HCl to gestating sheep at 81 and 180 mg Arg/(kg BW d) is safe for at least 82 and 40 d, respectively. Animals fed conventional diets can well tolerate large amounts of supplemental Arg [up to 630-mg Arg/(kg BW d) in pigs or 3.6-g Arg/(kg BW d) in rats] for 91 d, which are equivalent to 573-mg Arg/(kg BW d) for humans. Collectively, these results can help guide studies to determine the safety of long-term oral administration of Arg in humans.


Asunto(s)
Arginasa/metabolismo , Arginina/metabolismo , Arginina/farmacología , Suplementos Dietéticos , Animales , Arginina/efectos adversos , Humanos , Ratas , Ovinos , Porcinos
16.
Amino Acids ; 47(3): 435-47, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552397

RESUMEN

Reduced availability of nitric oxide (NO) in the vasculature is a major factor contributing to the impaired action of insulin on blood flow and, therefore, insulin resistance in obese and diabetic subjects. Available evidence shows that vascular insulin resistance plays an important role in the pathogenesis of cardiovascular disease, the leading cause of death in developed nations. Interestingly, increased concentrations of L-leucine in the plasma occur in obese humans and other animals with vascular dysfunction. Among branched-chain amino acids, L-leucine is unique in inhibiting NO synthesis from L-arginine in endothelial cells and may modulate cardiovascular homeostasis in insulin resistance. Results of recent studies indicate that L-leucine is an activator of glutamine:fructose-6-phosphate aminotransferase (GFAT), which is the first and a rate-controlling enzyme in the synthesis of glucosamine (an inhibitor of endothelial NO synthesis). Through stimulating the mammalian target of rapamycin signaling pathway and thus protein synthesis, L-leucine may enhance GFAT protein expression, thereby inhibiting NO synthesis in endothelial cells. We propose that reducing circulating levels of L-leucine or endothelial GFAT activity may provide a potentially novel strategy for preventing and/or treating cardiovascular disease in obese and diabetic subjects. Such means may include dietary supplementation with either α-ketoglutarate to enhance the catabolism of L-leucine in the small intestine and other tissues or with N-ethyl-L-glutamine to inhibit GFAT activity in endothelial cells. Preventing leucine-induced activation of GFAT by nutritional supplements or pharmaceutical drugs may contribute to improved cardiovascular function by enhancing vascular NO synthesis.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Diabetes Mellitus/sangre , Resistencia a la Insulina , Leucina/sangre , Óxido Nítrico/sangre , Obesidad/sangre , Animales , Endotelio Vascular/metabolismo , Humanos , Insulina/sangre , Transferasas de Grupos Nitrogenados/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
17.
Amino Acids ; 47(9): 1909-20, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25948162

RESUMEN

This study was conducted with rats to determine the safety of long-term dietary supplementation with L-arginine. Beginning at 6 weeks of age, male and female rats were fed a casein-based semi-purified diet containing 0.61 % L-arginine and received drinking water containing L-arginine-HCl (0, 1.8, or 3.6 g L-arginine/kg body-weight/day; n = 10/group). These supplemental doses of L-arginine were equivalent to 0, 286, and 573 mg L-arginine/kg body-weight/day, respectively, in humans. After a 13-week supplementation period, blood samples were obtained from rats for biochemical analyses. Supplementation with L-arginine increased plasma concentrations of arginine, ornithine, proline, homoarginine, urea, and nitric oxide metabolites without affecting those for lysine, histidine, or methylarginines, while reducing plasma concentrations of ammonia, glutamine, free fatty acids, and triglycerides. L-Arginine supplementation enhanced protein gain and reduced white-fat deposition in the body. Based on general appearance, feeding behavior, and physiological parameters, all animals showed good health during the entire experimental period; Plasma concentrations of all measured hormones (except leptin) did not differ between control and arginine-supplemented rats. L-Arginine supplementation reduced plasma levels of leptin. Additionally, L-arginine supplementation increased L-arginine:glycine amidinotransferase activity in kidneys but not in the liver or small intestine, suggesting tissue-specific regulation of enzyme expression by L-arginine. Collectively, these results indicate that dietary supplementation with L-arginine (e.g., 3.6 g/kg body-weight/day) is safe in rats for at least 91 days. This dose is equivalent to 40 g L-arginine/kg body-weight/day for a 70-kg person. Our findings help guide clinical studies to determine the safety of long-term oral administration of L-arginine to humans.


Asunto(s)
Adiposidad/efectos de los fármacos , Arginina/farmacología , Caseínas/farmacología , Proteínas en la Dieta/farmacología , Suplementos Dietéticos , Metabolismo de los Lípidos/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Arginina/efectos adversos , Caseínas/efectos adversos , Proteínas en la Dieta/efectos adversos , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
18.
Amino Acids ; 47(5): 925-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25655382

RESUMEN

This study was conducted with a swine model to determine the safety of long-term dietary supplementation with L-arginine-HCl or L-arginine free base. Beginning at 30 days of age, pigs were fed a corn- and soybean meal-based diet (31.5 g/kg body weight/day) supplemented with 0, 1.21, 1.81 or 2.42 % L-arginine-HCl (Experiment 1) or with 0, 1, 1.5 or 2 % L-arginine (Experiment 2). The supplemental doses of 0, 1, 1.5, and 2 % L-arginine provided pigs with 0, 315, 473, and 630 mg L-arginine/kg body weight/day, respectively, which were equivalent to 0, 286, 430, and 573 mg L-arginine/kg body weight/day, respectively, in humans. At 121 days of age (91 days after initiation of supplementation), blood samples were obtained from the jugular vein of pigs at 1 and 4 h after feeding for hematological and clinical chemistry tests. Dietary supplementation with L-arginine increased plasma concentrations of arginine, ornithine, proline, albumin and reticulocytes, while reducing plasma concentrations of ammonia, free fatty acids, triglyceride, cholesterol, and neutrophils. L-Arginine supplementation enhanced protein gain and reduced white-fat deposition in the body. Other variables in standard hematology and clinical chemistry tests, serum concentrations of insulin, growth hormone and insulin-like growth factor-I did not differ among all the groups of pigs. These results indicate that dietary supplementation with L-arginine (up to 630 mg/kg body weight/day) is safe in pigs for at least 91 days. Our findings help guide clinical studies to determine the safety of long-term oral administration of L-arginine to humans.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Arginina/administración & dosificación , Composición Corporal/efectos de los fármacos , Suplementos Dietéticos , Músculo Esquelético/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Alanina/sangre , Amoníaco/sangre , Animales , Animales Lactantes , Arginina/sangre , Colesterol/sangre , Femenino , Glutamina/sangre , Glicina/sangre , Masculino , Músculo Esquelético/metabolismo , Ornitina/sangre , Prolina/sangre , Porcinos , Factores de Tiempo , Triglicéridos/sangre , Destete
19.
Microcirculation ; 21(7): 640-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24750494

RESUMEN

OBJECTIVES: The knowledge of the basic principles of lymphatic function, still remains, to a large degree, rudimentary and will require significant research efforts. Recent studies of the physiology of the MLVs suggested the presence of an EDRF other than NO. In this study, we tested the hypothesis that lymphatic endothelium-derived histamine relaxes MLVs. METHODS: We measured and analyzed parameters of lymphatic contractility in isolated and pressurized rat MLVs under control conditions and after pharmacological blockade of NO by L-NAME (100 µM) or/and histamine production by α-MHD (10 µM). Effectiveness of α-MHD was confirmed immunohistochemically. We also used immunohistochemical labeling and Western blot analysis of the histamine-producing enzyme, HDC. In addition, we blocked HDC protein expression in MLVs by transient transfection with vivo-morpholino oligos. RESULTS: We found that only combined pharmacological blockade of NO and histamine production completely eliminates flow-dependent relaxation of lymphatic vessels, thus confirming a role for histamine as an EDRF in MLVs. We also confirmed the presence of HDC and histamine inside lymphatic endothelial cells. CONCLUSIONS: This study supports a role for histamine as an EDRF in MLVs.


Asunto(s)
Endotelio Linfático/fisiología , Histamina/fisiología , Vasos Linfáticos/fisiología , Óxido Nítrico/fisiología , Animales , Endotelio Linfático/citología , Endotelio Linfático/efectos de los fármacos , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/fisiología , Histamina/análisis , Histidina Descarboxilasa/fisiología , Vasos Linfáticos/efectos de los fármacos , Masculino , Mesenterio , Metilhistidinas/farmacología , Morfolinos/farmacología , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/antagonistas & inhibidores , Ratas , Ratas Endogámicas F344 , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/fisiología , Guanilil Ciclasa Soluble
20.
Circ Res ; 111(5): 553-63, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22777005

RESUMEN

RATIONALE: Endothelial adherens junction proteins constitute an important element in the control of microvascular permeability. Platelet-activating factor (PAF) increases permeability to macromolecules via translocation of endothelial nitric oxide synthase (eNOS) to cytosol and stimulation of eNOS-derived nitric oxide signaling cascade. The mechanisms by which nitric oxide signaling regulates permeability at adherens junctions are still incompletely understood. OBJECTIVE: We explored the hypothesis that PAF stimulates hyperpermeability via S-nitrosation (SNO) of adherens junction proteins. METHODS AND RESULTS: We measured PAF-stimulated SNO of ß-catenin and p120-catenin (p120) in 3 cell lines: ECV-eNOSGFP, EAhy926 (derived from human umbilical vein), and postcapillary venular endothelial cells (derived from bovine heart endothelium) and in the mouse cremaster muscle in vivo. SNO correlated with diminished abundance of ß-catenin and p120 at the adherens junction and with hyperpermeability. Tumor necrosis factor-α increased nitric oxide production and caused similar increase in SNO as PAF. To ascertain the importance of eNOS subcellular location in this process, we used ECV-304 cells transfected with cytosolic eNOS (GFPeNOSG2A) and plasma membrane eNOS (GFPeNOSCAAX). PAF induced SNO of ß-catenin and p120 and significantly diminished association between these proteins in cells with cytosolic eNOS but not in cells wherein eNOS is anchored to the cell membrane. Inhibitors of nitric oxide production and of SNO blocked PAF-induced SNO and hyperpermeability, whereas inhibition of the cGMP pathway had no effect. Mass spectrometry analysis of purified p120 identified cysteine 579 as the main S-nitrosated residue in the region that putatively interacts with vascular endothelial-cadherin. CONCLUSIONS: Our results demonstrate that agonist-induced SNO contributes to junctional membrane protein changes that enhance endothelial permeability.


Asunto(s)
Uniones Adherentes/metabolismo , Permeabilidad Capilar/fisiología , Cateninas/metabolismo , Células Endoteliales/metabolismo , Transducción de Señal/fisiología , beta Catenina/metabolismo , Secuencia de Aminoácidos , Animales , Permeabilidad Capilar/efectos de los fármacos , Cateninas/genética , Bovinos , Proteínas Fluorescentes Verdes/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitrosación/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Transducción de Señal/efectos de los fármacos , Vénulas/citología , Catenina delta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA