Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Nitric Oxide ; 148: 13-22, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38642795

ABSTRACT

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.


Subject(s)
Biopterins , Diabetes Mellitus, Experimental , Endothelium, Vascular , Nanoparticles , Animals , Biopterins/analogs & derivatives , Biopterins/pharmacology , Biopterins/administration & dosage , Biopterins/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Male , Nanoparticles/chemistry , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism
2.
Exp Biol Med (Maywood) ; 248(18): 1537-1549, 2023 09.
Article in English | MEDLINE | ID: mdl-37837386

ABSTRACT

This study tested the hypothesis that elevated L-leucine concentrations in plasma reduce nitric oxide (NO) synthesis by endothelial cells (ECs) and affect adiposity in obese rats. Beginning at four weeks of age, male Sprague-Dawley rats were fed a casein-based low-fat (LF) or high-fat (HF) diet for 15 weeks. Thereafter, rats in the LF and HF groups were assigned randomly into one of two subgroups (n = 8/subgroup) and received drinking water containing either 1.02% L-alanine (isonitrogenous control) or 1.5% L-leucine for 12 weeks. The energy expenditure of the rats was determined at weeks 0, 6, and 11 of the supplementation period. At the end of the study, an oral glucose tolerance test was performed on all the rats immediately before being euthanized for the collection of tissues. HF feeding reduced (P < 0.001) NO synthesis in ECs by 21% and whole-body insulin sensitivity by 19% but increased (P < 0.001) glutamine:fructose-6-phosphate transaminase (GFAT) activity in ECs by 42%. Oral administration of L-leucine decreased (P < 0.05) NO synthesis in ECs by 14%, increased (P < 0.05) GFAT activity in ECs by 35%, and reduced (P < 0.05) whole-body insulin sensitivity by 14% in rats fed the LF diet but had no effect (P > 0.05) on these variables in rats fed the HF diet. L-Leucine supplementation did not affect (P > 0.05) weight gain, tissue masses (including white adipose tissue, brown adipose tissue, and skeletal muscle), or antioxidative capacity (indicated by ratios of glutathione/glutathione disulfide) in LF- or HF-fed rats and did not worsen (P > 0.05) adiposity, whole-body insulin sensitivity, or metabolic profiles in the plasma of obese rats. These results indicate that high concentrations of L-leucine promote glucosamine synthesis and impair NO production by ECs, possibly contributing to an increased risk of cardiovascular disease in diet-induced obese rats.


Subject(s)
Insulin Resistance , Rats , Male , Animals , Leucine/pharmacology , Nitric Oxide , Rats, Sprague-Dawley , Endothelial Cells/metabolism , Obesity/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements
3.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982254

ABSTRACT

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.


Subject(s)
Carbon Dioxide , Hypoxia , Humans , Carbon Dioxide/metabolism , Oxygen/metabolism , Exercise , Homeostasis
4.
Amino Acids ; 54(2): 193-204, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34741684

ABSTRACT

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.


Subject(s)
Angiogenic Proteins , Placenta , Angiogenic Proteins/metabolism , Animals , Arginine/metabolism , Arginine/pharmacology , Dietary Supplements , Endothelial Cells/metabolism , Female , Placenta/metabolism , Placenta Growth Factor/metabolism , Polyamines/metabolism , Pregnancy , Sus scrofa/metabolism , Swine , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Adv Exp Med Biol ; 1332: 211-227, 2021.
Article in English | MEDLINE | ID: mdl-34251646

ABSTRACT

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.


Subject(s)
Amino Acids , Diet , Adult , Eating , Energy Intake , Female , Humans , Male , Nutrients
6.
Adv Exp Med Biol ; 1332: 167-187, 2021.
Article in English | MEDLINE | ID: mdl-34251644

ABSTRACT

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.


Subject(s)
COVID-19 , Nitric Oxide , Animals , Arginine/metabolism , Female , Humans , Male , Pregnancy , Protein Biosynthesis , SARS-CoV-2
7.
Front Immunol ; 11: 1234, 2020.
Article in English | MEDLINE | ID: mdl-32625213

ABSTRACT

Mast cells (MCs) are abundant in almost all vascularized tissues. Furthermore, their anatomical proximity to lymphatic vessels and their ability to synthesize, store and release a large array of inflammatory and vasoactive mediators emphasize their significance in the regulation of the lymphatic vascular functions. As a major secretory cell of the innate immune system, MCs maintain their steady-state granule release under normal physiological conditions; however, the inflammatory response potentiates their ability to synthesize and secrete these mediators. Activation of MCs in response to inflammatory signals can trigger adaptive immune responses by dendritic cell-directed T cell activation. In addition, through the secretion of various mediators, cytokines and growth factors, MCs not only facilitate interaction and migration of immune cells, but also influence lymphatic permeability, contractility, and vascular remodeling as well as immune cell trafficking through the lymphatic vessels. In summary, the consequences of these events directly affect the lymphatic niche, influencing inflammation at multiple levels. In this review, we have summarized the recent advancements in our understanding of the MC biology in the context of the lymphatic vascular system. We have further highlighted the MC-lymphatic interaction axis from the standpoint of the tumor microenvironment.


Subject(s)
Immunomodulation , Lymphatic System/physiology , Mast Cells/immunology , Mast Cells/metabolism , Cell Plasticity/immunology , Disease Susceptibility , Homeostasis , Humans , Inflammation/etiology , Inflammation/metabolism , Lymphatic Vessels/physiology , Neoplasms/etiology , Neoplasms/metabolism , Organ Specificity/immunology
8.
Am J Physiol Heart Circ Physiol ; 318(5): H1283-H1295, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32275470

ABSTRACT

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.


Subject(s)
Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Endothelial Cells/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Calcium/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Histamine/pharmacology , Humans , Interleukin-6/genetics , Interleukin-8/genetics , Lymphatic Vessels/cytology
9.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R590-R604, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913658

ABSTRACT

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.


Subject(s)
Autocrine Communication/physiology , Inflammation/metabolism , Mast Cells/metabolism , Mesentery/metabolism , Animals , Histamine/pharmacology , Homeostasis/physiology , Inflammation/physiopathology , Lymphatic Vessels/metabolism , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley
11.
Proc Natl Acad Sci U S A ; 116(24): 11590-11595, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31138695

ABSTRACT

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.


Subject(s)
Maternal Exposure/adverse effects , Metabolic Syndrome/chemically induced , Organogenesis/drug effects , Particulate Matter/adverse effects , Prenatal Exposure Delayed Effects/pathology , Air Pollution/adverse effects , Animals , Birth Weight/drug effects , Disease Susceptibility/blood , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Environmental Exposure/adverse effects , Fatty Acids/blood , Female , Glucose/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Organogenesis/physiology , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G217-G227, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30475062

ABSTRACT

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.


Subject(s)
Fatty Liver/drug therapy , Loratadine/analogs & derivatives , Lymphatic Vessels/drug effects , Metabolic Syndrome/etiology , Obesity/etiology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Fatty Liver/complications , Insulin Resistance/physiology , Lipids/blood , Loratadine/pharmacology , Lymphatic Vessels/metabolism , Male , Rats , Rats, Sprague-Dawley , Weight Gain/drug effects
13.
Microcirculation ; 25(7): e12492, 2018 10.
Article in English | MEDLINE | ID: mdl-30025187

ABSTRACT

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.


Subject(s)
Endothelium, Lymphatic/metabolism , Insulin Resistance , Animals , Cell Membrane Permeability/drug effects , Endothelium, Lymphatic/pathology , Glucose/pharmacology , Humans , Inflammation/chemically induced , Inflammation/metabolism , Insulin/pharmacology , Intercellular Junctions/drug effects , Mitochondria/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Signal Transduction/drug effects
14.
Amino Acids ; 50(9): 1215-1229, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29858688

ABSTRACT

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.


Subject(s)
Arginine/administration & dosage , Dietary Supplements/analysis , Adult , Amino Acids/blood , Arginine/adverse effects , Arginine/blood , Blood Pressure/drug effects , Dietary Supplements/adverse effects , Fatty Acids/blood , Female , Humans , Male , Middle Aged , Young Adult
15.
Int J Mol Sci ; 18(5)2017 May 03.
Article in English | MEDLINE | ID: mdl-28467354

ABSTRACT

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.


Subject(s)
Aging/physiology , Immunity/immunology , Inflammation/immunology , Lymphatic Vessels/physiology , Lymphoid Tissue/physiology , Animals , Histamine/metabolism , Humans , Mast Cells/metabolism , Mice , NF-kappa B/metabolism , Rats
16.
Lymphat Res Biol ; 15(2): 136-145, 2017 06.
Article in English | MEDLINE | ID: mdl-28453392

ABSTRACT

BACKGROUND: Knowledge of the mechanisms by which aging affects contracting lymphatic vessels remains incomplete; therefore, the functional role of histamine in the reaction of aged lymphatic vessels to increases in flow remains unknown. METHODS AND RESULTS: We measured and analyzed parameters of lymphatic contractility in isolated and pressurized rat mesenteric lymphatic vessels (MLVs) obtained from 9- and 24-month Fischer-344 rats under control conditions and after pharmacological blockade of nitric oxide (NO) by Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME, 100 µM) or/and blockade of histamine production by α-methyl-DL-histidine dihydrochloride (α-MHD, 10 µM). We also quantitatively compared results of immunohistochemical labeling of the histamine-producing enzyme, histidine decarboxylase (HDC) in adult and aged MLVs. Our data provide the first demonstration of an increased functional role of histamine as an endothelial-derived relaxing factor in aged MLVs, which appears in parallel with the abolished role of NO in the reactions of these lymph vessels to increases in flow. In addition, we found an increased expression of HDC in endothelium of aged MLVs. CONCLUSIONS: Our findings provide the basis for better understanding of the processes of aging in lymphatic vessels and for setting new important directions for investigations of the aging-associated disturbances in lymph flow and the immune response.


Subject(s)
Endothelium, Vascular/metabolism , Histamine/metabolism , Lymphatic Vessels/metabolism , Mesentery , Age Factors , Animals , Blood Pressure , Endothelial Cells/metabolism , Gene Expression , Histamine/pharmacology , Immunohistochemistry , Male , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Nitric Oxide/metabolism , Rats , Shear Strength
17.
J Nutr ; 146(12): 2587S-2593S, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27934649

ABSTRACT

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.


Subject(s)
Arginine/adverse effects , Arginine/pharmacology , Administration, Oral , Adult , Arginine/pharmacokinetics , Humans
18.
Aging (Albany NY) ; 8(11): 3065-3090, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27875806

ABSTRACT

This study aimed to establish mechanistic links between the aging-associated changes in the functional status of mast cells and the altered responses of mesenteric tissue and mesenteric lymphatic vessels (MLVs) to acute inflammation. We used an in vivo model of acute peritoneal inflammation induced by lipopolysaccharide treatment of adult (9-month) and aged (24-month) F-344 rats. We analyzed contractility of isolated MLVs, mast cell activation, activation of nuclear factor-κB (NF-κB) without and with stabilization of mast cells by cromolyn or blockade of all types of histamine receptors and production of 27 major pro-inflammatory cytokines in adult and aged perilymphatic mesenteric tissues and blood. We found that the reactivity of aged contracting lymphatic vessels to LPS-induced acute inflammation was abolished and that activated mast cells trigger NF-κB signaling in the mesentery through release of histamine. The aging-associated basal activation of mesenteric mast cells limits acute inflammatory NF-κB activation in aged mesentery. We conclude that proper functioning of the mast cell/histamine/NF-κB axis is necessary for reactions of the lymphatic vessels to acute inflammatory stimuli as well as for interaction and trafficking of immune cells near and within the collecting lymphatics.


Subject(s)
Cytokines/metabolism , Histamine/metabolism , Inflammation/metabolism , Mast Cells/metabolism , NF-kappa B/metabolism , Peritoneal Diseases/metabolism , Animals , Cromolyn Sodium/pharmacology , Inflammation/chemically induced , Inflammation Mediators/metabolism , Lipopolysaccharides , Lymphatic Vessels/metabolism , Male , Mesentery/metabolism , Peritoneal Diseases/chemically induced , Rats , Rats, Inbred F344
19.
Amino Acids ; 48(7): 1541-52, 2016 07.
Article in English | MEDLINE | ID: mdl-27156062

ABSTRACT

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.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Arginine/pharmacology , Dietary Supplements , Animals , Arginine/adverse effects , Humans , Rats , Sheep , Swine
20.
Amino Acids ; 48(10): 2401-10, 2016 10.
Article in English | MEDLINE | ID: mdl-27180260

ABSTRACT

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.


Subject(s)
Endothelial Cells/metabolism , Ornithine/metabolism , Putrescine/biosynthesis , Spermidine/biosynthesis , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cattle , Cells, Cultured , Cyclohexanecarboxylic Acids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...