Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Amino Acids ; 47(9): 1909-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25948162

ABSTRACT

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.


Subject(s)
Adiposity/drug effects , Arginine/pharmacology , Caseins/pharmacology , Dietary Proteins/pharmacology , Dietary Supplements , Lipid Metabolism/drug effects , Adipose Tissue, White/metabolism , Animals , Arginine/adverse effects , Caseins/adverse effects , Dietary Proteins/adverse effects , Female , Male , Rats , Rats, Sprague-Dawley , Time Factors
8.
Amino Acids ; 47(5): 925-36, 2015 May.
Article in English | MEDLINE | ID: mdl-25655382

ABSTRACT

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.


Subject(s)
Adipose Tissue, White/drug effects , Arginine/administration & dosage , Body Composition/drug effects , Dietary Supplements , Muscle, Skeletal/drug effects , Adipose Tissue, White/metabolism , Alanine/blood , Ammonia/blood , Animals , Animals, Suckling , Arginine/blood , Cholesterol/blood , Female , Glutamine/blood , Glycine/blood , Male , Muscle, Skeletal/metabolism , Ornithine/blood , Proline/blood , Swine , Time Factors , Triglycerides/blood , Weaning
9.
Amino Acids ; 47(3): 435-47, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552397

ABSTRACT

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.


Subject(s)
Cardiovascular Diseases/blood , Diabetes Mellitus/blood , Insulin Resistance , Leucine/blood , Nitric Oxide/blood , Obesity/blood , Animals , Endothelium, Vascular/metabolism , Humans , Insulin/blood , Nitrogenous Group Transferases/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
Melanoma Res ; 24(6): 556-67, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25304236

ABSTRACT

Metastatic melanoma is a deadly form of cancer with few therapeutic options and the cause of more than 9480 deaths annually in the USA alone. Novel treatment options for this disease are urgently needed. Here we test the efficacy of a novel melanoma drug, the human recombinant Co-arginase (CoArgIPEG), against an aggressive A375 melanoma mouse model. CoArgIPEG is a modification of the naturally occurring human enzyme with improved stability, catalytic activity, and potentially lower immunogenicity compared with current amino acid-depleting drugs. Marked tumor growth reductions (mean P=0.0057) with apoptosis induction and proliferation inhibition are noted with CoArgIPEG treatment, both in the presence and in the absence of supplemental citrulline. Further, improved therapeutic efficacy has been noted against A375 xenografts relative to the naturally occurring human recombinant arginase enzyme at lower doses of CoArgIPEG. Unfortunately, after 1 month, half of the relapsing tumors showed argininosuccinate synthase induction, which correlated with Ser62-phosphorylated cMyc. Although argininosuccinate synthase induction could not be induced in vitro, a drug targeting pathway previously demonstrated to be associated with Ser62 cMyc phosphorylation - U0126 - in combination with CoArgIPEG demonstrated an in-vitro synergistic response (combination indices 0.13±0.10 and 0.14±0.10 with or without citrulline, respectively). Overall, favorable efficacy and potential synergy with other antimelanoma drugs support CoArgIPEG as a potent, novel cancer therapeutic.


Subject(s)
Antineoplastic Agents/therapeutic use , Arginase/therapeutic use , Melanoma/drug therapy , Recombinant Proteins/therapeutic use , Skin Neoplasms/drug therapy , Animals , Cobalt/chemistry , Cobalt/therapeutic use , Female , Humans , Hydrolases/chemistry , Hydrolases/therapeutic use , Jurkat Cells , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Biofactors ; 39(5): 552-63, 2013.
Article in English | MEDLINE | ID: mdl-23804503

ABSTRACT

Male Zucker diabetic fatty (ZDF) rats were used to study effects of oral administration of interferon tau (IFNT) in reducing obesity. Eighteen ZDF rats (28 days of age) were assigned randomly to receive 0, 4, or 8 µg IFNT/kg body weight (BW) per day (n = 6/group) for 8 weeks. Water consumption was measured every two days. Food intake and BW were recorded weekly. Energy expenditure in 4-, 6-, 8-, and 10-week-old rats was determined using indirect calorimetry. Starting at 7 weeks of age, urinary glucose, and ketone bodies were tested daily. Rates of glucose and oleate oxidation in liver, brown adipose tissue, and abdominal adipose tissue, as well as leucine catabolism in skeletal muscle, and lipolysis in white and brown adipose tissues were greater for rats treated with 8 µg IFNT/kg BW/day in comparison with control rats. Treatment with 8 µg IFNT/kg BW/day increased heat production, reduced BW gain and adiposity, ameliorated fatty liver syndrome, delayed the onset of diabetes, and decreased concentrations of glucose, free fatty acids, triacylglycerol, cholesterol, and branched-chain amino acids in plasma, compared with control rats. Oral administration of 8 µg IFNT/kg BW/day ameliorated oxidative stress in skeletal muscle, liver, and adipose tissue, as indicated by decreased ratios of oxidized glutathione to reduced glutathione and increased concentrations of tetrahydrobiopterin. These results indicate that IFNT stimulates oxidation of energy substrates and reduces obesity in ZDF rats and may have broad important implications for preventing and treating obesity-related diseases in mammals.


Subject(s)
Adiposity/drug effects , Anti-Obesity Agents/administration & dosage , Diabetes Mellitus, Type 2/prevention & control , Interferon Type I/administration & dosage , Obesity/drug therapy , Pregnancy Proteins/administration & dosage , Adiponectin/blood , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Administration, Oral , Amino Acids/blood , Animals , Blood Glucose , Diabetes Mellitus, Type 2/blood , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Glycerol/metabolism , Insulin/blood , Leptin/blood , Leucine Transaminase/metabolism , Lipids/blood , Liver/drug effects , Liver/pathology , Male , Obesity/blood , Obesity/pathology , Organ Size , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Zucker
12.
Biofactors ; 39(4): 383-91, 2013.
Article in English | MEDLINE | ID: mdl-23553707

ABSTRACT

Nitric oxide (NO) is a signaling molecule synthesized from L-arginine by NO synthase in animals. Increasing evidence shows that NO regulates the mammalian metabolism of energy substrates and that these effects of NO critically depend on its concentrations at the reaction site and the period of exposure. High concentrations of NO (in the micromolar range) irreversibly inhibit complexes I, II, III, IV, and V in the mitochondrial respiratory chain, whereas physiological levels of NO (in the nanomolar range) reversibly reduce cytochomrome oxidase. Thus, NO reduces oxygen consumption by isolated mitochondria to various extents. In intact cells, through cGMP and AMP-activated protein kinase signaling, physiological levels of NO acutely stimulate uptake and oxidation of glucose and fatty acids by skeletal muscle, heart, liver, and adipose tissue, while inhibiting the synthesis of glucose, glycogen and fat in the insulin-sensitive tissues, and enhancing lipolysis in white adipocytes. Chronic effects of physiological levels of NO in vivo include stimulation of angiogenesis, blood flow, mitochondrial biogenesis, and brown adipocyte development. Modulation of NO-mediated pathways through dietary supplementation with L-arginine or its precursor L-citrulline may provide an effective, practical strategy to prevent and treat metabolic syndrome, including obesity, diabetes, and dyslipidemia in mammals, including humans.


Subject(s)
Energy Metabolism , Nitric Oxide/physiology , Adipose Tissue, Brown/physiopathology , Animals , Cell Respiration , Humans , Mitochondria/metabolism , Obesity/metabolism , Obesity/therapy , Oxidation-Reduction , Regional Blood Flow , Signal Transduction
13.
Curr Opin Clin Nutr Metab Care ; 16(1): 76-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23164986

ABSTRACT

PURPOSE OF REVIEW: The endothelial isoform of nitric oxide synthase (eNOS) is constitutively expressed but dynamically regulated by a number of factors. Building our knowledge of this regulation is necessary to understand and modulate the bioavailability of nitric oxide, central to the cardiovascular complications of diabetes and other diseases. This review will focus on the eNOS substrate (L-arginine), its cofactor (tetrahydrobiopterin), and mechanisms related to the uncoupling of eNOS activity. RECENT FINDINGS: The global arginine bioavailability ratio has been proposed as a biomarker reflective of L-arginine availability, arginase activity, and citrulline cycling, as all of these processes impact eNOS activity. The failure of oral supplementation of tetrahydrobiopterin to recouple eNOS has emphasized the importance of the tetrahydrobiopterin to dihydrobiopterin ratio. Identification of transporters for biopterin species as well as signals that regulate endogenous arginine production have provided insight for alternative strategies to raise endothelial tetrahydrobiopterin levels while reducing dihydrobiopterin and alter eNOS activity. Finally, new information about redox regulation of eNOS itself may point to ways of controlling oxidative stress in the vasculature. SUMMARY: Restoring proper eNOS activity is key to ameliorating or preventing cardiovascular complications of diabetes. Continued investigation is needed to uncover new means for maintaining endothelial nitric oxide bioavailability.


Subject(s)
Arginine/metabolism , Biopterins/analogs & derivatives , Diabetes Mellitus/physiopathology , Nitric Oxide/metabolism , Biomarkers/blood , Biopterins/metabolism , Cardiovascular Diseases/complications , Cardiovascular Diseases/physiopathology , Diabetes Mellitus/enzymology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Homeostasis , Humans , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Tetrahydrofolate Dehydrogenase/metabolism
14.
Amino Acids ; 39(2): 349-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20437186

ABSTRACT

Over the past 20 years, growing interest in the biochemistry, nutrition, and pharmacology of L-arginine has led to extensive studies to explore its nutritional and therapeutic roles in treating and preventing human metabolic disorders. Emerging evidence shows that dietary L-arginine supplementation reduces adiposity in genetically obese rats, diet-induced obese rats, finishing pigs, and obese human subjects with Type-2 diabetes mellitus. The mechanisms responsible for the beneficial effects of L-arginine are likely complex, but ultimately involve altering the balance of energy intake and expenditure in favor of fat loss or reduced growth of white adipose tissue. Recent studies indicate that L-arginine supplementation stimulates mitochondrial biogenesis and brown adipose tissue development possibly through the enhanced synthesis of cell-signaling molecules (e.g., nitric oxide, carbon monoxide, polyamines, cGMP, and cAMP) as well as the increased expression of genes that promote whole-body oxidation of energy substrates (e.g., glucose and fatty acids) Thus, L-arginine holds great promise as a safe and cost-effective nutrient to reduce adiposity, increase muscle mass, and improve the metabolic profile in animals and humans.


Subject(s)
Arginine/pharmacology , Arginine/therapeutic use , Obesity/drug therapy , Adipocytes/cytology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Dietary Supplements , Gene Expression Regulation/drug effects , Humans , Lipid Metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects
15.
Biofactors ; 35(1): 21-7, 2009.
Article in English | MEDLINE | ID: mdl-19319842

ABSTRACT

Obesity and type-II diabetes are growing major health issues worldwide. They are the leading risk factors for vascular insulin resistance, which plays an important role in the pathogenesis of cardiovascular disease, the leading cause of death in developed nations. Recent studies have shown that reduced synthesis of nitric oxide (NO; a major vasodilator) from L-arginine in endothelial cells is a major factor contributing to the impaired action of insulin in the vasculature of obese and diabetic subjects. The decreased NO generation results from a deficiency of (6R)-5,6,7,8-tetrahydrobiopterin [BH4; an essential cofactor for NO synthase (NOS)], as well as increased generation of glucosamine (an inhibitor of the pentose cycle for the production of NADPH, another cofactor for NOS) from glucose and L-glutamine. Accordingly, endothelial dysfunction can be prevented by (1) enhancement of BH4 synthesis through supplementation of its precursor (sepiapterin) via the salvage pathway; (2) transfer of the gene for GTP cyclohydrolase-I (the first and key regulatory enzyme for de novo synthesis of BH4); or (3) dietary supplementation of L-arginine (which stimulates GTP cyclohydrolase-I expression and inhibits hexosamine production). Modulation of the arginine-NO pathway by BH4 and arginine is beneficial for ameliorating vascular insulin resistance in obesity and diabetes.


Subject(s)
Insulin Resistance/physiology , Insulin/metabolism , Nitric Oxide/metabolism , Vascular Resistance/physiology , Animals , Arginine/metabolism , Biopterins/analogs & derivatives , Biopterins/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Humans , Models, Biological , Nitric Oxide Synthase/metabolism
16.
Amino Acids ; 37(1): 187-98, 2009 May.
Article in English | MEDLINE | ID: mdl-19212806

ABSTRACT

Dietary L-arginine (Arg) supplementation reduces white-fat gain in diet-induced obese rats but the underlying mechanisms are unknown. This study tested the hypothesis that Arg treatment affects expression of genes related to lipid metabolism in adipose tissue. Four-week-old male Sprague-Dawley rats were fed a low-fat (LF) or high-fat (HF) diet for 15 weeks. Thereafter, lean or obese rats continued to be fed their same respective diets and received drinking water containing 1.51% Arg-HCl or 2.55% L: -alanine (isonitrogenous control). After 12 weeks of Arg supplementation, rats were euthanized to obtain retroperitoneal adipose tissue for analyzing global changes in gene expression by microarray. The results were confirmed by RT-PCR analysis. HF feeding decreased mRNA levels for lipogenic enzymes, AMP-activated protein kinase, glucose transporters, heme oxygenase 3, glutathione synthetase, superoxide dismutase 3, peroxiredoxin 5, glutathione peroxidase 3, and stress-induced protein, while increasing expression of carboxypeptidase-A, peroxisome proliferator activated receptor (PPAR)-alpha, caspase 2, caveolin 3, and diacylglycerol kinase. In contrast, Arg supplementation reduced mRNA levels for fatty acid binding protein 1, glycogenin, protein phosphates 1B, caspases 1 and 2, and hepatic lipase, but increased expression of PPARgamma, heme oxygenase 3, glutathione synthetase, insulin-like growth factor II, sphingosine-1-phosphate receptor, and stress-induced protein. Biochemical analysis revealed oxidative stress in white adipose tissue of HF-fed rats, which was prevented by Arg supplementation. Collectively, these results indicate that HF diet and Arg supplementation differentially regulate gene expression to affect energy-substrate oxidation, redox state, fat accretion, and adipocyte differentiation in adipose tissue. Our findings provide a molecular mechanism to explain a beneficial effect of Arg on ameliorating diet-induced obesity in mammals.


Subject(s)
Adipose Tissue, White/drug effects , Arginine/administration & dosage , Dietary Fats/administration & dosage , Dietary Supplements , Gene Expression Regulation , Adipose Tissue, White/metabolism , Animals , Arginine/metabolism , Dietary Fats/metabolism , Down-Regulation , Gene Expression Profiling , Glutathione/metabolism , Male , Obesity/genetics , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
17.
J Nutr ; 139(2): 230-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19106310

ABSTRACT

Previous studies showed that dietary L-arginine supplementation decreased white fat mass in genetically obese rats. This study tested the effectiveness of L-arginine in diet-induced obesity. Male Sprague-Dawley rats were fed for 15 wk a high-fat (HF) (40% energy) or low-fat (LF) (10% energy) diet beginning at 4 wk of age, resulting in 18% higher body weight gains and 74% higher weights of major white fat pads (retroperitoneal, epididymal, subcutaneous, and mesenteric adipose tissues) in HF than in LF fed rats. Starting at 19 wk of age, rats in each dietary group were supplemented for 12 wk with 1.51% L-arginine-HCl or 2.55% L-alanine (isonitrogenous control) (n = 8 per treatment) in drinking water and arginine groups were individually pair-fed to alanine controls. Despite similar energy intake, absolute weights of white fat pads increased by 98% in control rats over a 12-wk period but only by 35% in arginine-supplemented rats. The arginine treatment reduced the relative weights of white fat pads by 30% and enhanced those of soleus muscle by 13%, extensor digitorum longus muscle by 11%, and brown fat by 34% compared with control rats. Serum concentrations of insulin, adiponectin, growth hormone, corticosterone, triiodothyronine, and thyroxine did not differ between control and arginine-supplemented rats. However, arginine treatment resulted in lower serum concentrations of leptin, glucose, triglycerides, urea, glutamine, and branched-chain amino acids, higher serum concentrations of nitric-oxide metabolites, and improvement in glucose tolerance. Thus, dietary arginine supplementation shifts nutrient partitioning to promote muscle over fat gain and may provide a useful treatment for improving the metabolic profile and reducing body white fat in diet-induced obese rats.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue/drug effects , Arginine/pharmacology , Diet , Dietary Supplements , Muscle, Skeletal/drug effects , Obesity/pathology , Adipose Tissue, Brown/pathology , Amino Acids/blood , Animals , Arginine/administration & dosage , Blood Glucose/analysis , Body Weight/drug effects , Glucose Tolerance Test , Leptin/blood , Male , Muscle, Skeletal/pathology , Nitric Oxide/blood , Organ Size/drug effects , Rats , Rats, Sprague-Dawley
18.
J Nutr ; 137(12): 2680-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18029483

ABSTRACT

Watermelon is rich in L-citrulline, an effective precursor of L-arginine. This study was conducted to determine whether dietary supplementation with watermelon pomace juice could ameliorate the metabolic syndrome in the Zucker diabetic fatty (ZDF) rat, an animal model of noninsulin-dependent diabetes mellitus. Nine-week-old ZDF rats were assigned randomly to receive drinking water containing 0% (control) or 0.2% L-arginine (as 0.24% L-arginine-HCl), 63% watermelon pomace juice, 0.01% lycopene, or 0.05% citrus pectin (n = 6 per treatment). At the end of the 4-wk supplementation period, blood samples, aortic rings, and hearts were obtained for biochemical and physiological analyses. Feed or energy intakes did not differ among the 5 groups of rats. However, dietary supplementation with watermelon pomace juice or L-arginine increased serum concentrations of arginine; reduced fat accretion; lowered serum concentrations of glucose, free fatty acids, homocysteine, and dimethylarginines; enhanced GTP cyclohydrolase-I activity and tetrahydrobiopterin concentrations in the heart; and improved acetylcholine-induced vascular relaxation. Compared with the control, dietary supplementation with lycopene or citrus pectin did not affect any measured parameter. These results provide the first evidence to our knowledge for a beneficial effect of watermelon pomace juice as a functional food for increasing arginine availability, reducing serum concentrations of cardiovascular risk factors, improving glycemic control, and ameliorating vascular dysfunction in obese animals with type-II diabetes.


Subject(s)
Citrullus , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Dietary Supplements , Metabolic Syndrome/complications , Metabolic Syndrome/drug therapy , Animals , Aorta/drug effects , Arginine/blood , Beverages , Blood Glucose , Citrulline/blood , Drinking , Eating , Energy Metabolism , GTP Cyclohydrolase/metabolism , Lipids/blood , Metabolic Syndrome/metabolism , Myocardium/enzymology , Nitric Oxide/blood , Nitric Oxide Synthase/metabolism , Rats , Rats, Zucker , Water
19.
J Nutr ; 137(6 Suppl 2): 1673S-1680S, 2007 06.
Article in English | MEDLINE | ID: mdl-17513446

ABSTRACT

Anticipating the future use of arginine to enhance fetal and neonatal growth as well as to treat diabetes and obesity, we performed studies in pigs, rats, and sheep to determine the pharmacokinetics of orally or i.v. administered arginine and the safety of its chronic supplementation. Our results indicate that all 3 species rapidly catabolized the supplemental arginine. The elevated circulating concentrations of arginine generally returned to baseline levels within 4-5 h after administration, with the rates varying with the age and physiological status of the animals. The clearance of arginine was greater in pregnant than in nonpregnant animals, in young than in adult animals, in lean than in obese animals, and in type-1 diabetic than in nondiabetic animals. I.v. administration of arginine-HCl to pregnant ewes (at least 0.081 g arginine.kg body weight-1.d-1) did not result in any undesirable treatment-related effect. Neonatal pigs, growing-finishing pigs, pregnant pigs, and adult rats tolerated large amounts of chronic supplemental arginine (e.g. 0.62, 0.32, 0.21, and 2.14 g.kg body weight-1.d-1, respectively) administered via enteral diets without the appearance of any adverse effect. On the basis of the comparative studies and a consideration of species differences in food intake per kilogram body weight, we estimate that a 70-kg human subject should be able to tolerate long-term parenteral and enteral supplemental doses of 6 and 15 g/d arginine, respectively, in addition to a basal amount of arginine (4-6 g/d) from regular diets.


Subject(s)
Animal Feed , Arginine/pharmacokinetics , Diabetes Mellitus, Type 1/metabolism , Dietary Supplements , Obesity/metabolism , Administration, Oral , Age Factors , Animals , Arginine/blood , Arginine/toxicity , Diabetes Mellitus, Type 2/metabolism , Female , Injections, Intravenous , Male , Pregnancy , Rats , Rats, Inbred BB , Rats, Zucker , Sheep , Sus scrofa
20.
J Nutr Biochem ; 17(9): 571-88, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16524713

ABSTRACT

Nitric oxide (NO) is synthesized from L-arginine by NO synthase in virtually all cell types. Emerging evidence shows that NO regulates the metabolism of glucose, fatty acids and amino acids in mammals. As an oxidant, pathological levels of NO inhibit nearly all enzyme-catalyzed reactions through protein oxidation. However, as a signaling molecule, physiological levels of NO stimulate glucose uptake as well as glucose and fatty acid oxidation in skeletal muscle, heart, liver and adipose tissue; inhibit the synthesis of glucose, glycogen, and fat in target tissues (e.g., liver and adipose); and enhance lipolysis in adipocytes. Thus, an inhibition of NO synthesis causes hyperlipidemia and fat accretion in rats, whereas dietary arginine supplementation reduces fat mass in diabetic fatty rats. The putative underlying mechanisms may involve multiple cyclic guanosine-3',5'-monophosphate-dependent pathways. First, NO stimulates the phosphorylation of adenosine-3',5'-monophosphate-activated protein kinase, resulting in (1) a decreased level of malonyl-CoA via inhibition of acetyl-CoA carboxylase and activation of malonyl-CoA decarboxylase and (2) a decreased expression of genes related to lipogenesis and gluconeogenesis (glycerol-3-phosphate acyltransferase, sterol regulatory element binding protein-1c and phosphoenolpyruvate carboxykinase). Second, NO increases the phosphorylation of hormone-sensitive lipase and perilipins, leading to the translocation of the lipase to the neutral lipid droplets and, hence, the stimulation of lipolysis. Third, NO activates expression of peroxisome proliferator-activated receptor-gamma coactivator-1alpha, thereby enhancing mitochondrial biogenesis and oxidative phosphorylation. Fourth, NO increases blood flow to insulin-sensitive tissues, promoting substrate uptake and product removal via the circulation. Modulation of the arginine-NO pathway through dietary supplementation with L-arginine or L-citrulline may aid in the prevention and treatment of the metabolic syndrome in obese humans and companion animals, and in reducing unfavorable fat mass in animals of agricultural importance.


Subject(s)
Arginine/metabolism , Energy Metabolism/physiology , Nitric Oxide/physiology , AMP-Activated Protein Kinases , Adipose Tissue/metabolism , Amino Acids/metabolism , Animals , Fatty Acids/metabolism , Gene Expression Regulation/physiology , Glucose/metabolism , Glycolysis/drug effects , Humans , Intestine, Small/metabolism , Lipid Metabolism/drug effects , Lipolysis/physiology , Liver/metabolism , Metabolic Syndrome/physiopathology , Mitochondria/physiology , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL