73 results on '"Morris SM Jr"'
Search Results
2. Decreased arginine bioavailability and increased serum arginase activity in asthma.
- Author
-
Morris CR, Poljakovic M, Lavrisha L, Machado L, Kuypers FA, Morris SM Jr., Morris, Claudia R, Poljakovic, Mirjana, Lavrisha, Lisa, Machado, Lorenzo, Kuypers, Frans A, and Morris, Sidney M Jr
- Abstract
Recent studies suggest that a nitric oxide (NO) deficiency and elevated arginase activity may play a role in the pathogenesis of asthma. Although much attention has been directed toward measurements of exhaled NO in asthma, no studies to date have evaluated levels of plasma arginase or arginine, the substrate for NO production, in patients with asthma. This study, therefore, measured amino acid levels, arginase activity, and nitric oxide metabolites in the blood of patients with asthma, as well as NO in exhaled breath. Although levels of virtually all amino acids were reduced, patients with asthma exhibited a striking reduction in plasma arginine levels compared with normal control subjects without asthma (45 +/- 22 vs. 94 +/- 29 microM, p < 0.0001), and serum arginase activity was elevated (1.6 +/- 0.8 vs. 0.5 +/- 0.3 micromol/ml/hour, asthma vs. control, p < 0.0001). High arginase activity in patients with asthma may contribute to low circulating arginine levels, thereby limiting arginine bioavailability and creating a NO deficiency that induces hyperreactive airways. Addressing the alterations in arginine metabolism may result in new strategies for treatment of asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
3. Isolation and characterization of a new hepatic epithelial-like cell line (AKN-1) from a normal liver
- Author
-
Nussler, AK, Vergani, G, Gollin, SM, Gansauge, S, Morris, SM, Jr., Demetris, AJ, Beger, HG, and Strom, SC
- Published
- 1998
- Full Text
- View/download PDF
4. Enhancing kidney DDAH-1 expression by adenovirus delivery reduces ADMA and ameliorates diabetic nephropathy.
- Author
-
Wetzel MD, Gao T, Stanley K, Cooper TK, Morris SM Jr, and Awad AS
- Subjects
- Albuminuria enzymology, Albuminuria genetics, Albuminuria prevention & control, Amidohydrolases genetics, Animals, Arginine metabolism, Cytokines genetics, Cytokines metabolism, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental enzymology, Diabetes Mellitus, Experimental genetics, Diabetic Nephropathies enzymology, Diabetic Nephropathies etiology, Diabetic Nephropathies genetics, Fibrosis, Inflammation Mediators metabolism, Kidney pathology, Male, Mice, Inbred DBA, Nitric Oxide metabolism, Nitric Oxide Synthase Type III genetics, Nitric Oxide Synthase Type III metabolism, Oxidative Stress, Signal Transduction, Thiobarbituric Acid Reactive Substances metabolism, Adenoviridae genetics, Amidohydrolases biosynthesis, Arginine analogs & derivatives, Diabetes Mellitus, Experimental therapy, Diabetic Nephropathies prevention & control, Genetic Therapy, Genetic Vectors, Kidney enzymology
- Abstract
Endothelial dysfunction, characterized by reduced bioavailability of nitric oxide and increased oxidative stress, is a hallmark characteristic in diabetes and diabetic nephropathy (DN). High levels of asymmetric dimethylarginine (ADMA) are observed in several diseases including DN and are a strong prognostic marker for cardiovascular events in patients with diabetes and end-stage renal disease. ADMA, an endogenous endothelial nitric oxide synthase (NOS3) inhibitor, is selectively metabolized by dimethylarginine dimethylaminohydrolase (DDAH). Low DDAH levels have been associated with cardiac and renal dysfunction, but its effects on DN are unknown. We hypothesized that enhanced renal DDAH-1 expression would improve DN by reducing ADMA and restoring NOS3 levels. DBA/2J mice injected with multiple low doses of vehicle or streptozotocin were subsequently injected intrarenally with adenovirus expressing DDAH-1 (Ad-h-DDAH-1) or vector control [Ad-green fluorescent protein (GFP)], and mice were followed for 6 wk. Diabetes was associated with increased kidney ADMA and reduced kidney DDAH activity and DDAH-1 expression but had no effect on kidney DDAH-2 expression. Ad-GFP-treated diabetic mice showed significant increases in albuminuria, histological changes, glomerular macrophage recruitment, inflammatory cytokine and fibrotic markers, kidney ADMA levels, and urinary thiobarbituric acid reactive substances excretion as an indicator of oxidative stress, along with a significant reduction in kidney DDAH activity and kidney NOS3 mRNA compared with normal mice. In contrast, Ad-h-DDAH-1 treatment of diabetic mice reversed these effects. These data indicate, for the first time, that DDAH-1 mediates renal tissue protection in DN via the ADMA-NOS3-interaction. Enhanced renal DDAH-1 activity could be a novel therapeutic tool for treating patients with diabetes.
- Published
- 2020
- Full Text
- View/download PDF
5. l-Homoarginine supplementation prevents diabetic kidney damage.
- Author
-
Wetzel MD, Gao T, Venkatachalam M, Morris SM Jr, and Awad AS
- Subjects
- Albuminuria drug therapy, Animals, Chemokine CXCL1 metabolism, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 1 metabolism, Diabetic Nephropathies metabolism, Diabetic Nephropathies pathology, Drug Evaluation, Preclinical methods, Homoarginine metabolism, Kidney metabolism, Kidney pathology, Kidney Glomerulus pathology, Macrophages pathology, Mice, Inbred DBA, Nitrates metabolism, Nitrites metabolism, Oxidative Stress drug effects, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Type 1 drug therapy, Diabetic Nephropathies prevention & control, Dietary Supplements, Homoarginine therapeutic use
- Abstract
l-homoarginine is an endogenous, non-proteinogenic amino acid that has emerged as a new player in health and disease. Specifically, low l-homoarginine levels are associated with cardiovascular diseases, stroke, and reduced kidney function. However, the role of l-homoarginine in the pathogenesis of diabetic nephropathy (DN) is not known. Experiments were conducted in 6-week-old Ins2
Akita mice supplemented with l-homoarginine via drinking water or mini osmotic pump for 12 weeks. Both plasma and kidney l-homoarginine levels were significantly reduced in diabetic mice compared to nondiabetic controls. Untreated Ins2Akita mice showed significant increases in urinary albumin excretion, histological changes, glomerular macrophage recruitment, the inflammatory cytokine KC-GRO/CXCL1, and urinary thiobarbituric acid reactive substances (TBARS) excretion as an indicator of oxidative stress, along with a significant reduction in kidney nitrate + nitrite levels compared to control mice at 18 weeks of age. In contrast, l-homoarginine supplementation for 12 weeks in Ins2Akita mice, via either drinking water or mini osmotic pump, significantly reduced albuminuria, renal histological changes, glomerular macrophage recruitment, KC-GRO/CXCL1 levels, urinary TBARS excretion, and largely restored kidney nitrate + nitrite levels. These data demonstrate that l-homoarginine supplementation attenuates specific features of DN in mice and could be a potential new therapeutic tool for treating diabetic patients., (© 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of The Physiological Society and the American Physiological Society.)- Published
- 2019
- Full Text
- View/download PDF
6. Distinct roles of arginases 1 and 2 in diabetic nephropathy.
- Author
-
Morris SM Jr, You H, Gao T, Vacher J, Cooper TK, and Awad AS
- Subjects
- Albuminuria enzymology, Albuminuria etiology, Animals, Diabetic Nephropathies complications, Fibronectins metabolism, Macrophages enzymology, Male, Mice, Renal Circulation, Tumor Necrosis Factor-alpha metabolism, Arginase metabolism, Diabetic Nephropathies enzymology
- Abstract
Diabetes is the leading cause of end-stage renal disease, resulting in a significant health care burden and loss of economic productivity by affected individuals. Because current therapies for progression of diabetic nephropathy (DN) are only moderately successful, identification of underlying mechanisms of disease is essential to develop more effective therapies. We showed previously that inhibition of arginase using S -(2-boronoethyl)-l-cysteine (BEC) or genetic deficiency of the arginase-2 isozyme was protective against key features of nephropathy in diabetic mouse models. However, those studies did not determine whether all markers of DN were dependent only on arginase-2 expression. The objective of this study was to identify features of DN that are associated specifically with expression of arginase-1 or -2. Elevated urinary albumin excretion rate and plasma urea levels, increases in renal fibronectin mRNA levels, and decreased renal medullary blood flow were associated almost completely and specifically with arginase-2 expression, indicating that arginase-2 selectively mediates major aspects of diabetic renal injury. However, increases in renal macrophage infiltration and renal TNF-α mRNA levels occurred independent of arginase-2 expression but were almost entirely abolished by treatment with BEC, indicating a distinct role for arginase-1. We therefore generated mice with a macrophage-specific deletion of arginase-1 ( CD11b
Cre / Arg1fl/fl ). CD11bCre / Arg1fl/fl mice had significantly reduced macrophage infiltration but had no effect on albuminuria compared with Arg1fl/fl mice after 12 wk of streptozotocin-induced diabetes. These results indicate that selective inhibition of arginase-2 would be effective in preventing or ameliorating major features of diabetic renal injury., (Copyright © 2017 the American Physiological Society.)- Published
- 2017
- Full Text
- View/download PDF
7. Arginase-2 mediates renal ischemia-reperfusion injury.
- Author
-
Raup-Konsavage WM, Gao T, Cooper TK, Morris SM Jr, Reeves WB, and Awad AS
- Subjects
- Acute Kidney Injury pathology, Adenosine Triphosphate metabolism, Animals, Arginase antagonists & inhibitors, Blood Urea Nitrogen, Creatinine blood, Kidney pathology, Male, Mice, Inbred C57BL, Mitochondria metabolism, Nitric Oxide Synthase Type III metabolism, Nitrites metabolism, Oxidative Stress, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Reperfusion Injury pathology, Acute Kidney Injury enzymology, Arginase metabolism, Reperfusion Injury enzymology
- Abstract
Novel therapeutic interventions for preventing or attenuating kidney injury following ischemia-reperfusion injury (IRI) remain a focus of significant interest. Currently, there are no definitive therapeutic or preventive approaches available for ischemic acute kidney injury (AKI). Our objective is to determine 1 ) whether renal arginase activity or expression is increased in renal IRI, and 2 ) whether arginase plays a role in development of renal IRI. The impact of arginase activity and expression on renal damage was evaluated in male C57BL/6J (wild type) and arginase-2 (ARG2)-deficient ( Arg2
-/- ) mice subjected to bilateral renal ischemia for 28 min, followed by reperfusion for 24 h. ARG2 expression and arginase activity significantly increased following renal IRI, paralleling the increase in kidney injury. Pharmacological blockade or genetic deficiency of Arg2 conferred kidney protection in renal IRI. Arg2-/- mice had significantly attenuated kidney injury and lower plasma creatinine and blood urea nitrogen levels after renal IRI. Blocking arginases using S -(2-boronoethyl)-l-cysteine (BEC) 18 h before ischemia mimicked arginase deficiency by reducing kidney injury, histopathological changes and kidney injury marker-1 expression, renal apoptosis, kidney inflammatory cell recruitment and inflammatory cytokines, and kidney oxidative stress; increasing kidney nitric oxide (NO) production and endothelial NO synthase (eNOS) phosphorylation, kidney peroxisome proliferator-activated receptor-γ coactivator-1α expression, and mitochondrial ATP; and preserving kidney mitochondrial ultrastructure compared with vehicle-treated IRI mice. Importantly, BEC-treated eNOS-knockout mice failed to reduce blood urea nitrogen and creatinine following renal IRI. These findings indicate that ARG2 plays a major role in renal IRI, via an eNOS-dependent mechanism, and that blocking ARG2 activity or expression could be a novel therapeutic approach for prevention of AKI., (Copyright © 2017 the American Physiological Society.)- Published
- 2017
- Full Text
- View/download PDF
8. Proposals for Upper Limits of Safe Intake for Arginine and Tryptophan in Young Adults and an Upper Limit of Safe Intake for Leucine in the Elderly.
- Author
-
Cynober L, Bier DM, Kadowaki M, Morris SM Jr, Elango R, and Smriga M
- Subjects
- Aged, Dietary Supplements, Dose-Response Relationship, Drug, Humans, Nutritional Requirements, Young Adult, Arginine administration & dosage, Arginine adverse effects, Leucine administration & dosage, Leucine adverse effects, Tryptophan administration & dosage, Tryptophan adverse effects
- Abstract
On the basis of research presented during the 9th Amino Acid Assessment Workshop, a No Observed Adverse Effect Level (NOAEL) for diet-added arginine (added mostly in the form of dietary supplements) of 30 g/d and an upper limit of safe intake (ULSI) for diet-added tryptophan (added mostly in the form of dietary supplements) of 4.5 g/d have been proposed. Both recommendations apply to healthy young adults. The total dietary leucine ULSI proposed for elderly individuals is 500 mg · kg
-1 · d-1 All 3 recommendations are relevant only to high-quality amino acid-containing products with specifications corresponding to those listed in the US Pharmacopeia Because the above amino acids are extensively utilized as dietary supplements for various real or perceived benefits, such as vasodilation, spermatogenesis, sleep, mood regulation, or muscle recovery, the above safety recommendations will have an important impact on regulatory and nutritional practices., (© 2016 American Society for Nutrition.)- Published
- 2016
- Full Text
- View/download PDF
9. Arginine Metabolism Revisited.
- Author
-
Morris SM Jr
- Subjects
- Animals, Arginine metabolism, Mammals physiology
- Abstract
Mammalian arginine metabolism is complex due to the expression of multiple enzymes that utilize arginine as substrate and to interactions or competition between specific enzymes involved in arginine metabolism. Moreover, cells may contain multiple intracellular arginine pools that are not equally accessible to all arginine metabolic enzymes, thus presenting additional challenges to more fully understanding arginine metabolism. At the whole-body level, arginine metabolism ultimately results in the production of a biochemically diverse range of products, including nitric oxide, urea, creatine, polyamines, proline, glutamate, agmatine, and homoarginine. Included in this group of compounds are the methylated arginines (e.g., asymmetric dimethylarginine), which are released upon degradation of proteins containing methylated arginine residues. Changes in arginine concentration also can regulate cellular metabolism and function via a variety of arginine sensors. Although much is known about arginine metabolism, elucidation of the physiologic or pathophysiologic roles for all of the pathways and their metabolites remains an active area of investigation, as exemplified by current findings highlighted in this review., (© 2016 American Society for Nutrition.)
- Published
- 2016
- Full Text
- View/download PDF
10. FoxO4 promotes early inflammatory response upon myocardial infarction via endothelial Arg1.
- Author
-
Zhu M, Goetsch SC, Wang Z, Luo R, Hill JA, Schneider J, Morris SM Jr, and Liu ZP
- Subjects
- Animals, Anti-Inflammatory Agents pharmacology, Arginase antagonists & inhibitors, Arginase genetics, Binding Sites, Cell Adhesion, Cell Cycle Proteins, Coculture Techniques, Disease Models, Animal, Endothelial Cells drug effects, Endothelial Cells immunology, Enzyme Induction, Forkhead Transcription Factors deficiency, Forkhead Transcription Factors genetics, HEK293 Cells, Humans, Inflammation genetics, Inflammation immunology, Inflammation pathology, Inflammation physiopathology, Inflammation prevention & control, Male, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction genetics, Myocardial Infarction immunology, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardial Infarction prevention & control, Myocytes, Cardiac drug effects, Myocytes, Cardiac immunology, Myocytes, Cardiac pathology, Neutrophil Infiltration, Nitric Oxide metabolism, Promoter Regions, Genetic, RNA Interference, Signal Transduction, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic, Transfection, U937 Cells, Arginase biosynthesis, Endothelial Cells enzymology, Forkhead Transcription Factors metabolism, Inflammation enzymology, Myocardial Infarction enzymology, Myocytes, Cardiac enzymology
- Abstract
Rationale: Inflammation in post-myocardial infarction (MI) is necessary for myocyte repair and wound healing. Unfortunately, it is also a key component of subsequent heart failure pathology. Transcription factor forkhead box O4 (FoxO4) regulates a variety of biological processes, including inflammation. However, its role in MI remains unknown., Objective: To test the hypothesis that FoxO4 promotes early post-MI inflammation via endothelial arginase 1 (Arg1)., Methods and Results: We induced MI in wild-type and FoxO4(-/-) mice. FoxO4(-/-) mice had a significantly higher post-MI survival, better cardiac function, and reduced infarct size. FoxO4(-/-) hearts had significantly fewer neutrophils, reduced expression of cytokines, and competitive nitric oxide synthase inhibitor Arg1. We generated conditional FoxO4 knockout mice with FoxO4 deleted in cardiac mycoytes or endothelial cells. FoxO4 endothelial cell-specific knockout mice showed significant post-MI improvement of cardiac function and reduction of neutrophil accumulation and cytokine expression, whereas FoxO4 cardiac mycoyte-specific knockout mice had no significant difference in cardiac function and post-MI inflammation from those of control littermates. FoxO4 binds the Foxo-binding site in the Arg1 promoter and activates Arg1 transcription. FoxO4 knockdown in human aortic endothelial cells upregulated nitric oxide on ischemia and suppressed monocyte adhesion that can be reversed by ectopic-expression of Arg1. Furthermore, chemical inhibition of Arg1 in wild-type mice had similar cardioprotection and reduced inflammation after MI as FoxO4 inactivation and administration of nitric oxide synthase inhibitor to FoxO4 KO mice reversed the beneficial effects of FoxO4 deletion on post-MI cardiac function., Conclusions: FoxO4 activates Arg1 transcription in endothelial cells in response to MI, leading to downregulation of nitric oxide and upregulation of neutrophil infiltration to the infarct area., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
11. Arginase inhibition: a new treatment for preventing progression of established diabetic nephropathy.
- Author
-
You H, Gao T, Cooper TK, Morris SM Jr, and Awad AS
- Subjects
- Albuminuria drug therapy, Albuminuria metabolism, Animals, Boronic Acids pharmacology, Captopril pharmacology, Diabetic Nephropathies metabolism, Disease Progression, Kidney drug effects, Kidney metabolism, Mice, Nitric Oxide Synthase Type III metabolism, Phosphorylation, Treatment Outcome, Angiotensin-Converting Enzyme Inhibitors therapeutic use, Arginase antagonists & inhibitors, Boronic Acids therapeutic use, Captopril therapeutic use, Diabetic Nephropathies drug therapy
- Abstract
Our previous publication showed that inhibition of arginase prevents the development of diabetic nephropathy (DN). However, identification of targets that retard the progression of established DN-which is more clinically relevant-is lacking. Therefore, we tested the hypothesis that arginase inhibition would prevent the progression of established DN. Effects of arginase inhibition were compared with treatment with the angiotensin-converting enzyme inhibitor captopril, a current standard of care in DN. Experiments were conducted in Ins2(Akita) mice treated with the arginase inhibitor S-(2-boronoethyl)-l-cysteine (BEC) or captopril starting at 6 wk of age for 12 wk (early treatment) or starting at 12 wk of age for 6 wk (late treatment). Early and late treatment with BEC resulted in protection from DN as indicated by reduced albuminuria, histological changes, kidney macrophage infiltration, urinary thiobarbituric acid-reactive substances, and restored nephrin expression, kidney nitrate/nitrite, kidney endothelial nitric oxide synthase phosphorylation, and renal medullary blood flow compared with vehicle-treated Ins2(Akita) mice at 18 wk of age. Interestingly, early treatment with captopril reduced albuminuria, histological changes, and kidney macrophage infiltration without affecting the other parameters, but late treatment with captopril was ineffective. These findings highlight the importance of arginase inhibition as a new potential therapeutic intervention in both early and late stages of diabetic renal injury., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
12. Diabetic nephropathy is resistant to oral L-arginine or L-citrulline supplementation.
- Author
-
You H, Gao T, Cooper TK, Morris SM Jr, and Awad AS
- Subjects
- Amino Acids blood, Amino Acids metabolism, Animals, Arginase metabolism, Arginine blood, Blood Pressure physiology, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Experimental pathology, Diabetic Nephropathies pathology, Fibronectins biosynthesis, Kidney pathology, Male, Membrane Proteins biosynthesis, Mice, Mice, Inbred DBA, Nitrates metabolism, Nitric Oxide Synthase Type III biosynthesis, Nitrites metabolism, Tumor Necrosis Factor-alpha biosynthesis, Arginine therapeutic use, Citrulline therapeutic use, Diabetic Nephropathies drug therapy, Dietary Supplements
- Abstract
Our recent publication showed that pharmacological blockade of arginases confers kidney protection in diabetic nephropathy via a nitric oxide (NO) synthase (NOS)3-dependent mechanism. Arginase competes with endothelial NOS (eNOS) for the common substrate L-arginine. Lack of L-arginine results in reduced NO production and eNOS uncoupling, which lead to endothelial dysfunction. Therefore, we hypothesized that L-arginine or L-citrulline supplementation would ameliorate diabetic nephropathy. DBA mice injected with multiple low doses of vehicle or streptozotocin (50 mg/kg ip for 5 days) were provided drinking water with or without L-arginine (1.5%, 6.05 g·kg(-1)·day(-1)) or L-citrulline (1.66%, 5.73 g·kg(-1)·day(-1)) for 9 wk. Nonsupplemented diabetic mice showed significant increases in albuminuria, blood urea nitrogen, glomerular histopathological changes, kidney macrophage recruitment, kidney TNF-α and fibronectin mRNA expression, kidney arginase activity, kidney arginase-2 protein expression, and urinary oxidative stress along with a significant reduction of nephrin and eNOS protein expression and kidney nitrite + nitrate compared with normal mice after 9 wk of diabetes. Surprisingly, L-arginine or L-citrulline supplementation in diabetic mice did not affect any of these parameters despite greatly increasing kidney and plasma arginine levels. These findings demonstrate that chronic L-arginine or L-citrulline supplementation does not prevent or reduce renal injury in a model of type 1 diabetes., (Copyright © 2014 the American Physiological Society.)
- Published
- 2014
- Full Text
- View/download PDF
13. Arginase inhibition mediates renal tissue protection in diabetic nephropathy by a nitric oxide synthase 3-dependent mechanism.
- Author
-
You H, Gao T, Cooper TK, Morris SM Jr, and Awad AS
- Subjects
- Albuminuria enzymology, Albuminuria prevention & control, Animals, Arginase metabolism, Biomarkers blood, Blood Urea Nitrogen, Boronic Acids administration & dosage, Creatinine blood, Diabetes Mellitus, Experimental blood, Diabetes Mellitus, Experimental chemically induced, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental enzymology, Diabetic Nephropathies blood, Diabetic Nephropathies enzymology, Diabetic Nephropathies etiology, Enzyme Inhibitors administration & dosage, Fibronectins metabolism, Infusions, Subcutaneous, Kidney enzymology, Macrophages drug effects, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Nitric Oxide Synthase Type III deficiency, Nitric Oxide Synthase Type III genetics, Ornithine metabolism, Oxidative Stress drug effects, Streptozocin, Time Factors, Tumor Necrosis Factor-alpha metabolism, Arginase antagonists & inhibitors, Boronic Acids pharmacology, Diabetes Mellitus, Experimental drug therapy, Diabetic Nephropathies prevention & control, Enzyme Inhibitors pharmacology, Kidney drug effects, Nitric Oxide Synthase Type III metabolism
- Abstract
Recently, we showed that pharmacological blockade or genetic deficiency of arginase-2 confers kidney protection in diabetic mouse models. Here, we tested whether the protective effect of arginase inhibition is nitric oxide synthase 3 (eNOS) dependent in diabetic nephropathy. Experiments were conducted in eNOS-knockout and their wild-type littermate mice using multiple low doses of vehicle or streptozotocin, and treated with continuous subcutaneous infusion of vehicle or the arginase inhibitor S-(2-boronoethyl)-L-cysteine by an osmotic pump. Inhibition of arginases for 6 weeks in diabetic wild-type mice significantly attenuated albuminuria, the increase in plasma creatinine and blood urea nitrogen, histopathological changes, kidney fibronectin and TNF-α expression, kidney macrophage recruitment, and oxidative stress compared with vehicle-treated diabetic wild-type mice. Arginase inhibition in diabetic eNOS-knockout mice failed to affect any of these parameters, but reduced kidney macrophage recruitment and kidney TNF-α expression compared with vehicle-treated diabetic eNOS-knockout mice. Furthermore, diabetic wild-type and eNOS-knockout mice exhibited increased kidney arginase-2 protein, arginase activity, and ornithine levels. Thus, arginase inhibition mediates renal tissue protection in diabetic nephropathy by an eNOS-dependent mechanism and has an eNOS-independent effect on kidney macrophage recruitment.
- Published
- 2013
- Full Text
- View/download PDF
14. A proposal for an upper limit of leucine safe intake in healthy adults.
- Author
-
Cynober L, Bier DM, Kadowaki M, Morris SM Jr, and Renwick AG
- Subjects
- Adult, Animals, Dietary Supplements, Humans, Male, Nutrition Policy, Rats, Rats, Sprague-Dawley, Leucine administration & dosage, Nutritional Requirements
- Abstract
Based on recent research, an upper limit of safe intake (ULSI) for leucine is proposed for healthy adults: 0.53 g/(kg·d). Because leucine has been used as a dietary supplement for many years in people practicing exercise and sport, further study with long-term exposure to leucine in this specific subpopulation should be performed to eventually adjust the ULSI.
- Published
- 2012
- Full Text
- View/download PDF
15. Selective endothelial overexpression of arginase II induces endothelial dysfunction and hypertension and enhances atherosclerosis in mice.
- Author
-
Vaisman BL, Andrews KL, Khong SM, Wood KC, Moore XL, Fu Y, Kepka-Lenhart DM, Morris SM Jr, Remaley AT, and Chin-Dusting JP
- Subjects
- Animals, Arginase genetics, Atherosclerosis genetics, Blood Pressure physiology, Blotting, Western, Endothelium, Vascular pathology, Hypertension genetics, Hypertension pathology, Macrophages, Peritoneal, Mice, Mice, Inbred C57BL, Mice, Transgenic, Reverse Transcriptase Polymerase Chain Reaction, Arginase metabolism, Atherosclerosis enzymology, Atherosclerosis pathology, Endothelium, Vascular enzymology, Hypertension enzymology
- Abstract
Background: Cardiovascular disorders associated with endothelial dysfunction, such as atherosclerosis, have decreased nitric oxide (NO) bioavailability. Arginase in the vasculature can compete with eNOS for L-arginine and has been implicated in atherosclerosis. The aim of this study was to evaluate the effect of endothelial-specific elevation of arginase II expression on endothelial function and the development of atherosclerosis., Methodology/principal Findings: Transgenic mice on a C57BL/6 background with endothelial-specific overexpression of human arginase II (hArgII) gene under the control of the Tie2 promoter were produced. The hArgII mice had elevated tissue arginase activity except in liver and in resident peritoneal macrophages, confirming endothelial specificity of the transgene. Using small-vessel myography, aorta from these mice exhibited endothelial dysfunction when compared to their non-transgenic littermate controls. The blood pressure of the hArgII mice was 17% higher than their littermate controls and, when crossed with apoE -/- mice, hArgII mice had increased aortic atherosclerotic lesions., Conclusion: We conclude that overexpression of arginase II in the endothelium is detrimental to the cardiovascular system.
- Published
- 2012
- Full Text
- View/download PDF
16. Arginase-2 mediates diabetic renal injury.
- Author
-
Morris SM Jr, Gao T, Cooper TK, Kepka-Lenhart D, and Awad AS
- Subjects
- Albuminuria etiology, Albuminuria prevention & control, Animals, Arginase antagonists & inhibitors, Arginase genetics, Diabetes Mellitus, Experimental chemically induced, Diabetes Mellitus, Experimental complications, Diabetic Nephropathies pathology, Diabetic Nephropathies physiopathology, Diabetic Nephropathies prevention & control, Disease Models, Animal, Enzyme Inhibitors toxicity, Gene Expression Regulation, Enzymologic drug effects, Histiocytes drug effects, Histiocytes pathology, Hyperglycemia etiology, Kidney blood supply, Kidney pathology, Kidney physiopathology, Male, Mice, Mice, Inbred DBA, Mice, Mutant Strains, Mice, Transgenic, Molecular Targeted Therapy, RNA, Messenger metabolism, Regional Blood Flow, Streptozocin toxicity, Arginase metabolism, Diabetic Nephropathies metabolism, Kidney metabolism
- Abstract
Objective: To determine 1) whether renal arginase activity or expression is increased in diabetes and 2) whether arginase plays a role in development of diabetic nephropathy (DN)., Research Design and Methods: The impact of arginase activity and expression on renal damage was evaluated in spontaneously diabetic Ins2(Akita) mice and in streptozotocin (STZ)-induced diabetic Dilute Brown Agouti (DBA) and arginase-2-deficient mice (Arg2(-/-))., Results: Pharmacological blockade or genetic deficiency of arginase-2 conferred kidney protection in Ins2(Akita) mice or STZ-induced diabetic renal injury. Blocking arginases using S-(2-boronoethyl)-L-cysteine for 9 weeks in Ins2(Akita) mice or 6 weeks in STZ-induced diabetic DBA mice significantly attenuated albuminuria, the increase in blood urea nitrogen, histopathological changes, and kidney macrophage recruitment compared with vehicle-treated Ins2(Akita) mice. Furthermore, kidney arginase-2 expression increased in Ins2(Akita) mice compared with control. In contrast, arginase-1 expression was undetectable in kidneys under normal or diabetes conditions. Arg2(-/-) mice mimicked arginase blockade by reducing albuminuria after 6 and 18 weeks of STZ-induced diabetes. In wild-type mice, kidney arginase activity increased significantly after 6 and 18 weeks of STZ-induced diabetes but remained very low in STZ-diabetic Arg2(-/-) mice. The increase in kidney arginase activity was associated with a reduction in renal medullary blood flow in wild-type mice after 6 weeks of STZ-induced diabetes, an effect significantly attenuated in diabetic Arg2(-/-) mice., Conclusions: These findings indicate that arginase-2 plays a major role in induction of diabetic renal injury and that blocking arginase-2 activity or expression could be a novel therapeutic approach for treatment of DN.
- Published
- 2011
- Full Text
- View/download PDF
17. LXRα regulates macrophage arginase 1 through PU.1 and interferon regulatory factor 8.
- Author
-
Pourcet B, Feig JE, Vengrenyuk Y, Hobbs AJ, Kepka-Lenhart D, Garabedian MJ, Morris SM Jr, Fisher EA, and Pineda-Torra I
- Subjects
- Animals, Arginase biosynthesis, Arginase genetics, Atherosclerosis genetics, Atherosclerosis metabolism, Atherosclerosis prevention & control, Cell Line, Gene Targeting methods, Genetic Loci, Interferon Regulatory Factors antagonists & inhibitors, Interferon Regulatory Factors metabolism, Liver X Receptors, Macrophages enzymology, Mice, Mice, Knockout, Orphan Nuclear Receptors deficiency, Orphan Nuclear Receptors genetics, Protein Binding genetics, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Trans-Activators antagonists & inhibitors, Trans-Activators metabolism, Arginase metabolism, Interferon Regulatory Factors physiology, Macrophages metabolism, Orphan Nuclear Receptors physiology, Proto-Oncogene Proteins physiology, Trans-Activators physiology
- Abstract
Rationale: Activation of liver X receptors (LXRs) inhibits the progression of atherosclerosis and promotes regression of existing lesions. In addition, LXRα levels are high in regressive plaques. Macrophage arginase 1 (Arg1) expression is inversely correlated with atherosclerosis progression and is markedly decreased in foam cells within the lesion., Objective: To investigate LXRα regulation of Arg1 expression in cultured macrophages and atherosclerotic regressive lesions., Methods and Results: We found that Arg1 expression is enhanced in CD68+ cells from regressive versus progressive lesions in a murine aortic arch transplant model. In cultured macrophages, ligand-activated LXRα markedly enhances basal and interleukin-4-induced Arg1 mRNA and protein expression as well as promoter activity. This LXRα-enhanced Arg1 expression correlates with a reduction in nitric oxide levels. Moreover, Arg1 expression within regressive atherosclerotic plaques is LXRα-dependent, as enhanced expression of Arg1 in regressive lesions is impaired in LXRα-deficient CD68+ cells. LXRα does not bind to the Arg1 promoter but instead promotes the interaction between PU.1 and interferon regulatory factor (IRF)8 transcription factors and induces their binding of a novel composite element. Accordingly, knockdown of either IRF8 or PU.1 strongly impairs LXRα regulation of Arg1 expression in macrophage cells. Finally, we demonstrate that LXRα binds the IRF8 locus and its activation increases IRF8 mRNA and protein levels in these cells., Conclusions: This work implicates Arg1 in atherosclerosis regression and identifies LXRα as a novel regulator of Arg1 and IRF8 in macrophages. Furthermore, it provides a unique molecular mechanism by which LXRα regulates macrophage target gene expression through PU.1 and IRF8.
- Published
- 2011
- Full Text
- View/download PDF
18. Nitrite-generated NO circumvents dysregulated arginine/NOS signaling to protect against intimal hyperplasia in Sprague-Dawley rats.
- Author
-
Alef MJ, Vallabhaneni R, Carchman E, Morris SM Jr, Shiva S, Wang Y, Kelley EE, Tarpey MM, Gladwin MT, Tzeng E, and Zuckerbraun BS
- Subjects
- Animals, Arginase metabolism, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p21 physiology, Hyperplasia pathology, Hyperplasia physiopathology, Hyperplasia prevention & control, Male, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, Nitric Oxide biosynthesis, Rats, Rats, Sprague-Dawley, Signal Transduction, Tunica Intima drug effects, Tunica Intima injuries, Tunica Intima pathology, Xanthine Dehydrogenase metabolism, Arginine physiology, Nitric Oxide physiology, Nitric Oxide Synthase Type III physiology, Sodium Nitrite administration & dosage, Tunica Intima physiology
- Abstract
Vascular disease, a significant cause of morbidity and mortality in the developed world, results from vascular injury. Following vascular injury, damaged or dysfunctional endothelial cells and activated SMCs engage in vasoproliferative remodeling and the formation of flow-limiting intimal hyperplasia (IH). We hypothesized that vascular injury results in decreased bioavailability of NO secondary to dysregulated arginine-dependent NO generation. Furthermore, we postulated that nitrite-dependent NO generation is augmented as an adaptive response to limit vascular injury/proliferation and can be harnessed for its protective effects. Here we report that sodium nitrite (intraperitoneal, inhaled, or oral) limited the development of IH in a rat model of vascular injury. Additionally, nitrite led to the generation of NO in vessels and SMCs, as well as limited SMC proliferation via p21Waf1/Cip1 signaling. These data demonstrate that IH is associated with increased arginase-1 levels, which leads to decreased NO production and bioavailability. Vascular injury also was associated with increased levels of xanthine oxidoreductase (XOR), a known nitrite reductase. Chronic inhibition of XOR and a diet deficient in nitrate/nitrite each exacerbated vascular injury. Moreover, established IH was reversed by dietary supplementation of nitrite. The vasoprotective effects of nitrite were counteracted by inhibition of XOR. These data illustrate the importance of nitrite-generated NO as an endogenous adaptive response and as a pathway that can be harnessed for therapeutic benefit.
- Published
- 2011
- Full Text
- View/download PDF
19. Arginase activities and global arginine bioavailability in wild-type and ApoE-deficient mice: responses to high fat and high cholesterol diets.
- Author
-
Erdely A, Kepka-Lenhart D, Salmen-Muniz R, Chapman R, Hulderman T, Kashon M, Simeonova PP, and Morris SM Jr
- Subjects
- Alanine Transaminase metabolism, Animal Feed, Animals, Arginase blood, Biological Availability, Citrulline genetics, Diet, Isoenzymes chemistry, Male, Mice, Mice, Inbred C57BL, Ornithine genetics, Reverse Transcriptase Polymerase Chain Reaction, Apolipoproteins E genetics, Arginase metabolism, Arginine pharmacology, Cholesterol metabolism
- Abstract
Increased catabolism of arginine by arginase is increasingly viewed as an important pathophysiological factor in cardiovascular disease, including atherosclerosis induced by high cholesterol diets. Whereas previous studies have focused primarily on effects of high cholesterol diets on arginase expression and arginine metabolism in specific blood vessels, there is no information regarding the impact of lipid diets on arginase activity or arginine bioavailability at a systemic level. We, therefore, evaluated the effects of high fat (HF) and high fat-high cholesterol (HC) diets on arginase activity in plasma and tissues and on global arginine bioavailability (defined as the ratio of plasma arginine to ornithine + citrulline) in apoE(-/-) and wild-type C57BL/6J mice. HC and HF diets led to reduced global arginine bioavailability in both strains. The HC diet resulted in significantly elevated plasma arginase in both strains, but the HF diet increased plasma arginase only in apoE(-/-) mice. Elevated plasma arginase activity correlated closely with increased alanine aminotransferase levels, indicating that liver damage was primarily responsible for elevated plasma arginase. The HC diet, which promotes atherogenesis, also resulted in increased arginase activity and expression of the type II isozyme of arginase in multiple tissues of apoE(-/-) mice only. These results raise the possibility that systemic changes in arginase activity and global arginine bioavailability may be contributing factors in the initiation and/or progression of cardiovascular disease.
- Published
- 2010
- Full Text
- View/download PDF
20. Thrombin induces endothelial arginase through AP-1 activation.
- Author
-
Zhu W, Chandrasekharan UM, Bandyopadhyay S, Morris SM Jr, DiCorleto PE, and Kashyap VS
- Subjects
- Activating Transcription Factor 2 genetics, Activating Transcription Factor 2 metabolism, Animals, Aorta cytology, Arginase genetics, Cells, Cultured, Endothelial Cells cytology, Mitogen-Activated Protein Kinases metabolism, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun metabolism, RNA Interference, Rats, Signal Transduction physiology, Thrombin genetics, Thrombin metabolism, Transcription Factor AP-1 genetics, Arginase metabolism, Endothelial Cells enzymology, Transcription Factor AP-1 metabolism
- Abstract
Arterial thrombosis is a common disease leading to severe ischemia beyond the obstructing thrombus. Additionally, endothelial dysfunction at the site of thrombosis can be rescued by l-arginine supplementation or arginase blockade in several animal models. Exposure of rat aortic endothelial cells (RAECs) to thrombin upregulates arginase I mRNA and protein levels. In this study, we further investigated the molecular mechanism of thrombin-induced arginase changes in endothelial cells. Thrombin strikingly increased arginase I promoter and enzyme activity in primary cultured RAECs. Using different deletion and point mutations of the promoter, we demonstrated that the activating protein-1 (AP-1) consensus site located at -3,157 bp in the arginase I promoter was a thrombin-responsive element. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay further confirmed that upon thrombin stimulation, c-Jun and activating transcription factor-2 (ATF-2) bound to the AP-1 site, which initiated the transactivation. Moreover, loss-of-function studies using small interfering RNA confirmed that recruitment of these two transcription factors to the AP-1 site was required for thrombin-induced arginase upregulation. In the course of defining the signaling pathway leading to the activation of AP-1 by thrombin, we found thrombin-induced phosphorylation of stress-activated protein kinase/c-Jun-NH(2)-terminal kinase (SAPK/JNK or JNK1/2/3) and p38 mitogen-activated protein kinase, which were followed by the phosphorylation of both c-Jun and ATF-2. These findings reveal the basis for thrombin induction of endothelial arginase I and indicate that arginase inhibition may be an attractive therapeutic alternative in the setting of arterial thrombosis and its associated endothelial dysfunction.
- Published
- 2010
- Full Text
- View/download PDF
21. A large-scale chemical screen for regulators of the arginase 1 promoter identifies the soy isoflavone daidzeinas a clinically approved small molecule that can promote neuronal protection or regeneration via a cAMP-independent pathway.
- Author
-
Ma TC, Campana A, Lange PS, Lee HH, Banerjee K, Bryson JB, Mahishi L, Alam S, Giger RJ, Barnes S, Morris SM Jr, Willis DE, Twiss JL, Filbin MT, and Ratan RR
- Subjects
- Analysis of Variance, Animals, Animals, Newborn, Arginase metabolism, CHO Cells, Cells, Cultured, Cerebellum cytology, Cricetinae, Cricetulus, Dose-Response Relationship, Drug, Embryo, Mammalian, Enzyme Inhibitors pharmacology, GAP-43 Protein metabolism, Ganglia, Spinal cytology, High-Throughput Screening Assays methods, Hippocampus cytology, Male, Myelin-Associated Glycoprotein pharmacology, Nerve Regeneration physiology, Neurons cytology, Optic Nerve Diseases drug therapy, Optic Nerve Diseases pathology, Oxidative Stress drug effects, Promoter Regions, Genetic drug effects, Rats, Rats, Sprague-Dawley, Receptors, Estrogen metabolism, Small Molecule Libraries, Arginase genetics, Cyclic AMP metabolism, Isoflavones pharmacology, Nerve Regeneration drug effects, Neurons drug effects, Neuroprotective Agents pharmacology, Promoter Regions, Genetic physiology
- Abstract
An ideal therapeutic for stroke or spinal cord injury should promote survival and regeneration in the CNS. Arginase 1 (Arg1) has been shown to protect motor neurons from trophic factor deprivation and allow sensory neurons to overcome neurite outgrowth inhibition by myelin proteins. To identify small molecules that capture Arg1's protective and regenerative properties, we screened a hippocampal cell line stably expressing the proximal promoter region of the arginase 1 gene fused to a reporter gene against a library of compounds containing clinically approved drugs. This screen identified daidzein as a transcriptional inducer of Arg1. Both CNS and PNS neurons primed in vitro with daidzein overcame neurite outgrowth inhibition from myelin-associated glycoprotein, which was mirrored by acutely dissociated and cultured sensory neurons primed in vivo by intrathecal or subcutaneous daidzein infusion. Further, daidzein was effective in promoting axonal regeneration in vivo in an optic nerve crush model when given intraocularly without lens damage, or most importantly, when given subcutaneously after injury. Mechanistically, daidzein requires transcription and induction of Arg1 activity for its ability to overcome myelin inhibition. In contrast to canonical Arg1 activators, daidzein increases Arg1 without increasing CREB phosphorylation, suggesting its effects are cAMP-independent. Accordingly, it may circumvent known CNS side effects of some cAMP modulators. Indeed, daidzein appears to be safe as it has been widely consumed in soy products, crosses the blood-brain barrier, and is effective without pretreatment, making it an ideal candidate for development as a therapeutic for spinal cord injury or stroke.
- Published
- 2010
- Full Text
- View/download PDF
22. Recent advances in arginine metabolism: roles and regulation of the arginases.
- Author
-
Morris SM Jr
- Subjects
- Animals, Arginase antagonists & inhibitors, Arginase biosynthesis, Arginase genetics, Arginine physiology, Cytosol enzymology, Disease Models, Animal, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes biosynthesis, Isoenzymes genetics, Isoenzymes physiology, Mitochondrial Proteins antagonists & inhibitors, Mitochondrial Proteins biosynthesis, Mitochondrial Proteins genetics, Mitochondrial Proteins physiology, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular enzymology, Arginase physiology, Arginine metabolism, Gene Expression Regulation, Enzymologic physiology
- Abstract
As arginine can serve as precursor to a wide range of compounds, including nitric oxide, creatine, urea, polyamines, proline, glutamate and agmatine, there is considerable interest in elucidating mechanisms underlying regulation of its metabolism. It is now becoming apparent that the two isoforms of arginase in mammals play key roles in regulation of most aspects of arginine metabolism in health and disease. In particular, work over the past several years has focused on the roles and regulation of the arginases in vascular disease, pulmonary disease, infectious disease, immune cell function and cancer. As most of these topics have been considered in recent review articles, this review will focus more closely on results of recent studies on expression of the arginases in endothelial and vascular smooth muscle cells, post-translational modulation of arginase activity and applications of arginase inhibitors in vivo.
- Published
- 2009
- Full Text
- View/download PDF
23. Arginine metabolism: boundaries of our knowledge.
- Author
-
Morris SM Jr
- Subjects
- Animals, Arginase metabolism, Humans, Protein Biosynthesis physiology, Arginine metabolism, Biogenic Polyamines biosynthesis, Nitric Oxide biosynthesis, Proline biosynthesis
- Abstract
Arginine has multiple metabolic fates and thus is one of the most versatile amino acids. Not only is it metabolically interconvertible with the amino acids proline and glutamate, but it also serves as a precursor for synthesis of protein, nitric oxide, creatine, polyamines, agmatine, and urea. These processes do not all occur within each cell but are differentially expressed according to cell type, age and developmental stage, diet, and state of health or disease. Arginine metabolism also is modulated by activities of various transporters that move arginine and its metabolites across the plasma and mitochondrial membranes. Moreover, several key enzymes in arginine metabolism are expressed as multiple isozymes whose expression can change rapidly and dramatically in response to a variety of different stimuli in health and disease. As illustrated by the questions raised in this article, we currently have an imperfect and incomplete picture of arginine metabolism for any mammalian species. It has become clear that a more complete understanding of arginine metabolism will require integration of information obtained from multiple approaches, including genomics, proteomics, and metabolomics.
- Published
- 2007
- Full Text
- View/download PDF
24. Lactate dehydrogenase as a biomarker of hemolysis-associated nitric oxide resistance, priapism, leg ulceration, pulmonary hypertension, and death in patients with sickle cell disease.
- Author
-
Kato GJ, McGowan V, Machado RF, Little JA, Taylor J 6th, Morris CR, Nichols JS, Wang X, Poljakovic M, Morris SM Jr, and Gladwin MT
- Subjects
- Adult, Anemia, Sickle Cell blood, Biomarkers blood, Clinical Enzyme Tests, Death, Female, Humans, Hypertension, Pulmonary diagnosis, Leg Ulcer diagnosis, Male, Middle Aged, Priapism diagnosis, Vascular Diseases diagnosis, Anemia, Sickle Cell complications, Hemolysis, L-Lactate Dehydrogenase blood
- Abstract
Pulmonary hypertension is prevalent in adult patients with sickle cell disease and is strongly associated with early mortality and markers of hemolysis, in particular, serum lactate dehydrogenase (LDH). Intravascular hemolysis leads to impaired bioavailability of nitric oxide (NO), mediated by NO scavenging by plasma oxyhemoglobin and by arginine degradation by plasma arginase. We hypothesized that serum LDH may represent a convenient biomarker of intravascular hemolysis and NO bioavailability, characterizing a clinical subphenotype of hemolysis-associated vasculopathy. In a cohort of 213 patients with sickle cell disease, we found statistically significant associations of steady-state LDH with low levels of hemoglobin and haptoglobin and high levels of reticulocytes, bilirubin, plasma hemoglobin, aspartate aminotransferase, arginase, and soluble adhesion molecules. LDH isoenzyme fractionation confirmed predominance of LD1 and LD2, the principal isoforms within erythrocytes. In a subgroup, LDH levels closely correlated with plasma cell-free hemoglobin, accelerated NO consumption by plasma, and impaired vasodilatory responses to an NO donor. Remarkably, this simple biomarker was associated with a clinical subphenotype of pulmonary hypertension, leg ulceration, priapism, and risk of death in patients with sickle cell disease. We propose that LDH elevation identifies patients with a syndrome of hemolysis-associated NO resistance, endothelial dysfunction, and end-organ vasculopathy.
- Published
- 2006
- Full Text
- View/download PDF
25. Inhibition of phosphodiesterase 4 amplifies cytokine-dependent induction of arginase in macrophages.
- Author
-
Erdely A, Kepka-Lenhart D, Clark M, Zeidler-Erdely P, Poljakovic M, Calhoun WJ, and Morris SM Jr
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Animals, Antineoplastic Agents pharmacology, Arginase genetics, Bronchoalveolar Lavage Fluid chemistry, Cell Line, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4, Drug Synergism, Enzyme Induction, Humans, Mice, Phosphodiesterase Inhibitors pharmacology, Promoter Regions, Genetic genetics, Rolipram pharmacology, 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, Arginase biosynthesis, Interleukin-4 pharmacology, Macrophages, Alveolar enzymology, Transforming Growth Factor beta pharmacology
- Abstract
Arginase is greatly elevated in asthma and is thought to play a role in the pathophysiology of this disease. As inhibitors of phosphodiesterase 4 (PDE4), the predominant PDE in macrophages, elevate cAMP levels and reduce inflammation, they have been proposed for use in treatment of asthma and chronic obstructive pulmonary disease. As cAMP is an inducer of arginase, we tested the hypothesis that a PDE4 inhibitor would enhance macrophage arginase induction by key cytokines implicated in asthma and other pulmonary diseases. RAW 264.7 cells were stimulated with IL-4 or transforming growth factor (TGF)-beta, with and without the PDE4 inhibitor rolipram. IL-4 and TGF-beta increased arginase activity 16- and 5-fold, respectively. Rolipram alone had no effect but when combined with IL-4 and TGF-beta synergistically enhanced arginase activity by an additional 15- and 5-fold, respectively. The increases in arginase I protein and mRNA levels mirrored increases in arginase activity. Induction of arginase II mRNA was also enhanced by rolipram but to a much lesser extent than arginase I. Unlike its effect in RAW 264.7 cells, IL-4 alone did not increase arginase activity in human alveolar macrophages (AM) from healthy volunteers. However, combining IL-4 with agents to induce cAMP levels induced arginase activity in human AM significantly above the level obtained with cAMP-inducing agents alone. In conclusion, agents that elevate cAMP significantly enhance induction of arginase by cytokines. Therefore, consequences of increased arginase expression should be evaluated whenever PDE inhibitors are proposed for treatment of inflammatory disorders in which IL-4 and/or TGF-beta predominate.
- Published
- 2006
- Full Text
- View/download PDF
26. Arginine: beyond protein.
- Author
-
Morris SM Jr
- Subjects
- Amino Acid Transport Systems, Animals, Arginase metabolism, Arginine metabolism, Humans, Arginine physiology, Gene Expression Regulation physiology
- Abstract
Arginine, a semiessential or conditionally essential amino acid in humans, is one of the most metabolically versatile amino acids and serves as a precursor for the synthesis of urea, nitric oxide, polyamines, proline, glutamate, creatine, and agmatine. Arginine is metabolized through a complex and highly regulated set of pathways that remain incompletely understood at both the whole-body and the cellular levels. Adding to the metabolic complexity is the fact that limited arginine availability can selectively affect the expression of specific genes, most of which are themselves involved in some aspect of arginine metabolism. This overview highlights selected aspects of arginine metabolism, including areas in which our knowledge remains fragmentary and incomplete.
- Published
- 2006
- Full Text
- View/download PDF
27. Application of branched-chain amino acids in experimental animals: discussion of session 2.
- Author
-
Morris SM Jr
- Subjects
- Animals, Models, Animal, Muscle, Skeletal metabolism, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Amino Acids, Branched-Chain metabolism
- Published
- 2006
- Full Text
- View/download PDF
28. Elevated levels of the 64-kDa cleavage stimulatory factor (CstF-64) in lipopolysaccharide-stimulated macrophages influence gene expression and induce alternative poly(A) site selection.
- Author
-
Shell SA, Hesse C, Morris SM Jr, and Milcarek C
- Subjects
- 3' Untranslated Regions, Algorithms, Animals, Binding Sites, Blotting, Western, Cell Proliferation, Cells, Cultured, Genes, Reporter, Lipopolysaccharides pharmacology, Mice, Mice, Inbred C57BL, Models, Biological, Models, Genetic, Models, Statistical, Oligonucleotide Array Sequence Analysis, Oligonucleotides chemistry, Open Reading Frames, Polyadenylation, Promoter Regions, Genetic, Protein Binding, Protein Conformation, RNA chemistry, RNA Processing, Post-Transcriptional, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Transcription, Genetic, Cleavage Stimulation Factor chemistry, Cleavage Stimulation Factor physiology, Gene Expression Regulation, Lipopolysaccharides metabolism, Macrophages metabolism, Poly A chemistry
- Abstract
Lipopolysaccharide (LPS) activation of murine RAW 264.7 macrophages influences the expression of multiple genes through transcriptional and post-transcriptional mechanisms. We observed a 5-fold increase in CstF-64 expression following LPS treatment of RAW macrophages. The increase in CstF-64 protein was specific in that several other factors involved in 3'-end processing were not affected by LPS stimulation. Activation of RAW macrophages with LPS caused an increase in proximal poly(A) site selection within a reporter mini-gene containing two linked poly(A) sites that occurred concomitant with the increase in CstF-64 expression. Furthermore, forced overexpression of the CstF-64 protein also induced alternative poly(A) site selection on the reporter minigene. Microarray analysis performed on CstF-64 overexpressing RAW macrophages revealed that elevated levels of CstF-64 altered the expression of 51 genes, 14 of which showed similar changes in gene expression with LPS stimulation. Sequence analysis of the 3'-untranslated regions of these 51 genes revealed that over 45% possess multiple putative poly(A) sites. Two of these 51 genes demonstrated alternative polyadenylation under both LPS-stimulating and CstF-64-overexpressing conditions. We concluded that the physiologically increased levels of CstF-64 observed in LPS-stimulated RAW macrophages contribute to the changes in expression and alternative polyadenylation of a number of genes, thus identifying another level of gene regulation that occurs in macrophages activated with LPS.
- Published
- 2005
- Full Text
- View/download PDF
29. Arginine metabolism in vascular biology and disease.
- Author
-
Morris SM Jr
- Subjects
- Animals, Arginase metabolism, Cardiovascular Diseases enzymology, Erythrocytes enzymology, Humans, Muscle, Smooth, Vascular enzymology, Nitric Oxide metabolism, Nitric Oxide Synthase metabolism, Arginine metabolism, Endothelium, Vascular enzymology
- Abstract
Arginine metabolism plays a major role in cardiovascular physiology and pathophysiology, largely via nitric oxide (NO)-dependent processes. It is becoming increasingly apparent, however, that arginine metabolic enzymes other than the NO synthases can also play important roles via both NO-dependent and -independent processes. There are three sources of arginine in vivo and at least five mammalian enzymes or enzyme families that utilize arginine as substrate. Changes in arginine availability or in production of the different end products of the various arginine metabolic pathways can have distinct and profound physiologic consequences. However, our knowledge regarding the complex interplay between these pathways at the level of the whole body, specific tissues, and even individual cells, is incomplete. This review will highlight recent findings in this area that may suggest additional avenues of investigation that will allow a fuller understanding of cardiovascular physiology in health and disease.
- Published
- 2005
- Full Text
- View/download PDF
30. Induction of arginase I transcription by IL-4 requires a composite DNA response element for STAT6 and C/EBPbeta.
- Author
-
Gray MJ, Poljakovic M, Kepka-Lenhart D, and Morris SM Jr
- Subjects
- Animals, Base Sequence, Binding Sites genetics, Binding, Competitive, Cell Line, DNA genetics, DNA metabolism, Electrophoretic Mobility Shift Assay, Gene Expression Regulation, Enzymologic drug effects, Luciferases genetics, Luciferases metabolism, Macrophages cytology, Macrophages drug effects, Macrophages metabolism, Mice, Mice, Inbred Strains, Molecular Sequence Data, Promoter Regions, Genetic genetics, Protein Binding, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, STAT6 Transcription Factor, Sequence Homology, Nucleic Acid, Transcription, Genetic drug effects, Transfection, Arginase genetics, CCAAT-Enhancer-Binding Protein-beta metabolism, Interleukin-4 pharmacology, Response Elements genetics, Trans-Activators metabolism
- Abstract
Arginine metabolism in macrophages during infection and inflammation is complex, owing to differential regulation of inducible nitric oxide synthase (iNOS) and arginases by cytokines and other agents. Changes in levels of Th2 cytokines such as interleukin-4 (IL-4) can play important roles in these conditions via effects on arginine metabolism. IL-4 alters macrophage arginine metabolism by inducing arginase I expression and inhibiting nitric oxide production. To determine the molecular basis for induction of arginase I, the promoter of the murine arginase I gene was cloned and analyzed by transfection in RAW 264.7 macrophage cells. IL-4 induction required a composite response element containing STAT6 and C/EBP sites located 2.86 kb upstream of the transcription start site. Competition experiments showed that STAT6 and C/EBPbeta bind to the STAT6 and C/EBP sites non-cooperatively. Elucidation of the mechanisms involved in regulation of arginase I transcription may provide a basis for developing strategies to modulate arginase expression in Th2 cytokine-predominant diseases.
- Published
- 2005
- Full Text
- View/download PDF
31. Macrophage arginase regulation by CCAAT/enhancer-binding protein beta.
- Author
-
Albina JE, Mahoney EJ, Daley JM, Wesche DE, Morris SM Jr, and Reichner JS
- Subjects
- 8-Bromo Cyclic Adenosine Monophosphate metabolism, Animals, Arginase metabolism, CCAAT-Enhancer-Binding Protein-beta metabolism, Cell Line, Cell Nucleus metabolism, Cells, Cultured, Cyclic AMP metabolism, Escherichia coli metabolism, Hypoxia, Immunoblotting, Inflammation, Interleukin-4 metabolism, Lipopolysaccharides metabolism, Macrophages metabolism, Mice, Mice, Transgenic, Peritoneum metabolism, Phenotype, STAT6 Transcription Factor, Trans-Activators metabolism, Arginase chemistry, CCAAT-Enhancer-Binding Protein-beta chemistry, Gene Expression Regulation, Enzymologic, Macrophages enzymology
- Abstract
Arginase activity is expressed by macrophages in healing wounds and other sites of inflammation and has been shown to modulate the synthesis of nitric oxide, polyamines, and collagen. The role of CCAAT/enhancer-binding protein beta (C/EBPbeta) in the regulation of macrophage arginase by different agonists was investigated using C/EBPbeta-/- and +/+ macrophage cell lines. 8-Bromo-cyclic adenosine monophosphate (8-Br-cAMP, 0.5 mM), recombinant murine interleukin 4 (rmIL-4, 20 U/mL), Escherichia coli lipopolysaccharide (100 ng/mL), and hypoxia (1% O2) induced arginase activity in C/EBPbeta+/+ macrophages, where enzyme activity correlated with arginase I protein. Only rmIL-4 increased arginase activity in C/EBPbeta-/- cells. Arginase II protein was expressed constitutively in wild-type and C/EBPbeta-/- cell lines and was unaltered by 8-Br-cAMP or rmIL-4. rmIL-4-stimulated immortalized C/EBPbeta-/- macrophages demonstrated higher nuclear signal transducer and activator of transcription-6 (STAT6) and phospho-STAT6 content than their +/+ counterparts. Validating the biological relevance of findings with the cell lines, additional experiments examined wound fluids and peritoneal macrophages from C/EBPbeta-/- mice and demonstrated that both contained less arginase activity than those from wild-type controls. Wounds in C/EBPbeta-/- animals showed signs of delayed maturation, as manifested by the persistence of neutrophils in the inflammatory infiltrate. Peritoneal macrophages from C/EBPbeta+/+ animals responded to 8-Br-cAMP and rmIL-4 with increased arginase activity, whereas those from C/EBPbeta-/- mice did not respond to cAMP. Results demonstrate a key mechanistic role for C/EBPbeta in the modulation of macrophage arginase I expression in vivo and in vitro.
- Published
- 2005
- Full Text
- View/download PDF
32. Enzymes of arginine metabolism.
- Author
-
Morris SM Jr
- Subjects
- Animals, Arginine biosynthesis, Arginine physiology, Gene Expression Regulation physiology, Humans, Proteins genetics, Tissue Distribution, Arginine metabolism, Enzymes metabolism
- Abstract
In mammals, L-arginine is classified as a semiessential or conditionally essential amino acid, depending on the developmental stage and health status of the individual. It can be derived from proline or glutamate, with the ultimate synthetic step catalyzed by argininosuccinate lyase. L-arginine is catabolized by arginases, nitric oxide synthases, arginine:glycine amidinotransferase, and possibly also by arginine decarboxylase, resulting ultimately in the production of urea, proline, glutamate, polyamines, nitric oxide, creatine, or agmatine. There is considerable diversity in tissue-specific and stimulus-dependent regulation of expression within this group of enzymes, and the expression of several of them can be regulated at transcriptional and translational levels by changes in the concentration of L-arginine itself. Consequently, the interplay among these enzymes in the regulation of specific aspects of arginine metabolism can be quite complex. For example, nitric oxide production can be affected by the interplay between nitric oxide synthases, arginases, and argininosuccinate synthetase. This metabolic complexity can pose challenges for analyses of arginine metabolism not only because L-arginine is a substrate for several different enzymes but also because ornithine and citrulline, key products of arginine metabolism, can each be produced by multiple enzymes. This overview highlights key features of the arginine metabolic enzymes and their interactions.
- Published
- 2004
- Full Text
- View/download PDF
33. Arginine therapy: a new treatment for pulmonary hypertension in sickle cell disease?
- Author
-
Morris CR, Morris SM Jr, Hagar W, Van Warmerdam J, Claster S, Kepka-Lenhart D, Machado L, Kuypers FA, and Vichinsky EP
- Subjects
- Administration, Oral, Adolescent, Adult, Amino Acids blood, Arginase blood, Arginase drug effects, Arginine metabolism, Biological Availability, Case-Control Studies, Echocardiography, Female, Humans, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary physiopathology, Male, Middle Aged, Nitric Oxide metabolism, Ornithine blood, Oximetry, Pulmonary Wedge Pressure drug effects, Treatment Outcome, Anemia, Sickle Cell complications, Arginine therapeutic use, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary etiology
- Abstract
Pulmonary hypertension is a life-threatening complication of sickle cell disease. L-Arginine is the nitrogen donor for synthesis of nitric oxide, a potent vasodilator that is deficient during times of sickle cell crisis. This deficiency may play a role in pulmonary hypertension. The enzyme arginase hydrolyzes arginine to ornithine and urea, and thus, it may compete with nitric oxide synthase, leading to decreased nitric oxide production. Nitric oxide therapy by inhalation has improved pulmonary hypertension associated with acute chest syndrome in sickle cell disease, and several studies demonstrate therapeutic benefits of arginine therapy for primary and secondary pulmonary hypertension. We sought to determine the effects of arginine therapy on pulmonary hypertension in patients with sickle cell disease. Arginase activity was also determined. Oral arginine produced a 15.2% mean reduction in estimated pulmonary artery systolic pressure (63.9 +/- 13 to 54.2 +/- 12 mm Hg, p = 0.002) after 5 days of therapy in 10 patients. Arginase activity was elevated almost twofold (p = 0.07) in patients with pulmonary hypertension and may limit arginine bioavailability. With limited treatment options and a high mortality rate for patients with sickle cell disease who develop pulmonary hypertension, arginine is a promising new therapy that warrants further investigation.
- Published
- 2003
- Full Text
- View/download PDF
34. Translational control of inducible nitric oxide synthase expression by arginine can explain the arginine paradox.
- Author
-
Lee J, Ryu H, Ferrante RJ, Morris SM Jr, and Ratan RR
- Subjects
- Animals, Arginase biosynthesis, Arginase genetics, Arginine metabolism, Astrocytes drug effects, Astrocytes metabolism, Cytokines pharmacology, Enzyme Stability drug effects, Extracellular Space metabolism, Humans, Mice, Models, Biological, Nitric Oxide biosynthesis, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type II, Protein Biosynthesis drug effects, Protein Kinases metabolism, Protein Serine-Threonine Kinases, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Arginine pharmacology, Nitric Oxide Synthase genetics
- Abstract
L-Arginine is the only endogenous nitrogen-containing substrate of NO synthase (NOS), and it thus governs the production of NO during nervous system development as well as in disease states such as stroke, multiple sclerosis, Parkinson's disease, and HIV dementia. The "arginine paradox" refers to the dependence of cellular NO production on exogenous L-arginine concentration despite the theoretical saturation of NOS enzymes with intracellular L-arginine. Herein, we report that decreased availability of L-arginine blocked induction of NO production in cytokine-stimulated astrocytes, owing to inhibition of inducible NOS (iNOS) protein expression. However, activity of the promoter of the iNOS gene, induction of iNOS mRNA, and stability of iNOS protein were not inhibited under these conditions. Our results indicate that inhibition of iNOS activity by arginine depletion in stimulated astrocyte cultures occurs via inhibition of translation of iNOS mRNA. After stimulation by cytokines, uptake of L-arginine negatively regulates the phosphorylation status of the eukaryotic initiation factor (eIF2 alpha), which, in turn, regulates translation of iNOS mRNA. eIF2 alpha phosphorylation correlates with phosphorylation of the mammalian homolog of yeast GCN2 eIF2 alpha kinase. As the kinase activity of GCN2 is activated by phosphorylation, these findings suggest that GCN2 activity represents a proximal step in the iNOS translational regulation by availability of l-arginine. These results provide an explanation for the arginine paradox for iNOS and define a distinct mechanism by which a substrate can regulate the activity of its associated enzyme.
- Published
- 2003
- Full Text
- View/download PDF
35. Cloning of human agmatinase. An alternate path for polyamine synthesis induced in liver by hepatitis B virus.
- Author
-
Mistry SK, Burwell TJ, Chambers RM, Rudolph-Owen L, Spaltmann F, Cook WJ, and Morris SM Jr
- Subjects
- Brain enzymology, Cloning, Molecular, Gene Expression Regulation, Enzymologic, Humans, Kidney enzymology, Molecular Sequence Data, Putrescine metabolism, RNA, Messenger analysis, Sequence Homology, Amino Acid, Hepatitis B metabolism, Hepatitis B virus, Liver enzymology, Polyamines metabolism, Ureohydrolases genetics, Ureohydrolases metabolism
- Abstract
Agmatinase, which hydrolyzes agmatine to putrescine and urea, not only represents a potentially important mechanism for regulating the biological effects of agmatine in mammalian cells but also represents an alternative to ornithine decarboxylase for polyamine biosynthesis. We have isolated a full-length cDNA encoding human agmatinase whose function was confirmed by complementation in yeast. The single-copy human agmatinase gene located on chromosome 1 encodes a 352-residue protein with a putative mitochondrial targeting sequence at the NH(3)-terminus. Human agmatinase has about 30% identity to bacterial agmatinases and <20% identity to mammalian arginases. Residues required for binding of Mn(2+) at the active site in bacterial agmatinase and other members of the arginase superfamily are fully conserved in human agmatinase. Agmatinase mRNA is most abundant in human liver and kidney but also is expressed in several other tissues, including skeletal muscle and brain. Its expression in human liver is induced during hepatitis B virus infection, suggesting that agmatinase may play a role in the pathophysiology of this disease.
- Published
- 2002
- Full Text
- View/download PDF
36. Activities of arginase I and II are limiting for endothelial cell proliferation.
- Author
-
Li H, Meininger CJ, Kelly KA, Hawker JR Jr, Morris SM Jr, and Wu G
- Subjects
- Animals, Arginase genetics, Cattle, Cell Division physiology, Cells, Cultured, Coronary Vessels cytology, Eflornithine pharmacology, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Enzymologic, Isoenzymes genetics, Lac Operon, Ornithine metabolism, Ornithine Decarboxylase Inhibitors, Polyamines metabolism, Putrescine metabolism, Spermidine metabolism, Transfection, Arginase metabolism, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Isoenzymes metabolism
- Abstract
Polyamines are essential for cell proliferation; therefore, we hypothesized that arginase I or arginase II activities, via production of ornithine for polyamine synthesis, may be limiting for proliferation of endothelial cells (EC). Bovine coronary venular EC stably transfected with a lacZ gene (lacZ-EC, control), rat arginase I cDNA (AI-EC), or mouse arginase II cDNA (AII-EC) were utilized to test this hypothesis. Cell-proliferation assays showed that EC proliferation was markedly increased in AI-EC and AII-EC compared with lacZ-EC. Expression of proliferating cell nuclear antigen was also enhanced in AI-EC and AII-EC. DL-alpha-difluoromethylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, was used to establish that increased polyamine synthesis was involved in mediating the enhanced growth of AI-EC and AII-EC. Addition of 5 mM DFMO to the culture medium completely abolished the differences in cellular putrescine concentrations and reduced the differences in spermidine concentrations among AI-EC, AII-EC, and lacZ-EC. The DFMO treatment also prevented an increase in AI-EC and AII-EC proliferation compared with lacZ-EC. Addition of 10 and 50 microM putrescine dose-dependently increased AI-EC, AII-EC, and lacZ-EC growth to the same extent. These results demonstrate that either arginase isoform can potentially play a role in modulating EC proliferation by regulating polyamine synthesis.
- Published
- 2002
- Full Text
- View/download PDF
37. Elevated arginase I expression in rat aortic smooth muscle cells increases cell proliferation.
- Author
-
Wei LH, Wu G, Morris SM Jr, and Ignarro LJ
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Arginase antagonists & inhibitors, Arginase metabolism, Biogenic Polyamines biosynthesis, Cells, Cultured, Enzyme Inhibitors pharmacology, Genetic Vectors, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Rats, Transfection, Aorta enzymology, Arginase genetics, Cell Division, Muscle, Smooth, Vascular enzymology
- Abstract
Arginase, which exists as the isoforms arginase I and II, catalyzes the hydrolysis of arginine to ornithine and urea. Ornithine is the principal precursor for production of polyamines, which are required for cell proliferation. Rat aortic smooth muscle cells (RASMC) contain constitutive arginase I, and arginase inhibitors cause inhibition of cell proliferation. The objective of this study was to determine whether the elevated expression of arginase I in RASMC causes increased cell proliferation. RASMC were stably transfected with either rat arginase I cDNA or a beta-galactosidase control expression plasmid. Western blots and arginase enzymatic assays revealed high-level expression of cytosolic arginase I in arginase I-transfected RASMC. Moreover, this observation was associated with the increased production of urea and polyamines and higher rates of RASMC proliferation. The two selective inhibitors of arginase, N(G)-hydroxy-l-arginine and S-(2-boronoethyl)-l-cysteine, inhibited arginase and decreased the production of urea and polyamines in arginase I-transfected RASMC, all of which were associated with the inhibition of cell proliferation. This study demonstrates that elevated arginase I expression increases RASMC proliferation by mechanisms involving increased production of polyamines. These observations suggest that arginase I plays a potentially important role in controlling RASMC proliferation.
- Published
- 2001
- Full Text
- View/download PDF
38. Alterations in arginine metabolic enzymes in trauma.
- Author
-
Bernard AC, Mistry SK, Morris SM Jr, O'Brien WE, Tsuei BJ, Maley ME, Shirley LA, Kearney PA, Boulanger BR, and Ochoa JB
- Subjects
- Animals, Arginase genetics, Arginase metabolism, Enzymes genetics, Gene Expression Regulation, Enzymologic, Isoenzymes, Mice, Mice, Inbred C3H, Nitric Oxide metabolism, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type II, RNA, Messenger metabolism, Arginine metabolism, Enzymes metabolism, Spleen metabolism, Wounds and Injuries metabolism
- Abstract
Arginine is the sole substrate for nitric oxide (NO) synthesis by NO synthases (NOS) and promotes the proliferation and maturation of human T-cells. Arginine is also metabolized by the enzyme arginase, producing urea and ornithine, the precursor for polyamine production. We sought to determine the molecular mechanisms regulating arginase and NOS in splenic immune cells after trauma. C3H/HeN mice underwent laparotomy as simulated moderate trauma or anesthesia alone (n = 24 per group). Six, 12, 24, or 48 h later, 6 animals from each group were sacrificed, and splenectomy was performed and plasma collected. Six separate animals had neither surgery nor anesthesia and were sacrificed to provide resting values (t = 0 h). Spleen arginase I and II and iNOS mRNA abundance, arginase I protein expression, and arginase activity were determined. Plasma NO metabolites (nitrite + nitrate) were also measured. Trauma increased spleen arginase I protein expression and activity (P = 0.01) within 12 and for at least 48 h after injury and coincided with up-regulated arginase I mRNA abundance at 24 h. Neither arginase II nor iNOS mRNA abundance in the spleen was significantly increased by trauma at 24 h. Plasma nitrite + nitrate was decreased in animals 48 h post-injury compared to anesthesia controls (P < 0.05). Trauma induces up-regulation of arginase I gene expression in splenic immune cells within 24 h of injury. Arginase II is not significantly up-regulated at that time point. Arginase I, rather than iNOS appears to be the dominant route for arginine metabolism in splenic immune cells 24 h after trauma.
- Published
- 2001
- Full Text
- View/download PDF
39. Arginase I expression and activity in human mononuclear cells after injury.
- Author
-
Ochoa JB, Bernard AC, O'Brien WE, Griffen MM, Maley ME, Rockich AK, Tsuei BJ, Boulanger BR, Kearney PA, and Morris SM Jr
- Subjects
- Adult, Aged, Biomarkers, Case-Control Studies, Citrulline blood, Female, Humans, Male, Middle Aged, Nitric Oxide blood, Ornithine blood, Prognosis, Prospective Studies, Statistics, Nonparametric, Trauma Severity Indices, Wounds and Injuries diagnosis, Arginase blood, Leukocytes, Mononuclear metabolism, Wounds and Injuries immunology
- Abstract
Objective: To determine the effect of trauma on arginase, an arginine-metabolizing enzyme, in cells of the immune system in humans., Summary Background Data: Arginase, classically considered an enzyme exclusive to the liver, is now known to exist in cells of the immune system. Arginase expression is induced in these cells by cytokines interleukin (IL) 4, IL-10, and transforming growth factor beta, corresponding to a T-helper 2 cytokine profile. In contrast, nitric oxide synthase expression is induced by IL-1, tumor necrosis factor, and gamma interferon, a T-helper 1 cytokine profile. Trauma is associated with a decrease in the production of nitric oxide metabolites and a state of immunosuppression characterized by an increase in the production of IL-4, IL-10, and transforming growth factor beta. This study tests the hypothesis that trauma increases arginase activity and expression in cells of the immune system., Methods: Seventeen severely traumatized patients were prospectively followed up in the intensive care unit for 7 days. Twenty volunteers served as controls. Peripheral mononuclear cells were isolated and assayed for arginase activity and expression, and plasma was collected for evaluation of levels of arginine, citrulline, ornithine, nitrogen oxides, and IL-10., Results: Markedly increased mononuclear cell arginase activity was observed early after trauma and persisted throughout the intensive care unit stay. Increased arginase activity corresponded with increased arginase I expression. Increased arginase activity coincided with decreased plasma arginine concentration. Plasma arginine and citrulline levels were decreased throughout the study period. Ornithine levels decreased early after injury but recovered by postinjury day 3. Increased arginase activity correlated with the severity of trauma, early alterations in lactate level, and increased levels of circulating IL-10. Increased arginase activity was associated with an increase in length of stay. Plasma nitric oxide metabolites were decreased during this same period., Conclusions: Markedly altered arginase expression and activity in cells of the human immune system after trauma have not been reported previously. Increased mononuclear cell arginase may partially explain the benefit of arginine supplementation for trauma patients. Arginase, rather than nitric oxide synthase, appears to be the dominant route for arginine metabolism in immune cells after trauma.
- Published
- 2001
- Full Text
- View/download PDF
40. Generation of a mouse model for arginase II deficiency by targeted disruption of the arginase II gene.
- Author
-
Shi O, Morris SM Jr, Zoghbi H, Porter CW, and O'Brien WE
- Subjects
- Amino Acids blood, Animals, Arginase physiology, Arginine blood, Base Sequence, DNA Primers genetics, Gene Targeting, Humans, Mice, Mice, Knockout, Models, Animal, Phenotype, Polyamines metabolism, Arginase genetics, Hyperargininemia
- Abstract
Mammals express two isoforms of arginase, designated types I and II. Arginase I is a component of the urea cycle, and inherited defects in arginase I have deleterious consequences in humans. In contrast, the physiologic role of arginase II has not been defined, and no deficiencies in arginase II have been identified in humans. Mice with a disruption in the arginase II gene were created to investigate the role of this enzyme. Homozygous arginase II-deficient mice were viable and apparently indistinguishable from wild-type mice, except for an elevated plasma arginine level which indicates that arginase II plays an important role in arginine homeostasis.
- Published
- 2001
- Full Text
- View/download PDF
41. Regulatory role of arginase I and II in nitric oxide, polyamine, and proline syntheses in endothelial cells.
- Author
-
Li H, Meininger CJ, Hawker JR Jr, Haynes TE, Kepka-Lenhart D, Mistry SK, Morris SM Jr, and Wu G
- Subjects
- Animals, Arginase genetics, Arginine pharmacokinetics, Carbon Radioisotopes, Cattle, Cells, Cultured, Coronary Vessels cytology, Cytosol enzymology, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Enzyme Activation physiology, Gene Expression Regulation, Enzymologic, Glutamic Acid biosynthesis, Isoenzymes genetics, Lac Operon, Microcirculation physiology, Mitochondria enzymology, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Ornithine metabolism, Rats, Transfection, Arginase metabolism, Coronary Vessels enzymology, Isoenzymes metabolism, Nitric Oxide biosynthesis, Polyamines metabolism, Proline biosynthesis
- Abstract
Endothelial cells (EC) metabolize L-arginine mainly by arginase, which exists as two distinct isoforms, arginase I and II. To understand the roles of arginase isoforms in EC arginine metabolism, bovine coronary venular EC were stably transfected with the Escherichia coli lacZ gene (lacZ-EC, control), rat arginase I cDNA (AI-EC), or mouse arginase II cDNA (AII-EC). Western blots and enzymatic assays confirmed high-level expression of arginase I in the cytosol of AI-EC and of arginase II in mitochondria of AII-EC. For determining arginine catabolism, EC were cultured for 24 h in DMEM containing 0.4 mM L-arginine plus [1-(14)C]arginine. Urea formation, which accounted for nearly all arginine consumption by these cells, was enhanced by 616 and 157% in AI-EC and AII-EC, respectively, compared with lacZ-EC. Arginine uptake was 31-33% greater in AI-EC and AII-EC than in lacZ-EC. Intracellular arginine content was 25 and 11% lower in AI-EC and AII-EC, respectively, compared with lacZ-EC. Basal nitric oxide (NO) production was reduced by 60% in AI-EC and by 47% in AII-EC. Glutamate and proline production from arginine increased by 164 and 928% in AI-EC and by 79 and 295% in AII-EC, respectively, compared with lacZ-EC. Intracellular content of putrescine and spermidine was increased by 275 and 53% in AI-EC and by 158 and 43% in AII-EC, respectively, compared with lacZ-EC. Our results indicate that arginase expression can modulate NO synthesis in bovine venular EC and that basal levels of arginase I and II are limiting for endothelial syntheses of polyamines, proline, and glutamate and may have important implications for wound healing, angiogenesis, and cardiovascular function.
- Published
- 2001
- Full Text
- View/download PDF
42. Arginase I: a limiting factor for nitric oxide and polyamine synthesis by activated macrophages?
- Author
-
Kepka-Lenhart D, Mistry SK, Wu G, and Morris SM Jr
- Subjects
- 8-Bromo Cyclic Adenosine Monophosphate pharmacology, Animals, Arginine pharmacology, Cell Line, Gene Expression Regulation, Enzymologic, Genes, Reporter, Macrophages drug effects, Mice, Putrescine metabolism, Transfection, beta-Galactosidase genetics, Arginase genetics, Arginase metabolism, Lipopolysaccharides pharmacology, Macrophage Activation physiology, Macrophages physiology, Nitric Oxide biosynthesis, Polyamines metabolism
- Abstract
Because arginase hydrolyzes arginine to produce ornithine and urea, it has the potential to regulate nitric oxide (NO) and polyamine synthesis. We tested whether expression of the cytosolic isoform of arginase (arginase I) was limiting for NO or polyamine production by activated RAW 264.7 macrophage cells. RAW 264.7 cells, stably transfected to overexpress arginase I or beta-galactosidase, were treated with interferon-gamma to induce type 2 NO synthase or with lipopolysaccharide or 8-bromo-cAMP (8-BrcAMP) to induce ornithine decarboxylase. Overexpression of arginase I had no effect on NO synthesis. In contrast, cells overexpressing arginase I produced twice as much putrescine after activation than did cells expressing beta-galactosidase. Cells overexpressing arginase I also produced more spermidine after treatment with 8-BrcAMP than did cells expressing beta-galactosidase. Thus endogenous levels of arginase I are limiting for polyamine synthesis, but not for NO synthesis, by activated macrophage cells. This study also demonstrates that it is possible to alter arginase I levels sufficiently to affect polyamine synthesis without affecting induced NO synthesis.
- Published
- 2000
- Full Text
- View/download PDF
43. A cortisol surge mediates the enhanced expression of pig intestinal pyrroline-5-carboxylate synthase during weaning.
- Author
-
Wu G, Meininger CJ, Kelly K, Watford M, and Morris SM Jr
- Subjects
- Animals, Citrulline biosynthesis, Female, Glutamine metabolism, Male, Ornithine-Oxo-Acid Transaminase genetics, Pulsatile Flow, RNA, Messenger metabolism, Swine, Hydrocortisone blood, Intestines enzymology, Ornithine-Oxo-Acid Transaminase biosynthesis, Weaning
- Abstract
Citrulline synthesis from glutamine is enhanced remarkably in enterocytes of weanling pigs, but the molecular mechanism(s) involved are not known. The objective of this study was to determine whether a cortisol surge mediates the enhanced expression of intestinal citrulline-synthetic enzymes during weaning. Jejunal enterocytes were prepared from 29-d-old weanling pigs treated with or without metyrapone (an inhibitor of cortisol synthesis), or from age-matched unweaned pigs. The mRNA levels and activities of phosphate-dependent glutaminase (PDG), pyrroline-5-carboxylate synthase (P5CS), ornithine aminotransferase (OAT), carbamoyl-phosphate synthase I (CPS-I) and ornithine carbamoyltransferase (OCT) were determined. The mRNA levels for PDG, P5CS, OAT and OCT were 139, 157, 102 and 55% higher, respectively, in weanling pigs compared with suckling pigs. The activities of PDG and P5CS were 38 and 692% higher, respectively, in weanling pigs compared with unweaned pigs, but the activities of OAT, CPS-I and OCT did not differ between these two groups of pigs. The effects of metyrapone administration to weanling pigs were as follows: 1) prevention of a cortisol surge, 2) abolition of the increases in both mRNA levels and activity of P5CS, 3) no alteration in the mRNA levels and activities of PDG and CPS-I, 4) increases in the mRNA levels for OAT (216%) and OCT (39%) and in OAT activity (30%), and 5) prevention of the increase in intestinal synthesis of citrulline from glutamine. These results suggest that increased P5CS activity reflects in large part the increased levels of P5CS mRNA and is responsible for the increased synthesis of citrulline from glutamine in enterocytes of weanling pigs; these increases may be mediated by a cortisol surge during weaning that can be blocked by metyrapone administration.
- Published
- 2000
- Full Text
- View/download PDF
44. IL-4 and IL-13 upregulate arginase I expression by cAMP and JAK/STAT6 pathways in vascular smooth muscle cells.
- Author
-
Wei LH, Jacobs AT, Morris SM Jr, and Ignarro LJ
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Cell Division drug effects, Cells, Cultured, Dexamethasone pharmacology, Enzyme Induction drug effects, Glucocorticoids pharmacology, Interferon-gamma pharmacology, Muscle, Smooth, Vascular cytology, Phosphorylation, Rats, STAT6 Transcription Factor, Tyrosine metabolism, Up-Regulation, Arginase metabolism, Cyclic AMP metabolism, Interleukin-13 pharmacology, Interleukin-4 pharmacology, Isoenzymes metabolism, Muscle, Smooth, Vascular metabolism, Protein-Tyrosine Kinases metabolism, Trans-Activators metabolism
- Abstract
The objectives of this study were to determine whether rat aortic smooth muscle cells (RASMC) express arginase and to elucidate the possible mechanisms involved in the regulation of arginase expression. The results show that RASMC contain basal arginase I (AI) activity, which is significantly enhanced by stimulating the cells with either interleukin (IL)-4 or IL-13, but arginase II (AII) expression was not detected under any condition studied here. We further investigated the signal transduction pathways responsible for AI induction. AI mRNA and protein levels were enhanced by addition of forskolin (1 microM) and inhibited by H-89 (30 microM), suggesting positive regulation of AI by a protein kinase A pathway. Genistein (10 microgramg/ml) and sodium orthovanadate (Na(3)VO(4); 10 microM) were used to investigate the role of tyrosine phosphorylation in the control of AI expression. Genistein inhibited, whereas Na(3)VO(4) enhanced the induction of AI by IL-4 or IL-13. Along with immunoprecipitation and immunoblot analyses, these data implicate the JAK/STAT6 pathway in AI regulation. Dexamethasone (Dex) and interferon (IFN)-gamma were investigated for their effects on AI induction. Dex (1 microM) and IFN-gamma (100 U/ml) alone had no effect on basal AI expression in RASMC, but both reduced AI induction by IL-4 and IL-13. In combination, Dex and IFN-gamma abolished AI induction by IL-4 and IL-13. Finally, both IL-4 and IL-13 significantly increased RASMC DNA synthesis as monitored by [(3)H]thymidine incorporation, demonstrating that upregulation of AI is correlated with an increase in cell proliferation. Blockade of AI induction by IFN-gamma, H-89, or genistein also blocked the increase in cell proliferation. These observations are consistent with the possibility that upregulation of AI might play an important role in the pathophysiology of vascular disorders characterized by excessive smooth muscle growth.
- Published
- 2000
- Full Text
- View/download PDF
45. cAMP induces CD14 expression in murine macrophages via increased transcription.
- Author
-
Liu S, Morris SM Jr, Nie S, Shapiro RA, and Billiar TR
- Subjects
- 1-Methyl-3-isobutylxanthine pharmacology, 8-Bromo Cyclic Adenosine Monophosphate metabolism, 8-Bromo Cyclic Adenosine Monophosphate pharmacology, Animals, Cell Line, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Cyclic AMP-Dependent Protein Kinases metabolism, Gene Expression, Lipopolysaccharide Receptors biosynthesis, Macrophages cytology, Macrophages drug effects, Macrophages, Peritoneal cytology, Macrophages, Peritoneal drug effects, Male, Mice, Phosphodiesterase Inhibitors pharmacology, Rats, Rats, Sprague-Dawley, Solubility, Cyclic AMP metabolism, Gene Expression Regulation drug effects, Lipopolysaccharide Receptors genetics, Macrophages metabolism, Macrophages, Peritoneal metabolism, Transcription, Genetic drug effects
- Abstract
CD14, a glycoprotein that binds bacterial lipopolysaccharide, plays a critical role in the inflammatory response to infection by gram-negative bacteria. Studies were undertaken to determine whether cyclic adenosine monophosphate (cAMP) regulates CD14 expression in macrophages. Incubation of RAW 264.7 cells with 8-Br-cAMP resulted in a significant increase in steady-state CD14 mRNA levels. The increase in mRNA levels was also associated with both cell-associated and soluble CD14 protein. H89 completely blocked the 8-Br-cAMP-induced CD14 mRNA up-regulation. There was no change in CD 14 mRNA half-life in the presence of 8-Br-cAMP. The CD14 gene transcription rate was increased about twofold after exposure to 8-Br-cAMP. cAMP-dependent increases in CD14 mRNA were also observed in rat peritoneal macrophages, demonstrating that this is an authentic response of mature macrophages. This study provides evidence that cAMP and protein kinase A are important regulators of CD14 expression in macrophages.
- Published
- 2000
- Full Text
- View/download PDF
46. Salicylate-enhanced activation of transcription factors induced by interferon-gamma.
- Author
-
Chen LC, Kepka-Lenhart D, Wright TM, and Morris SM Jr
- Subjects
- Animals, Aspirin pharmacology, Cells, Cultured, Electrophoresis, Polyacrylamide Gel, Interferon Regulatory Factor-1, Mice, RNA, Messenger metabolism, STAT1 Transcription Factor, Trans-Activators metabolism, DNA-Binding Proteins metabolism, Interferon-gamma pharmacology, Phosphoproteins metabolism, Salicylates pharmacology, Transcription Factors genetics, Transcriptional Activation drug effects
- Abstract
Salicylate enhanced the interferon-gamma-dependent activation of two transcription factors in a murine macrophage cell line: signal transducer and activator of transcription (STAT)1 and interferon-gamma-responsive factor 1. Salicylate alone did not activate these transcription factors. This enhancement was reflected by increased DNA-binding activities and was the consequence of prolonged tyrosine phosphorylation of these transcription factors following interferon-gamma treatment. However, salicylate did not directly inhibit protein-tyrosine phosphatase activity in nuclear extracts of interferon-gamma-treated cells. The enhanced activation of STAT1 resulted in increased induction of mRNA encoding interferon regulatory factor-1. These results not only demonstrate that aspirin and its metabolite salicylate may have pro-inflammatory as well as anti-inflammatory effects but also raise the possibility that new cellular targets may be identified for modulating the Janus kinase-STAT signalling pathway.
- Published
- 1999
47. Glucocorticoids mediate the enhanced expression of intestinal type II arginase and argininosuccinate lyase in postweaning pigs.
- Author
-
Flynn NE, Meininger CJ, Kelly K, Ing NH, Morris SM Jr, and Wu G
- Subjects
- Analysis of Variance, Animals, Animals, Suckling, Arginase classification, Intestine, Small drug effects, RNA, Messenger isolation & purification, Swine, Weaning, Arginase metabolism, Argininosuccinate Lyase metabolism, Glucocorticoids physiology, Hormone Antagonists pharmacology, Intestine, Small enzymology, Mifepristone pharmacology
- Abstract
Arginine metabolism is enhanced in the small intestine of weanling pigs, but the molecular mechanism(s) involved is not known. The objectives of this study were to determine the following: 1) whether glucocorticoids play a role in induction of intestinal arginine metabolic enzymes during weaning; 2) whether the induction of enzyme activities was due to increases in corresponding mRNA levels; and 3) the identity of the arginase isoform(s) expressed in the small intestine. Jejunum was obtained from 29-d-old weaned pigs that were or were not treated with 17-beta-hydroxy-11beta-(4-dimethylaminophenyl)17alpha-(prop- 1-ynyl)es tra-4,9-dien-3-one (RU486, an antagonist of glucocorticoid receptors), or from age-matched suckling pigs. Activities and mRNA levels for type I and type II arginases, argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL) were determined. Activities of arginase, ASL and ASS increased by 635, 56 and 106%, respectively, in weanling pigs, compared with suckling pigs. RU486 treatment attenuated the increase in arginase activity by 74% and completely prevented the ASL induction in weanling pigs, but had no effect on ASS activity. Pig intestine expresses both type I and type II arginases. On the basis of immunoblot analyses, there was no significant difference in levels of intestinal type I arginase among these three groups of pigs, indicating that changes in arginase activity were due only to type II arginase. The mRNA levels for type II arginase and ASL increased by 135 and 198%, respectively, in weanling pigs compared with suckling pigs, and this induction was completely prevented by RU486. In contrast, ASS mRNA levels did not differ between suckling and weanling pigs. These results suggest that intestinal type II arginase, ASS and ASL are regulated differentially at transcriptional and post-translational levels and that glucocorticoids play a major role in the induction of type II arginase and ASL mRNAs in the small intestine of weanling pigs.
- Published
- 1999
- Full Text
- View/download PDF
48. Isolation and characterization of a human hepatic epithelial-like cell line (AKN-1) from a normal liver.
- Author
-
Nussler AK, Vergani G, Gollin SM, Dorko K, Morris SM Jr, Demetris AJ, Nomoto M, Beger HG, and Strom SC
- Subjects
- Biomarkers, Cell Differentiation, Epithelial Cells physiology, Humans, Karyotyping, Liver physiology, Cell Line, Epithelial Cells cytology, Liver cytology
- Abstract
The isolation and characterization of human liver cell lines are rather difficult due to limited material and poor growth in cell culture. In this report, we present the isolation, culture and characterization of a new epithelial-like liver cell line (AKN-1) with a heterogeneous cell population and many characteristics of the biliary epithelium. The AKN-1 cell line stained positively with antibodies to epithelial cytokeratin polypetides CK 8, 18, and 19. In addition, the cell line expressed the anti-human epithelial-related antigen (MOC-31), the human epithelial antigen (HEA), and the gamma-glutamyl transpeptidase, the hematopoietic growth factor, stem cell factor, and also its receptor, c-kit. The cell line failed to express albumin and factor 8 by immunohistochemistry. It did show, however, a twofold increase in 7-ethoxyresorufin-O-deethylase activity. Cytogenetic characterization revealed rare breakpoints in chromosome 2, which to our knowledge, have not yet been reported in liver cells.
- Published
- 1999
- Full Text
- View/download PDF
49. Arginine metabolism: nitric oxide and beyond.
- Author
-
Wu G and Morris SM Jr
- Subjects
- Amidinotransferases metabolism, Animals, Arginase metabolism, Arginine biosynthesis, Biological Transport, Carboxy-Lyases metabolism, Intestinal Mucosa metabolism, Kidney metabolism, Liver metabolism, Nitric Oxide Synthase metabolism, Arginine metabolism, Nitric Oxide metabolism
- Abstract
Arginine is one of the most versatile amino acids in animal cells, serving as a precursor for the synthesis not only of proteins but also of nitric oxide, urea, polyamines, proline, glutamate, creatine and agmatine. Of the enzymes that catalyse rate-controlling steps in arginine synthesis and catabolism, argininosuccinate synthase, the two arginase isoenzymes, the three nitric oxide synthase isoenzymes and arginine decarboxylase have been recognized in recent years as key factors in regulating newly identified aspects of arginine metabolism. In particular, changes in the activities of argininosuccinate synthase, the arginases, the inducible isoenzyme of nitric oxide synthase and also cationic amino acid transporters play major roles in determining the metabolic fates of arginine in health and disease, and recent studies have identified complex patterns of interaction among these enzymes. There is growing interest in the potential roles of the arginase isoenzymes as regulators of the synthesis of nitric oxide, polyamines, proline and glutamate. Physiological roles and relationships between the pathways of arginine synthesis and catabolism in vivo are complex and difficult to analyse, owing to compartmentalized expression of various enzymes at both organ (e.g. liver, small intestine and kidney) and subcellular (cytosol and mitochondria) levels, as well as to changes in expression during development and in response to diet, hormones and cytokines. The ongoing development of new cell lines and animal models using cDNA clones and genes for key arginine metabolic enzymes will provide new approaches more clearly elucidating the physiological roles of these enzymes.
- Published
- 1998
- Full Text
- View/download PDF
50. Multiple NF-kappaB enhancer elements regulate cytokine induction of the human inducible nitric oxide synthase gene.
- Author
-
Taylor BS, de Vera ME, Ganster RW, Wang Q, Shapiro RA, Morris SM Jr, Billiar TR, and Geller DA
- Subjects
- Base Sequence, Cell Line, DNA Mutational Analysis, DNA-Binding Proteins analysis, Humans, Interleukin-1 pharmacology, Molecular Sequence Data, Mutagenesis, Site-Directed genetics, Nitric Oxide Synthase Type II, Nuclear Proteins analysis, Proline analogs & derivatives, Proline pharmacology, Promoter Regions, Genetic genetics, RNA, Messenger metabolism, Sequence Analysis, DNA, Thiocarbamates pharmacology, Tumor Necrosis Factor-alpha pharmacology, Cytokines pharmacology, Enhancer Elements, Genetic genetics, Gene Expression Regulation, Enzymologic drug effects, NF-kappa B genetics, Nitric Oxide Synthase genetics
- Abstract
The human inducible nitric oxide synthase (iNOS) gene is overexpressed in a number of human inflammatory diseases. Previously, we observed that the human iNOS gene is transcriptionally regulated by cytokines and demonstrated that the cytokine-responsive regions are upstream of -3.8 kilobase pairs (kb). Therefore, the purpose of this study was to further localize the functional enhancer elements and to assess the role of the transcription factor NF-kappaB in both human liver (AKN-1) and human lung (A549) epithelial cell lines. The addition of NF-kappaB inhibitors significantly suppressed cytokine-stimulated iNOS mRNA expression and NO synthesis, indicating that NF-kappaB is involved in the induction of the human iNOS gene. Analysis of the first 4.7 kb of the 5'-flanking region demonstrated basal promoter activity and failed to show any cytokine-inducible activity. However, promoter constructs extending to -5.8 and -7.2 kb revealed 2-3-fold and 4-5-fold induction, respectively, in the presence of cytokines. DNA sequence analysis from -3.8 to -7.2 kb identified five putative NF-kappaB cis-regulatory transcription factor binding sites upstream of -4.7 kb. Site-directed mutagenesis of these sites revealed that the NF-kappaB motif at -5.8 kb is required for cytokine-induced promoter activity, while the sites at -5.2, -5.5, and -6.1 kb elicit a cooperative effect. Electromobility shift assays using a site-specific oligonucleotide and nuclear extracts from cells stimulated with cytokine-mixture, tumor necrosis factor-alpha or interleukin-1beta, but not interferon-gamma, exhibited inducible DNA binding activity for NF-kappaB. These data indicate that NF-kappaB activation is required for cytokine induction of the human iNOS gene and identifies four NF-kappaB enhancer elements upstream in the human iNOS promoter that confer inducibility to tumor necrosis factor-alpha and interleukin-1beta.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.