125 results on '"Morris SM Jr"'
Search Results
2. Amplified expression profiling of platelet transcriptome reveals changes in arginine metabolic pathways in patients with sickle cell disease.
- Author
-
Raghavachari N, Xu X, Harris A, Villagra J, Logun C, Barb J, Solomon MA, Suffredini AF, Danner RL, Kato G, Munson PJ, Morris SM Jr, Gladwin MT, Raghavachari, Nalini, Xu, Xiuli, Harris, Amy, Villagra, Jose, Logun, Carolea, Barb, Jennifer, and Solomon, Michael A
- Published
- 2007
3. 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
4. Nitric oxide, sepsis, and arginine metabolism.
- Author
-
Kelly E, Morris SM Jr., Billiar TR, Kelly, E, Morris, S M Jr, and Billiar, T R
- Published
- 1995
- Full Text
- View/download PDF
5. Arginine metabolism, pulmonary hypertension, and sickle cell disease.
- Author
-
Waugh WH, Kielstein JT, Cooke JP, Morris CR, Vichinsky EP, Kato GJ, Gladwin MT, Hazen S, Morris SM Jr., and Waugh, William H
- Published
- 2005
- Full Text
- View/download PDF
6. 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
7. 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
8. 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
9. 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
10. 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
11. 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
12. 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
13. 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
14. 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
15. 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
16. The central role of arginine catabolism in T-cell dysfunction and increased susceptibility to infection after physical injury.
- Author
-
Zhu X, Pribis JP, Rodriguez PC, Morris SM Jr, Vodovotz Y, Billiar TR, and Ochoa JB
- Subjects
- Animals, Disease Models, Animal, Listeriosis physiopathology, Mice, Mice, Inbred C57BL, Wounds and Injuries physiopathology, Arginase biosynthesis, Arginine biosynthesis, Listeriosis immunology, Myeloid Cells metabolism, T-Lymphocytes metabolism, Wounds and Injuries immunology
- Abstract
Objective: To explore the hypothesis that decreased arginine availability by myeloid-derived suppressor cells (MDSCs) is a cause of T-cell dysfunction after physical injury (PI)., Background: Arginine is an essential amino acid for normal T-cell function whose availability becomes limited after PI. MDSCs expressing arginase 1 are induced by PI. T-cell dysfunction after PI seems to increase the risk of infection but the mechanisms that cause it are unclear., Methods: PI was created using a standard laparotomy model. Phenotypical and functional alterations in T cells were evaluated in vivo. MDSCs expressing arginase 1 were measured by flow cytometry. Infection after PI was created by intraperitoneal injection of Listeria monocytogenes. N-Hydroxy-Nor-L-arginine (Nor-NOHA) was used as an arginase inhibitor. The effect of arginine depletion on T-cell function and susceptibility to infection was assessed through adoptive transfer of MDSC or injection of arginase into noninjured mice., Results: PI caused a decrease in intracellular arginine in T cells, loss of the T-cell receptor (TCR) CD3-ζ chain, inhibition of in vivo T-cell proliferation, memory, and cytotoxicity. PI exponentially increased bacterial growth and mortality to L. monocytogenes. T-cell dysfunction and increased infection were reversed by arginase inhibitor Nor-NOHA but were reproduced by adoptively transferring MDSC or injecting arginase 1 to noninjured mice., Conclusions: Arginine availability is decreased after PI coinciding with an induction of MDSC expressing arginase 1. Decreased arginine may inhibit T-cell function and increase susceptibility to infection after injury.
- Published
- 2014
- Full Text
- View/download PDF
17. 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
18. Shaping the murine macrophage phenotype: IL-4 and cyclic AMP synergistically activate the arginase I promoter.
- Author
-
Sheldon KE, Shandilya H, Kepka-Lenhart D, Poljakovic M, Ghosh A, and Morris SM Jr
- Subjects
- Animals, Arginase genetics, Arginase immunology, Cell Line, Chromatin Immunoprecipitation, Cyclic AMP immunology, Immunoblotting, Interleukin-4 immunology, Macrophages immunology, Mice, Phenotype, Polymerase Chain Reaction, Transfection, Arginase biosynthesis, Cyclic AMP metabolism, Gene Expression Regulation immunology, Interleukin-4 metabolism, Macrophage Activation physiology, Macrophages metabolism, Promoter Regions, Genetic genetics
- Abstract
Arginase I is a marker of murine M2 macrophages and is highly expressed in many inflammatory diseases. The basis for high arginase I expression in macrophages in vivo is incompletely understood but likely reflects integrated responses to combinations of stimuli. Our objective was to elucidate mechanisms involved in modulating arginase I induction by IL-4, the prototypical activator of M2 macrophages. IL-4 and 8-bromo-cAMP individually induce arginase I, but together they rapidly and synergistically induce arginase I mRNA, protein, and promoter activity in murine macrophage cells. Arginase I induction by IL-4 requires binding of the transcription factors STAT6 and C/EBPβ to the IL-4 response element of the arginase I gene. Chromatin immunoprecipitation showed that the synergistic response involves binding of both transcription factors to the IL-4 response element at levels significantly greater than in response to IL-4 alone. The results suggest that C/EBPβ is a limiting factor for the level of STAT6 bound to the IL-4 response element. The enhanced binding in the synergistic response was not due to increased expression of either STAT6 or C/EBPβ but was correlated primarily with increased nuclear abundance of C/EBPβ. Our findings also suggest that induction of arginase I expression is stochastic; that is, differences in induction reflect differences in probability of transcriptional activation and not simply differences in rate of transcription. Results of the present study also may be useful for understanding mechanisms underlying regulated expression of other genes in macrophages and other myeloid-derived cells in health and disease.
- Published
- 2013
- Full Text
- View/download PDF
19. Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms.
- Author
-
Mattila JT, Ojo OO, Kepka-Lenhart D, Marino S, Kim JH, Eum SY, Via LE, Barry CE 3rd, Klein E, Kirschner DE, Morris SM Jr, Lin PL, and Flynn JL
- Subjects
- Animals, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic metabolism, CD11c Antigen metabolism, Cellular Microenvironment, Humans, Leukocyte L1 Antigen Complex metabolism, Lung microbiology, Lung pathology, Macaca, Mycobacterium tuberculosis immunology, Myeloid Cells, Neutrophils metabolism, Protein Isoforms metabolism, Receptors, Cell Surface metabolism, Tuberculosis microbiology, Tuberculosis pathology, Arginase metabolism, Granuloma immunology, Macrophages cytology, Nitric Oxide Synthase Type II metabolism, Nitric Oxide Synthase Type III metabolism, Tuberculosis immunology
- Abstract
Macrophages in granulomas are both antimycobacterial effector and host cell for Mycobacterium tuberculosis, yet basic aspects of macrophage diversity and function within the complex structures of granulomas remain poorly understood. To address this, we examined myeloid cell phenotypes and expression of enzymes correlated with host defense in macaque and human granulomas. Macaque granulomas had upregulated inducible and endothelial NO synthase (iNOS and eNOS) and arginase (Arg1 and Arg2) expression and enzyme activity compared with nongranulomatous tissue. Immunohistochemical analysis indicated macrophages adjacent to uninvolved normal tissue were more likely to express CD163, whereas epithelioid macrophages in regions where bacteria reside strongly expressed CD11c, CD68, and HAM56. Calprotectin-positive neutrophils were abundant in regions adjacent to caseum. iNOS, eNOS, Arg1, and Arg2 proteins were identified in macrophages and localized similarly in granulomas across species, with greater eNOS expression and ratio of iNOS/Arg1 expression in epithelioid macrophages as compared with cells in the lymphocyte cuff. iNOS, Arg1, and Arg2 expression in neutrophils was also identified. The combination of phenotypic and functional markers support that macrophages with anti-inflammatory phenotypes localized to outer regions of granulomas, whereas the inner regions were more likely to contain macrophages with proinflammatory, presumably bactericidal, phenotypes. Together, these data support the concept that granulomas have organized microenvironments that balance antimicrobial anti-inflammatory responses to limit pathology in the lungs.
- Published
- 2013
- Full Text
- View/download PDF
20. Retinoic acid promotes the development of Arg1-expressing dendritic cells for the regulation of T-cell differentiation.
- Author
-
Chang J, Thangamani S, Kim MH, Ulrich B, Morris SM Jr, and Kim CH
- Subjects
- Animals, Arginase genetics, Binding Sites, Dendritic Cells metabolism, Forkhead Transcription Factors metabolism, Gene Expression Regulation drug effects, Interleukin-4 pharmacology, Mice, Mice, Knockout, Promoter Regions, Genetic, Receptors, Retinoic Acid metabolism, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, Arginase metabolism, Cell Differentiation immunology, Dendritic Cells drug effects, Dendritic Cells immunology, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, Tretinoin pharmacology
- Abstract
Arginase I (Arg1), an enzyme expressed by many cell types including myeloid cells, can regulate immune responses. Expression of Arg1 in myeloid cells is regulated by a number of cytokines and tissue factors that influence cell development and activation. Retinoic acid, produced from vitamin A, regulates the homing and differentiation of lymphocytes and plays important roles in the regulation of immunity and immune tolerance. We report here that optimal expression of Arg1 in DCs requires retinoic acid. Induction of Arg1 by retinoic acid is directly mediated by retinoic acid-responsive elements in the 5' noncoding region of the Arg1 gene. Arg1, produced by DCs in response to retinoic acid, promotes the generation of FoxP3(+) regulatory T (Treg) cells. Importantly, blocking the retinoic acid receptor makes DCs hypo-responsive to known inducers of Arg1 such as IL-4 and GM-CSF in Arg1 expression. We found that intestinal CD103(+) DCs that are known to produce retinoic acid highly express Arg1. Our results establish retinoic acid as a key signal in expression of Arg1 in DCs., (© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2013
- Full Text
- View/download PDF
21. 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
22. Adenosine promotes alternative macrophage activation via A2A and A2B receptors.
- Author
-
Csóka B, Selmeczy Z, Koscsó B, Németh ZH, Pacher P, Murray PJ, Kepka-Lenhart D, Morris SM Jr, Gause WC, Leibovich SJ, and Haskó G
- Subjects
- Adenosine pharmacology, Adenosine-5'-(N-ethylcarboxamide) pharmacology, Animals, Arginase metabolism, CCAAT-Enhancer-Binding Protein-beta metabolism, Cell Line, Cyclic AMP Response Element-Binding Protein metabolism, Extracellular Space metabolism, Inflammation immunology, Interleukin-13 metabolism, Interleukin-4 metabolism, Macrophages drug effects, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor, Adenosine A2A genetics, Receptor, Adenosine A2A immunology, Receptor, Adenosine A2B genetics, Receptor, Adenosine A2B immunology, STAT6 Transcription Factor metabolism, Tissue Inhibitor of Metalloproteinase-1 metabolism, Toll-Like Receptor 4 immunology, Toll-Like Receptor 4 metabolism, Vasodilator Agents pharmacology, Adenosine metabolism, Inflammation metabolism, Macrophages immunology, Macrophages metabolism, Receptor, Adenosine A2A metabolism, Receptor, Adenosine A2B metabolism
- Abstract
Adenosine has been implicated in suppressing the proinflammatory responses of classically activated macrophages induced by Th1 cytokines. Alternative macrophage activation is induced by the Th2 cytokines interleukin (IL)-4 and IL-13; however, the role of adenosine in governing alternative macrophage activation is unknown. We show here that adenosine treatment of IL-4- or IL-13-activated macrophages augments the expression of alternative macrophage markers arginase-1, tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), and macrophage galactose-type C-type lectin-1. The stimulatory effect of adenosine required primarily A(2B) receptors because the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased both arginase activity (EC(50)=261.8 nM) and TIMP-1 production (EC(50)=80.67 nM), and both pharmacologic and genetic blockade of A(2B) receptors prevented the effect of NECA. A(2A) receptors also contributed to the adenosine augmentation of IL-4-induced TIMP-1 release, as both adenosine and NECA were less efficacious in augmenting TIMP-1 release by A(2A) receptor-deficient than control macrophages. Of the transcription factors known to drive alternative macrophage activation, CCAAT-enhancer-binding protein β was required, while cAMP response element-binding protein and signal transducer and activator of transcription 6 were dispensable in mediating the effect of adenosine. We propose that adenosine receptor activation suppresses inflammation and promotes tissue restitution, in part, by promoting alternative macrophage activation.
- Published
- 2012
- Full Text
- View/download PDF
23. 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
24. From inflammation to wound healing: using a simple model to understand the functional versatility of murine macrophages.
- Author
-
Childs LM, Paskow M, Morris SM Jr, Hesse M, and Strogatz S
- Subjects
- Animals, Immunity, Innate, Inflammation immunology, Macrophage Activation, Mathematical Concepts, Mice, Models, Immunological, Wound Healing immunology, Macrophages immunology
- Abstract
Macrophages are fundamental cells of the innate immune system. Their activation is essential for such distinct immune functions as inflammation (pathogen-killing) and tissue repair (wound healing). An open question has been the functional stability of an individual macrophage cell: whether it can change its functional profile between different immune responses such as between the repair pathway and the inflammatory pathway. We studied this question theoretically by constructing a rate equation model for the key substrate, enzymes and products of the pathways; we then tested the model experimentally. Both our model and experiments show that individual macrophages can switch from the repair pathway to the inflammation pathway but that the reverse switch does not occur.
- Published
- 2011
- Full Text
- View/download PDF
25. 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
26. Regulation of macrophage arginase expression and tumor growth by the Ron receptor tyrosine kinase.
- Author
-
Sharda DR, Yu S, Ray M, Squadrito ML, De Palma M, Wynn TA, Morris SM Jr, and Hankey PA
- Subjects
- Animals, Arginase genetics, Arginase immunology, Cell Separation, Flow Cytometry, Gene Expression, Macrophage Activation immunology, Macrophages immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasms, Experimental immunology, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic, Receptor Protein-Tyrosine Kinases genetics, Receptor Protein-Tyrosine Kinases immunology, Signal Transduction genetics, Signal Transduction immunology, Transcription Factor AP-1 genetics, Transcription Factor AP-1 immunology, Arginase biosynthesis, Gene Expression Regulation immunology, Macrophages enzymology, Neoplasms, Experimental metabolism, Receptor Protein-Tyrosine Kinases metabolism, Transcription Factor AP-1 metabolism
- Abstract
M1 activation of macrophages promotes inflammation and immunity to intracellular pathogens, whereas M2 macrophage activation promotes resolution of inflammation, wound healing, and tumor growth. These divergent phenotypes are characterized, in part, by the expression of inducible NO synthase and arginase I (Arg1) in M1 versus M2 activated macrophages, respectively. In this study, we demonstrate that the Ron receptor tyrosine kinase tips the balance of macrophage activation by attenuating the M1 phenotype while promoting expression of Arg1 through a Stat6-independent mechanism. Induction of the Arg1 promoter by Ron is mediated by an AP-1 site located 433 bp upstream of the transcription start site. Treatment of primary macrophages with macrophage stimulating protein, the ligand for Ron, induces potent MAPK activation, upregulates Fos, and enhances binding of Fos to the AP-1 site in the Arg1 promoter. In vivo, Arg1 expression in tumor-associated macrophages (TAMs) from Ron(-/-) mice was significantly reduced compared with that in TAMs from control animals. Furthermore, we show that Ron is expressed specifically by Tie2-expressing macrophages, a TAM subset that exhibits a markedly skewed M2 and protumoral phenotype. Decreased Arg1 in TAMs from Ron(-/-) mice was associated with reduced syngeneic tumor growth in these animals. These findings indicate that Ron induces Arg1 expression in macrophages through a previously uncharacterized AP-1 site in the Arg1 promoter and that Ron could be therapeutically targeted in the tumor microenvironment to inhibit tumor growth by targeting expression of Arg1.
- Published
- 2011
- Full Text
- View/download PDF
27. 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
28. 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
29. 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
30. Arginine: master and commander in innate immune responses.
- Author
-
Morris SM Jr
- Subjects
- Arginase metabolism, Arginine metabolism, Models, Immunological, Mycobacterium metabolism, Nitric Oxide Synthase Type II metabolism, Arginine immunology, Immunity, Innate physiology, Macrophage Activation immunology, Nitric Oxide immunology, Signal Transduction immunology
- Abstract
The activation of macrophages through Toll-like receptor (TLR) signaling pathways is a major component of innate immune responses to infection. Because the production of nitric oxide (NO) from arginine by the inducible isoform of NO synthase (iNOS) in activated macrophages is essential for host defense against many pathogens, arginine availability is a critical determinant of resistance to infection. Thus, induction of the arginine catabolic enzyme arginase is exploited by some pathogens as a means of immune evasion. Details of this mechanism are revealed by studies that demonstrate that mycobacteria use a component of the TLR pathway to induce the type I isoform of arginase in macrophages through an autocrine-paracrine mechanism that involves macrophage-produced cytokines. Separate studies show that, in addition to inhibiting NO synthesis by substrate limitation, reducing the availability of arginine simply by nutrient deprivation can blunt the innate immune response by impairing a specific mitogen-activated protein kinase (MAPK) pathway downstream of TLR4. These findings illustrate the growing complexity of the roles of arginine as an enzyme substrate and also as a regulatory molecule in signal transduction pathways in immune cells.
- Published
- 2010
- Full Text
- View/download PDF
31. Effect of eculizumab on haemolysis-associated nitric oxide depletion, dyspnoea, and measures of pulmonary hypertension in patients with paroxysmal nocturnal haemoglobinuria.
- Author
-
Hill A, Rother RP, Wang X, Morris SM Jr, Quinn-Senger K, Kelly R, Richards SJ, Bessler M, Bell L, Hillmen P, and Gladwin MT
- Subjects
- Antibodies, Monoclonal, Humanized, Biomarkers blood, Blood Pressure drug effects, Dyspnea etiology, Female, Hemoglobins metabolism, Hemoglobinuria, Paroxysmal blood, Hemoglobinuria, Paroxysmal complications, Hemolysis physiology, Humans, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Male, Natriuretic Peptide, Brain blood, Peptide Fragments blood, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Dyspnea drug therapy, Hemoglobinuria, Paroxysmal drug therapy, Hemolysis drug effects, Hypertension, Pulmonary drug therapy, Nitric Oxide deficiency
- Abstract
Pulmonary hypertension (PH) is a common complication of haemolytic anaemia. Intravascular haemolysis leads to nitric oxide (NO) depletion, endothelial and smooth muscle dysregulation, and vasculopathy, characterized by progressive hypertension. PH has been reported in patients with paroxysmal nocturnal haemoglobinuria (PNH), a life-threatening haemolytic disease. We explored the relationship between haemolysis, systemic NO, arginine catabolism and measures of PH in 73 PNH patients enrolled in the placebo-controlled TRIUMPH (Transfusion Reduction Efficacy and Safety Clinical Investigation Using Eculizumab in Paroxysmal Nocturnal Haemoglobinuria) study. At baseline, intravascular haemolysis was associated with elevated NO consumption (P < 0.0001) and arginase-1 release (P < 0.0001). Almost half of the patients in the trial had elevated levels (> or =160 pg/ml) of N-terminal pro-brain natriuretic peptide (NT-proBNP), a marker of pulmonary vascular resistance and right ventricular dysfunction previously shown to indicate PH. Eculizumab treatment significantly reduced haemolysis (P < 0.001), NO depletion (P < 0.001), vasomotor tone (P < 0.05), dyspnoea (P = 0.006) and resulted in a 50% reduction in the proportion of patients with elevated NT-proBNP (P < 0.001) within 2 weeks of treatment. Importantly, the significant improvements in dyspnoea and NT-proBNP levels occurred without significant changes in anaemia. These data demonstrated that intravascular haemolysis in PNH produces a state of NO catabolism leading to signs of PH, including elevated NT pro-BNP and dyspnoea that are significantly improved by treatment with eculizumab.
- Published
- 2010
- Full Text
- View/download PDF
32. 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
33. 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
34. 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
35. Nitric oxide-mediated suppression of detrusor overactivity by arginase inhibitor in rats with chronic spinal cord injury.
- Author
-
Sasatomi K, Hiragata S, Miyazato M, Chancellor MB, Morris SM Jr, and Yoshimura N
- Subjects
- Animals, Arginase biosynthesis, Chronic Disease, Female, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Spinal Cord Injuries complications, Spine pathology, Urinary Bladder drug effects, Urinary Bladder, Neurogenic complications, Arginase antagonists & inhibitors, Gene Expression Regulation, Enzymologic, Nitric Oxide metabolism, Spinal Cord Injuries therapy, Urinary Bladder, Neurogenic drug therapy
- Abstract
Objectives: We investigated the effects of an arginase inhibitor on bladder overactivity and measured bladder arginase I and II mRNA levels in rats with chronic spinal cord injury (SCI)., Methods: We performed awake cystometrograms 3 to 4 weeks after spinal cord transection in female rats. Cystometric parameters such as mean amplitudes and number of non-voiding contractions (NVCs), voided volume, voiding efficiency, and micturition pressure were evaluated before and after intravenous (i.v.) injection of an arginase inhibitor (nor-NOHA: N(omega)-hydroxy-nor-L-arginine) in SCI rats. We also examined the effects of an NOS inhibitor (L-NAME: N(omega)-nitro-L-arginine methyl ester hydrochloride) to determine whether suppression of bladder overactivity by arginase inhibition is mediated by increased production of NO. In addition, we measured mRNA levels of arginase I and II in SCI bladders using quantitative real-time polymerase chain reaction (qRT-PCR)., Results: We found that nor-NOHA (10 mg/kg, i.v.) significantly decreased the amplitude and number of NVCs. There were no significant changes in other parameters before and after administration of vehicle or nor-NOHA at any dose. When we administered L-NAME (20 mg/kg, i.v.) before nor-NOHA injection (10 mg/kg, i.v.), nor-NOHA-induced inhibition of NVCs was prevented. The relative levels of both arginase I and II mRNA in the bladder were significantly higher in SCI rats compared with spinal cord-intact rats., Conclusions: These results suggest that arginase inhibition can suppress SCI-induced bladder overactivity as indicated by a reduction in NVCs. Thus, arginase inhibition could be an effective treatment for neurogenic bladder overactivity in pathological conditions such as SCI.
- Published
- 2008
- Full Text
- View/download PDF
36. 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
37. Cell- and isoform-specific increases in arginase expression in acute silica-induced pulmonary inflammation.
- Author
-
Poljakovic M, Porter DW, Millecchia L, Kepka-Lenhart D, Beighley C, Wolfarth MG, Castranova V, and Morris SM Jr
- Subjects
- Albumins metabolism, Animals, Arginase genetics, Bronchoalveolar Lavage Fluid chemistry, Bronchoalveolar Lavage Fluid cytology, Isoenzymes genetics, Isoenzymes metabolism, L-Lactate Dehydrogenase metabolism, Leukocyte Count, Lung metabolism, Macrophages, Alveolar drug effects, Macrophages, Alveolar metabolism, Male, Neutrophils drug effects, Neutrophils enzymology, Nitric Oxide Synthase Type II metabolism, Pneumonia chemically induced, Pneumonia metabolism, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Arginase metabolism, Lung drug effects, Silicon Dioxide toxicity
- Abstract
Arginase induction was reported in several inflammatory lung diseases, suggesting that this may be a common feature underlying the pathophysiology of such diseases. As little is known regarding arginase expression in silicosis, the induction and cellular localization of arginase were elucidated in lungs of Sprague-Dawley rats 24 h following exposure to varying doses of silica by intratracheal instillation. Arginase expression was evaluated by activity assay, quantification of arginase I and arginase II mRNA levels using real-time polymerase chain reaction (PCR), and immunohistochemistry. Analyses of cells and fluid obtained by bronchoalveolar lavage (BAL) showed that markers of pulmonary inflammation, tissue damage, activation of alveolar macrophages (AM) and NO production were significantly increased by all silica doses. Arginase activity was increased also in AMs isolated from BAL fluid of silica-treated rats. Silica produced two- and three-fold increases in arginase activity of whole lung at doses of 1 and 5 mg/100 g body weight, respectively. Levels of arginase I mRNA, but not of arginase II mRNA, were similarly elevated. In control lungs, arginase I immunoreactivity was observed only in AMs sparsely dispersed throughout the lung; no inducible nitric oxide synthase (iNOS) immunoreactivity was detected. In silica-treated lungs, arginase I and iNOS were co-expressed in most AMs that were abundantly clustered at inflammatory foci. The rapid induction of arginase I expression in inflammatory lung cells, similar to induction of arginase in other inflammatory lung diseases, implicates elevated arginase activity as a factor in the development of lung damage following exposure to silica.
- Published
- 2007
- Full Text
- View/download PDF
38. 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
39. 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
40. 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
41. 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
42. 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
43. Dysregulated arginine metabolism, hemolysis-associated pulmonary hypertension, and mortality in sickle cell disease.
- Author
-
Morris CR, Kato GJ, Poljakovic M, Wang X, Blackwelder WC, Sachdev V, Hazen SL, Vichinsky EP, Morris SM Jr, and Gladwin MT
- Subjects
- Adult, Aged, Amino Acids blood, Anemia, Sickle Cell blood, Anemia, Sickle Cell complications, Anemia, Sickle Cell mortality, Arginase blood, Case-Control Studies, Female, Humans, Male, Middle Aged, Nitric Oxide blood, Ornithine blood, Proportional Hazards Models, Anemia, Sickle Cell enzymology, Arginase metabolism, Arginine metabolism, Hypertension, Pulmonary etiology
- Abstract
Context: Sickle cell disease is characterized by a state of nitric oxide resistance and limited bioavailability of l-arginine, the substrate for nitric oxide synthesis. We hypothesized that increased arginase activity and dysregulated arginine metabolism contribute to endothelial dysfunction, pulmonary hypertension, and patient outcomes., Objective: To explore the role of arginase in sickle cell disease pathogenesis, pulmonary hypertension, and mortality., Design: Plasma amino acid levels, plasma and erythrocyte arginase activities, and pulmonary hypertension status as measured by Doppler echocardiogram were prospectively obtained in outpatients with sickle cell disease. Patients were followed up for survival up to 49 months., Setting: Urban tertiary care center and community clinics in the United States between February 2001 and March 2005., Participants: Two hundred twenty-eight patients with sickle cell disease, aged 18 to 74 years, and 36 control participants., Main Outcome Measures: Plasma amino acid levels, plasma and erythrocyte arginase activities, diagnosis of pulmonary hypertension, and mortality., Results: Plasma arginase activity was significantly elevated in patients with sickle cell disease, with highest activity found in patients with secondary pulmonary hypertension. Arginase activity correlated with the arginine-ornithine ratio, and lower ratios were associated with greater severity of pulmonary hypertension and with mortality in this population (risk ratio, 2.5; 95% confidence interval [CI], 1.2-5.2; P = .006). Global arginine bioavailability, characterized by the ratio of arginine to ornithine plus citrulline, was also strongly associated with mortality (risk ratio, 3.6; 95% CI, 1.5-8.3; P<.001). Increased plasma arginase activity was correlated with increased intravascular hemolytic rate and, to a lesser extent, with markers of inflammation and soluble adhesion molecule levels., Conclusions: These data support a novel mechanism of disease in which hemolysis contributes to reduced nitric oxide bioavailability and endothelial dysfunction via release of erythrocyte arginase, which limits arginine bioavailability, and release of erythrocyte hemoglobin, which scavenges nitric oxide. The ratios of arginine to ornithine and arginine to ornithine plus citrulline are independently associated with pulmonary hypertension and increased mortality in patients with sickle cell disease.
- Published
- 2005
- Full Text
- View/download PDF
44. 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
45. 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
46. 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
47. 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
48. Biochemical responses of healthy subjects during dietary supplementation with L-arginine.
- Author
-
Evans RW, Fernstrom JD, Thompson J, Morris SM Jr, and Kuller LH
- Subjects
- Arginine adverse effects, Arginine blood, Biological Availability, Blood drug effects, Blood Glucose analysis, Citrulline blood, Cyclic GMP blood, Dose-Response Relationship, Drug, Female, Glycine blood, Humans, Insulin blood, Lysine blood, Male, Ornithine blood, Arginine analogs & derivatives, Arginine pharmacology, Blood metabolism, Dietary Supplements
- Abstract
Dietary supplements of L-arginine, a substrate for nitric oxide synthases, may promote formation of nitric oxide and thus may be of clinical benefit. However, the optimal level of L-arginine supplementation is unclear. The objective of this study was to evaluate the response of healthy individuals to increasing doses of L-arginine (as free acid). Twelve healthy subjects were recruited and instructed to take L-arginine for 1-week periods at daily doses of 3, 9, 21, and 30 g. At baseline and at the end of each week, 24-hour urine and fasting blood samples were collected, and weight, diastolic blood pressure, and systolic blood pressure were recorded. Samples were analyzed for L-arginine, L-citrulline, glycine, L-lysine, L-ornithine, asymmetric dimethy L-arginine, symmetric dimethy L-arginine, glucose, insulin (serum), creatinine, cGMP (urine), and total nitrates (serum and urine). Ten subjects reported adverse side effects at initial L-arginine doses of 21 g/day (five subjects) or 30 g/day (five subjects), respectively. Blood pressure and weight did not change during the supplementation period. Of the individual biochemical measures, only L-arginine, glycine, and L-ornithine concentrations changed significantly. The mean concentration of L-arginine reached a peak during supplementation at 9 g/d; however, individuals differed markedly in their response. Availability of L-arginine, relative to that of asymmetric dimethy L-arginine, increased significantly at both 9 g/day and 21 g/day. Mean values indicate that supplementation with 9 g/day of L-arginine, a dose associated with minimal adverse side effects, is sufficient to increase circulating L-arginine concentrations. However, subjects varied widely in their responses, indicating that L-arginine supplementation needs to be tailored to individuals.
- Published
- 2004
- Full Text
- View/download PDF
49. Recent advances in arginine metabolism.
- Author
-
Morris SM Jr
- Subjects
- Agmatine metabolism, Arginase antagonists & inhibitors, Arginase metabolism, Humans, Respiratory Tract Diseases metabolism, Urea metabolism, Arginine metabolism
- Abstract
Purpose of Review: Arginine metabolism has been a topic of intense interest over the past 15-20 years, primarily with regard to the role of arginine as the nitrogen donor for nitric oxide synthesis. However, other important aspects of arginine metabolism, such as arginine transport and arginine catabolism via the arginases, arginine decarboxylase or agmatinase, have been less well studied. The purpose of this review is to highlight recent studies on the urea cycle, agmatine metabolism, and the arginases., Recent Findings: Recent advances include the cloning of complementary DNA encoding agmatinase, N-acetylglutamate synthetase, and proteins involved in mitochondrial arginine transport, as well as initial investigations of their regulation and tissue-specific expression. The most exciting results of studies in this area over the past year or so have indicated new roles for the arginases in health and disease, as a result of their effects on the synthesis of nitric oxide, proline, or polyamines, or on the expression of specific genes by their ability to limit the availability of free arginine., Summary: Recent studies have led to refinements in our understanding of the urea cycle. Agmatine metabolism is still largely a mystery, although the isolation of cloned cDNA for agmatinase and possibly also arginine decarboxylase should stimulate much needed investigations in this area. The most exciting findings in the field are coming from studies indicating new roles for the arginases in various diseases.
- Published
- 2004
- Full Text
- View/download PDF
50. Vertebrate agmatinases: what role do they play in agmatine catabolism?
- Author
-
Morris SM Jr
- Subjects
- Amino Acid Sequence, Animals, Arginase chemistry, Arginase genetics, Arginase metabolism, Humans, Molecular Sequence Data, Molecular Structure, Sequence Alignment, Agmatine metabolism, Ureohydrolases genetics, Ureohydrolases metabolism
- Abstract
Whereas agmatine in vertebrates may be derived from multiple sources such as the diet, endogenous synthesis via arginine decarboxylase, and possibly also from enteric bacteria, agmatinase is the only enzyme specific for agmatine catabolism. As it hydrolyzes a guanidino group within agmatine and also contains signature amino acid residues that act as ligand binding sites for the Mn(++) cofactor, agmatinase is classified as a member of the arginase superfamily. Very little information is available regarding how much agmatine in vertebrate species is catabolized by agmatinase versus other enzymes such as diamine and amine oxidases. Moreover, comparisons of primary sequences of several vertebrate agmatinases demonstrate that several residues essential for catalytic activity are not conserved in the mouse. This leads to the prediction that the agmatinase protein in mouse has little or no catalytic activity, not only raising questions about the physiologic routes of agmatine disposal in this organism, but also suggesting the existence of species-specific differences in mechanisms for regulating agmatine levels.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.