Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
1.
Bull Environ Contam Toxicol ; 110(1): 12, 2022 Dec 13.
Article En | MEDLINE | ID: mdl-36512146

The widespread occurrence of cyanobacteria blooms damages the water ecosystem and threatens the safety of potable water and human health. Exogenous L-lysine significantly inhibits the growth of a dominant cyanobacteria Microcystis aeruginosa in freshwater. However, the molecular mechanism of how lysine inhibits the growth of M. aeruginosa is unclear. In this study, both non-target and target metabolomic analysis were performed to investigate the effects of algicide L-lysine. The results showed that 8 mg L- 1 lysine most likely disrupts the metabolism of amino acids, especially the arginine and proline metabolism. According to targeted amino acid metabolomics analysis, only 3 amino acids (L-arginine, ornithine, and citrulline), which belong to the ornithine-ammonia cycle (OAC) in arginine metabolic pathway, showed elevated levels. The intracellular concentrations of ornithine, citrulline, and arginine increased by 115%, 124%, and 19.4%, respectively. These results indicate that L-lysine may affect arginine metabolism and OAC to inhibit the growth of M. aeruginosa.


Cyanobacteria , Herbicides , Microcystis , Humans , Microcystis/metabolism , Lysine/toxicity , Lysine/metabolism , Citrulline/metabolism , Ecosystem , Herbicides/metabolism , Cyanobacteria/metabolism , Ornithine/toxicity , Ornithine/metabolism , Arginine/chemistry , Arginine/metabolism , Ammonia , Microcystins/metabolism
2.
Biol Pharm Bull ; 42(9): 1581-1589, 2019.
Article En | MEDLINE | ID: mdl-31474718

As a bacterium used in industry for production of several amino acids, an endotoxin-free Corynebacterium (C.) glutamicum is well known. However, it is also true that the endotoxin-producing other Corynebacterium species is present. An aim of this study is to obtain a lactic acid bacterium (LAB) that produces ornithine and citrulline at high levels. We successfully isolated a strain, designated K-28, and identified it as Weissella (W.) confusa. The production of ornithine and citrulline by K-28 was 18 ± 1 and 10 ± 2 g/L, respectively, with a 100 ± 9% conversion rate when arginine was continuously fed into a jar fermenter. Although the ornithine high production using C. glutamicum is industrially present, the strains have been genetically modified. In that connection, the wild-type of C. glutamicum produces only 0.5 g/L ornithine, indicating that W. confusa K-28 is superior to C. glutamicum to use a probiotic microorganism. We confirmed that W. confusa K-28 harbors an arginine deiminase (ADI) gene cluster, wkaABDCR. The production of ornithine and the expression of these genes significantly decreased under the aerobic condition rather than anaerobic one. The expression level of the five genes did not differ with or without arginine, suggesting that the production of amino acids in the K-28 strain was not induced by exogenous arginine.


Citrulline/biosynthesis , Flowers/microbiology , Ornithine/biosynthesis , Senna Plant/microbiology , Weissella/metabolism , Animals , Citrulline/toxicity , Male , Ornithine/toxicity , Probiotics , Rats, Sprague-Dawley , Rats, Wistar , Toxicity Tests, Acute , Weissella/isolation & purification
3.
J Vis Exp ; (109)2016 Mar 17.
Article En | MEDLINE | ID: mdl-27023377

Stroke affecting white matter accounts for up to 25% of clinical stroke presentations, occurs silently at rates that may be 5-10 fold greater, and contributes significantly to the development of vascular dementia. Few models of focal white matter stroke exist and this lack of appropriate models has hampered understanding of the neurobiologic mechanisms involved in injury response and repair after this type of stroke. The main limitation of other subcortical stroke models is that they do not focally restrict the infarct to the white matter or have primarily been validated in non-murine species. This limits the ability to apply the wide variety of murine research tools to study the neurobiology of white matter stroke. Here we present a methodology for the reliable production of a focal stroke in murine white matter using a local injection of an irreversible eNOS inhibitor. We also present several variations on the general protocol including two unique stereotactic variations, retrograde neuronal tracing, as well as fresh tissue labeling and dissection that greatly expand the potential applications of this technique. These variations allow for multiple approaches to analyze the neurobiologic effects of this common and understudied form of stroke.


Axons/pathology , Disease Models, Animal , Enzyme Inhibitors/toxicity , Nerve Degeneration/pathology , Ornithine/analogs & derivatives , Stroke/pathology , White Matter/drug effects , Animals , Axons/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nitric Oxide Synthase Type III/antagonists & inhibitors , Ornithine/toxicity , Stroke/chemically induced , White Matter/pathology
4.
J Neurosci Methods ; 245: 44-57, 2015 Apr 30.
Article En | MEDLINE | ID: mdl-25745859

BACKGROUND: Ischemic stroke is the most frequent cause of persistent neurological disability in Western societies. New treatment strategies are required and effective in vivo models are crucial to their development. NEW METHOD: The current study establishes a novel in vivo rat model of focal striatal ischemia using the vasoconstrictive agent N5-(1-iminoethyl)-L-ornithine (L-NIO). Adult male Sprague Dawley rats received a unilateral intrastriatal infusion of L-NIO in combination with jugular vein occlusion. RESULTS: L-NIO infusion was associated with zero mortality, low surgical complexity and a reproducible infarct, providing advantages over established models of focal ischemia. The mean infarct volume of 8.5±5.3% of the volume of the contralateral striatum resulted in blood-brain barrier dysfunction, neuronal hypoxia and ongoing neurodegeneration. Further characteristics of ischemic stroke were exhibited, including robust microglia/macrophage and astroglial responses lasting at least 35 days post-ischemia, in addition to chronic motor function impairment. COMPARISON WITH EXISTING METHODS: When compared to other models such as the MCAo models, the consistency in regions affected, high success rate, zero mortality, reduced surgical complexity and minimal welfare requirements of the L-NIO model make it ideal for initial high-throughput investigations into preclinical efficacy and proof of principle studies of acute ischemic stroke interventions. CONCLUSION: We propose that the L-NIO rat model of focal striatal ischemia does not replace the use of other ischemic stroke models. Rather it provides a new, complementary tool for initial preclinical investigations into the treatment of ischemic stroke.


Brain Ischemia/etiology , Disease Models, Animal , Enzyme Inhibitors/toxicity , Ornithine/analogs & derivatives , Animals , Blood-Brain Barrier/physiopathology , Brain Infarction/etiology , Brain Ischemia/complications , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dizocilpine Maleate/therapeutic use , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Jugular Veins/physiopathology , Male , Movement Disorders/etiology , Movement Disorders/prevention & control , Nerve Degeneration/etiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Ornithine/toxicity , Rats , Rats, Sprague-Dawley , Time Factors
5.
J Neurochem ; 130(4): 555-62, 2014 Aug.
Article En | MEDLINE | ID: mdl-24762056

Neonatal hypoxic ischaemic (HI) injury frequently causes neural impairment in surviving infants. Our knowledge of the underlying molecular mechanisms is still limited. Protein deimination is a post-translational modification caused by Ca(+2) -regulated peptidylarginine deiminases (PADs), a group of five isozymes that display tissue-specific expression and different preference for target proteins. Protein deimination results in altered protein conformation and function of target proteins, and is associated with neurodegenerative diseases, gene regulation and autoimmunity. In this study, we used the neonatal HI and HI/infection [lipopolysaccharide (LPS) stimulation] murine models to investigate changes in protein deimination. Brains showed increases in deiminated proteins, cell death, activated microglia and neuronal loss in affected brain areas at 48 h after hypoxic ischaemic insult. Upon treatment with the pan-PAD inhibitor Cl-amidine, a significant reduction was seen in microglial activation, cell death and infarct size compared with control saline or LPS-treated animals. Deimination of histone 3, a target protein of the PAD4 isozyme, was increased in hippocampus and cortex specifically upon LPS stimulation and markedly reduced following Cl-amidine treatment. Here, we demonstrate a novel role for PAD enzymes in neural impairment in neonatal HI Encephalopathy, highlighting their role as promising new candidates for drug-directed intervention in neurotrauma. Hypoxic Ischaemic Insult (HI) results in activation of peptidylarginine deiminases (PADs) because of calcium dysregulation. Target proteins undergo irreversible changes of protein bound arginine to citrulline, resulting in protein misfolding. Infection in synergy with HI causes up-regulation of TNFα, nuclear translocation of PAD4 and change in gene regulation as a result of histone deimination. Pharmacological PAD inhibition significantly reduced HI brain damage.


Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Animals , Animals, Newborn , Brain Infarction/drug therapy , Brain Infarction/pathology , Cell Death/drug effects , Central Nervous System Bacterial Infections/drug therapy , Central Nervous System Bacterial Infections/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neurons/drug effects , Neurons/pathology , Ornithine/analogs & derivatives , Ornithine/toxicity , Protein-Arginine Deiminases
6.
Regul Toxicol Pharmacol ; 67(3): 360-71, 2013 Dec.
Article En | MEDLINE | ID: mdl-23994624

L-Ornithine monohydrochloride was evaluated in two in vitro genotoxicity assays and a rat 90-day oral toxicity study. No evidence of genotoxicity was observed in the reverse bacterial mutation assay or the chromosome aberration test at doses of up to 5000 µg/plate or 1686 µg/mL, respectively, both in the presence and absence of metabolic activation. Rats were administered L-ornithine monohydrochloride at dietary concentrations of 0 (basal diet), 1.25%, 2.5%, or 5.0% for 90 days. No changes in body weight, food consumption, ophthalmoscopy, or hematology were observed. Transient increases in water intake and urinary volume, and a decrease in specific gravity were observed in males receiving 5.0% L-ornithine monohydrochloride; however, these were likely attributable to the central role of ornithine in the urea cycle and the consequent increase in urea production. A decrease in serum chloride concentration and an increase in urinary chloride excretion were observed; however, these were likely attributable to administration of the hydrochloride salt of ornithine and were not considered to be of any toxicological significance. No remarkable findings were noted at necropsy. Based on the results of the study, a no-observed-adverse effect level (NOAEL) of 3445 and 3986 mg/kg body weight/day was established for male and female rats.


Ornithine/toxicity , Administration, Oral , Animals , Cell Line , Chromosome Aberrations/chemically induced , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Male , Mutagenicity Tests , No-Observed-Adverse-Effect Level , Organ Specificity , Ornithine/administration & dosage , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Toxicity Tests, Subchronic
7.
Metab Brain Dis ; 27(4): 521-30, 2012 Dec.
Article En | MEDLINE | ID: mdl-22798168

Ornithine, ammonia and homocitrulline are the major metabolites accumulating in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome, a genetic disorder characterized by neurological regression whose pathogenesis is still not understood. The present work investigated the in vivo effects of intracerebroventricular administration of ornithine and homocitrulline in the presence or absence of hyperammonemia induced by intraperitoneal urease treatment on a large spectrum of oxidative stress parameters in cerebral cortex from young rats in order to better understand the role of these metabolites on brain damage. Ornithine increased thiobarbituric acid-reactive substances (TBA-RS) levels and carbonyl formation and decreased total antioxidant status (TAS) levels. We also observed that the combination of hyperammonemia with ornithine resulted in significant decreases of sulfhydryl levels, reduced glutathione (GSH) concentrations and the activities of catalase (CAT) and glutathione peroxidase (GPx), highlighting a synergistic effect of ornithine and ammonia. Furthermore, homocitrulline caused increases of TBA-RS values and carbonyl formation, as well as decreases of GSH concentrations and GPx activity. Hcit with hyperammonemia (urease treatment) decreased TAS and CAT activity. We also showed that urease treatment per se was able to enhance TBA-RS levels. Finally, nitric oxide production was not altered by Orn and Hcit alone or in combination with hyperammonemia. Our data indicate that the major metabolites accumulating in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome provoke lipid and protein oxidative damage and a reduction of the antioxidant defenses in the brain. Therefore, it is presumed that oxidative stress may represent a relevant pathomechanism involved in the brain damage found in patients affected by this disease.


Amino Acid Metabolism, Inborn Errors/metabolism , Ammonia/metabolism , Brain/metabolism , Citrulline/analogs & derivatives , Homeostasis/physiology , Ornithine/metabolism , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/chemically induced , Ammonia/blood , Animals , Antioxidants/metabolism , Catalase/metabolism , Citrulline/metabolism , Citrulline/urine , Glutathione Peroxidase/metabolism , Injections, Intraperitoneal , Injections, Intraventricular , Male , Nitric Oxide/metabolism , Ornithine/blood , Ornithine/toxicity , Oxidation-Reduction , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Sulfhydryl Compounds/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Urease
8.
Metab Brain Dis ; 27(4): 479-86, 2012 Dec.
Article En | MEDLINE | ID: mdl-22699997

Hyperornithinemia is the biochemical hallmark of hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome, an inherited metabolic disease clinically characterized by mental retardation whose pathogenesis is still poorly known. In the present work, we produced a chemical animal model of hyperornithinemia induced by a subcutaneous injection of saline-buffered Orn (2-5 µmol/g body weight) to rats. High brain Orn concentrations were achieved, indicating that Orn is permeable to the blood brain barrier. We then investigated the effect of early chronic postnatal administration of Orn on physical development and on the performance of adult rats in the open field, the Morris water maze and in the step down inhibitory avoidance tasks. Chronic Orn treatment had no effect on the appearance of coat, eye opening or upper incisor eruption, nor on the free-fall righting reflex and on the adult rat performance in the Morris water maze and in the inhibitory avoidance tasks, suggesting that physical development, aversive and spatial localization were not changed by Orn. However, Orn-treated rats did not habituate to the open field apparatus, implying a deficit of learning/memory. Motor activity was the same for Orn- and saline- injected animals. We also verified that Orn subcutaneous injections provoked lipid peroxidation in the brain, as determined by a significant increase of thiobarbituric acid-reactive substances levels. Our results indicate that chronic early postnatal hyperornithinemia may impair the central nervous system, causing minor disabilities which result in specific learning deficiencies.


Amino Acid Metabolism, Inborn Errors/chemically induced , Learning Disabilities/chemically induced , Learning Disabilities/psychology , Ornithine/toxicity , Amino Acid Metabolism, Inborn Errors/psychology , Ammonia/blood , Animals , Animals, Newborn , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Citrulline/analogs & derivatives , Citrulline/blood , Cognition/drug effects , Cognition/physiology , Developmental Disabilities/chemically induced , Disease Models, Animal , Half-Life , Maze Learning/drug effects , Memory/drug effects , Memory, Long-Term/drug effects , Ornithine/pharmacokinetics , Postural Balance/drug effects , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
9.
Int J Pharm ; 423(2): 392-400, 2012 Feb 28.
Article En | MEDLINE | ID: mdl-22210000

The development of new nonviral vectors characterized by high transfection efficiency and low cytotoxicity remains an important challenge in the field of gene delivery. Unsymmetrical bolaamphiphiles (bolas) appear as new emerging candidates for this application. In this work, new unsymmetrical bolas, bearing neutral lactonic acid and cationic ornithine residues at the two ends of a hydrophobic spacer, were synthesized and their properties were compared to analogues bearing a gluconic acid residue. The new bolas showed DNA binding and condensation at higher N/P ratios than their gluconic analogues, probably due to their larger neutral head group. Whereas the size of the complexes of the new bolas with DNA (bolaplexes) increased with N/P, as a result of charge neutralization, their formulations with DOPE at high N/P were of small size (ca. 200 nm). These DOPE formulations showed high transfection efficiency in different cell lines (HeLa, COS-7 and HepG2), close to that of jetPEI. Their cytotoxicity was relatively low, which allowed repetitive transfection in vitro. Fluorescence imaging showed that the bolaplexes bind rapidly to cell surface and internalize mainly through endocytosis. This work suggests a new type of efficient nonviral vectors based on bolaamphiphiles.


DNA/metabolism , Furans/chemistry , Lactose/chemistry , Ornithine/chemistry , Pyridones/chemistry , Surface-Active Agents/chemistry , Transfection/methods , Animals , COS Cells , Chlorocebus aethiops , DNA/chemistry , Endocytosis , Furans/toxicity , Gluconates/chemistry , HeLa Cells , Hep G2 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Lactose/analogs & derivatives , Lactose/toxicity , Microscopy, Fluorescence , Nanotechnology , Ornithine/analogs & derivatives , Ornithine/toxicity , Particle Size , Phosphatidylethanolamines/chemistry , Pyridones/toxicity , Surface-Active Agents/toxicity , Time Factors
10.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G929-38, 2011 Jun.
Article En | MEDLINE | ID: mdl-21415415

Inflammatory bowel diseases (IBDs), mainly Crohn's disease and ulcerative colitis, are dynamic, chronic inflammatory conditions that are associated with an increased colon cancer risk. Inflammatory cell apoptosis is a key mechanism for regulating IBD. Peptidylarginine deiminases (PADs) catalyze the posttranslational conversion of peptidylarginine to peptidylcitrulline in a calcium-dependent, irreversible reaction and mediate the effects of proinflammatory cytokines. Because PAD levels are elevated in mouse and human colitis, we hypothesized that a novel small-molecule inhibitor of the PADs, i.e., chloramidine (Cl-amidine), could suppress colitis in a dextran sulfate sodium mouse model. Results are consistent with this hypothesis, as demonstrated by the finding that Cl-amidine treatment, both prophylactic and after the onset of disease, reduced the clinical signs and symptoms of colitis, without any indication of toxic side effects. Interestingly, Cl-amidine drives apoptosis of inflammatory cells in vitro and in vivo, providing a mechanism by which Cl-amidine suppresses colitis. In total, these data help validate the PADs as therapeutic targets for the treatment of IBD and further suggest Cl-amidine as a candidate therapy for this disease.


Anti-Inflammatory Agents/pharmacology , Colitis/prevention & control , Colon/drug effects , Enzyme Inhibitors/pharmacology , Gastrointestinal Agents/pharmacology , Hydrolases/antagonists & inhibitors , Ornithine/analogs & derivatives , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/toxicity , Apoptosis/drug effects , Arginine/metabolism , Citrulline/metabolism , Colitis/chemically induced , Colitis/enzymology , Colitis/pathology , Colon/enzymology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/toxicity , HT29 Cells , Humans , Hydrolases/metabolism , Mice , Mice, Inbred C57BL , Ornithine/administration & dosage , Ornithine/pharmacology , Ornithine/toxicity , Protein Processing, Post-Translational/drug effects , Protein-Arginine Deiminases , Up-Regulation
12.
Brain Res ; 1369: 235-44, 2011 Jan 19.
Article En | MEDLINE | ID: mdl-21059345

Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is an autosomal recessive disorder caused by a defect in the mitochondrial ornithine transporter, leading to accumulation of ornithine (Orn), homocitrulline (Hcit) and ammonia. Progressive neurological regression whose pathogenesis is not well established is common in this disease. The present work investigated the in vivo effects of intracerebroventricular administration of Orn and Hcit on important parameters of oxidative stress and energy metabolism in cerebral cortex from young rats. Orn and Hcit significantly increased thiobarbituric acid-reactive substances values and carbonyl formation, indicators of lipid and protein oxidative damage, respectively. Furthermore, N-acetylcysteine and the combination of the free radical scavengers ascorbic acid plus α-tocopherol attenuated the lipid oxidation and totally prevented the protein oxidative damage provoked by Orn and Hcit, suggesting that reactive species were involved in these effects. Hcit, but not Orn administration, also decreased glutathione concentrations, as well as the activity of catalase and glutathione peroxidase, indicating that Hcit provokes a reduction of brain antioxidant defenses. As regards to the parameters of energy metabolism, we verified that Orn and Hcit significantly inhibited the citric acid cycle function (inhibition of CO(2) synthesis from [1-(14)C] acetate), the aerobic glycolytic pathway (reduced CO(2) production from [U-(14)C] glucose) and complex I-III activity of the respiratory chain. Hcit also inhibited the activity of aconitase, an enzyme very susceptible to free radical attack. Taken together, our data indicate that mitochondrial homeostasis is disturbed by Orn and especially by Hcit. It is presumed that the impairment of brain bioenergetics and the oxidative damage induced by these metabolites may possibly contribute to the brain deterioration and neurological symptoms affecting patients with HHH syndrome.


Cerebral Cortex/metabolism , Citrulline/analogs & derivatives , Ornithine/toxicity , Oxidative Stress/drug effects , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Citrulline/administration & dosage , Citrulline/metabolism , Citrulline/toxicity , Hyperammonemia/metabolism , Hyperammonemia/physiopathology , Injections, Intraventricular , Lipid Peroxidation/drug effects , Ornithine/administration & dosage , Ornithine/deficiency , Ornithine/metabolism , Rats , Rats, Wistar , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/physiopathology
13.
Pancreas ; 39(7): 1047-56, 2010 Oct.
Article En | MEDLINE | ID: mdl-20531247

OBJECTIVES: l-Ornithine is a precursor of polyamine synthesis that is essential for cell survival. In contrast, intraperitoneal (IP) administration of a large dose of l-ornithine results in death of pancreatic acinar cells in rats. We investigated changes in pancreatic and extrapancreatic polyamine homeostasis after injection of l-ornithine and tested the effects of the stable polyamine analogue methylspermidine (MeSpd) on l-ornithine-induced pancreatitis. METHODS: Male Wistar rats were injected IP with 3 g/kg l-ornithine and were untreated, pretreated, or treated with 50 mg/kg MeSpd IP. Rats were killed after 0 to 168 hours for determinations of polyamines and activities of ornithine decarboxylase and spermidine/spermine N(1)-acetyltransferase (SSAT). Pancreatitis severity was assessed by measuring standard laboratory and histological parameters. RESULTS: Injection of l-ornithine paradoxically induced pancreatic spermidine catabolism, possibly via activation of SSAT, after (>6 hours) appearance of the first histological signs of acute pancreatitis. Polyamine levels generally increased in the lung and liver with the exception of lung spermidine levels, which decreased. Methylspermidine did not influence polyamine levels and SSAT activity and did not ameliorate the severity of l-ornithine-induced pancreatitis. CONCLUSIONS: l-Ornithine-induced pancreatitis was associated with activation of pancreatic polyamine catabolism. However, administration of a metabolically stable polyamine analogue did not affect disease severity.


Biogenic Polyamines/metabolism , Homeostasis , Ornithine/toxicity , Pancreatitis/metabolism , Acetyltransferases/metabolism , Acute Disease , Animals , Creatinine/blood , HSP72 Heat-Shock Proteins/analysis , I-kappa B Proteins/analysis , Interleukin-1beta/biosynthesis , Liver/metabolism , Lung/metabolism , Male , NF-KappaB Inhibitor alpha , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/pathology , Rats , Rats, Wistar
14.
Crit Care Med ; 36(7): 2117-27, 2008 Jul.
Article En | MEDLINE | ID: mdl-18594222

OBJECTIVE: Intraperitoneal administration of large doses of L-arginine is known to induce severe acute pancreatitis in rats. We therefore set out to determine whether metabolites of L-arginine (L-ornithine, L-citrulline, and nitric oxide) cause pancreatitis. DESIGN: The authors conducted an in vivo animal study. SETTING: This study was conducted at a university research laboratory. SUBJECTS: Study subjects were male Wistar rats. INTERVENTIONS: Dose-response and time course changes of laboratory and histologic parameters of pancreatitis were determined after L-arginine, L-ornithine, L-citrulline, or sodium nitroprusside (nitric oxide donor) injection. MEASUREMENTS AND MAIN RESULTS: Intraperitoneal injection of 3 g/kg L-ornithine but not L-citrulline or nitroprusside caused severe acute pancreatitis; 4 to 6 g/kg L-ornithine killed the animals within hours. Serum and ascitic amylase activities were significantly increased, whereas pancreatic amylase activity was decreased after intraperitoneal injection of 3 g/kg L-ornithine. The increase in pancreatic trypsin activity (9-48 hrs) correlated with the degradation of IkappaB proteins and elevated interleukin-1beta levels. Oxidative stress in the pancreas was evident from 6 hrs; HSP72 synthesis was increased from 4 hrs after L-ornithine administration. Morphologic examination of the pancreas showed massive interstitial edema, apoptosis, and necrosis of acinar cells and infiltration of neutrophil granulocytes and monocytes 18 to 36 hrs after 3 g/kg L-ornithine injection. One month after L-ornithine injection, the pancreas appeared almost normal; the destructed parenchyma was partly replaced by fat. Equimolar administration of L-arginine resulted in lower pancreatic weight/body weight ratio, pancreatic myeloperoxidase activity, and histologic damage compared with the L-ornithine-treated group. L-ornithine levels in the blood were increased 54-fold after intraperitoneal administration of L-arginine. CONCLUSIONS: We have developed a simple, noninvasive model of acute necrotizing pancreatitis in rats by intraperitoneal injection of 3 g/kg L-ornithine. Interestingly, we found that, compared with L-arginine, L-ornithine was even more effective at inducing pancreatitis. Large doses of L-arginine produce a toxic effect on the pancreas, at least in part, through L-ornithine.


Ornithine/toxicity , Pancreatitis, Acute Necrotizing/chemically induced , Animals , Apoptosis/drug effects , Arginine/blood , Arginine/toxicity , Citrulline/blood , Citrulline/toxicity , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Ornithine/administration & dosage , Ornithine/blood , Pancreatic alpha-Amylases , Pancreatitis, Acute Necrotizing/metabolism , Pancreatitis, Acute Necrotizing/pathology , Peroxidase/metabolism , Rats , Rats, Wistar , Time Factors , Trypsin/metabolism , alpha-Amylases/metabolism
16.
Am J Physiol Cell Physiol ; 293(2): C729-37, 2007 Aug.
Article En | MEDLINE | ID: mdl-17494634

We previously showed that ornithine was mainly transported via cationic amino acid transporter (CAT)-1 in human retinal pigment epithelial (RPE) cell line, human telomerase RT (hTERT)-RPE, and that CAT-1 was involved in ornithine cytotoxicity in ornithine-delta-aminotransferase (OAT)-deficient cell produced by a OAT specific inhibitor, 5-fluoromethylornithine (5-FMO). We showed here that CAT-1 mRNA expression was increased by ornithne in OAT-deficient RPE cells, which was reversed by an inhibitor of ornithine decarboxylase (ODC), alpha-difluoromethylornithine (DFMO). Polyamines, especially spermine, one of the metabolites of ODC, also enhanced the expression of CAT-1 mRNA. ODC mRNA expression was also increased by ornithine and polyamines, and gene silencing of ODC by siRNA decreased ornithine transport activity and its cytotoxicity. In addition, the mRNA of nuclear protein c-myc was also increased in 5-FMO- and ornithine-treated hTERT-RPE cells, and gene silencing of c-myc prevented the induction of CAT-1 and ODC. Increases in expression of CAT-1, ODC, and c-myc, and the inhibition of these stimulated expression by DFMO were also observed in primary porcine RPE cells. These results suggest that spermine plays an important role in stimulation of mRNA expression of CAT-1, which is a crucial role in ornithine cytotoxicity in OAT-deficient hTERT-RPE cells.


Cationic Amino Acid Transporter 1/biosynthesis , Epithelial Cells/metabolism , Ornithine/metabolism , Pigment Epithelium of Eye/metabolism , RNA, Messenger/biosynthesis , Animals , Cationic Amino Acid Transporter 1/genetics , Cell Line , Dose-Response Relationship, Drug , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Humans , Large Neutral Amino Acid-Transporter 1/biosynthesis , Large Neutral Amino Acid-Transporter 1/genetics , Ornithine/analogs & derivatives , Ornithine/pharmacology , Ornithine/toxicity , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Ornithine-Oxo-Acid Transaminase/metabolism , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/drug effects , Pigment Epithelium of Eye/enzymology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Putrescine/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Spermidine/metabolism , Spermine/metabolism , Swine , Telomerase/genetics , Telomerase/metabolism , Time Factors , Transfection , Up-Regulation
17.
Invest Ophthalmol Vis Sci ; 48(1): 455-63, 2007 Jan.
Article En | MEDLINE | ID: mdl-17197567

PURPOSE: A prior study showed inactivation of ornithine-delta-aminotransferase (OAT)-deficient human retinal pigment epithelial (RPE) cells by a specific irreversible inhibitor (5-fluoromethylornithine; 5-FMO) leading to cell death, in an in vitro model of gyrate atrophy (GA) of the choroid and retina. In the present study, the cytotoxicity of metabolites of ornithine, especially spermine, in RPE cells was investigated, to clarify the mechanism of ornithine cytotoxicity in RPE cells. METHODS: RPE cells were incubated with ornithine or compounds involved in ornithine metabolic pathways. The effects on RPE cell viability and proliferative activity were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric and [(3)H]thymidine incorporation assays. Incorporation of spermine into RPE cells was examined by using [(14)C]spermine and dansyl-spermine. To assess spermine-induced RPE cell death, cells were double stained with annexin V and propidium iodide and subjected to flow cytometry. RESULTS: Ornithine, arginine, glutamate, proline, creatine, glycine, and putrescine exhibited no effects on the viability and proliferative activities of RPE cells, whereas spermidine and spermine (10 mM) inhibited [(3)H]thymidine incorporation by 13% and 89%, respectively. The inhibition of [(3)H]thymidine incorporation by spermine was dose dependent and was observed as early as 4 hours after addition. Further, spermine was incorporated and accumulated in the perinuclear region of RPE cells. Apoptotic RPE cell death was induced by spermine in a dose-dependent manner. CONCLUSIONS: The present results demonstrated that excessive spermine is cytotoxic to RPE cells and suggest that metabolites of ornithine, especially spermine, may be involved in the mechanism of RPE degeneration in GA.


Apoptosis/drug effects , Pigment Epithelium of Eye/drug effects , Spermine/toxicity , Animals , Annexin A5/metabolism , Cattle , Cell Culture Techniques , Cell Proliferation/drug effects , Colorimetry , DNA/biosynthesis , Dose-Response Relationship, Drug , Flow Cytometry , Microscopy, Confocal , Ornithine/toxicity , Pigment Epithelium of Eye/pathology , Propidium/metabolism , Tetrazolium Salts , Thiazoles , Thymidine/metabolism
18.
Invest Ophthalmol Vis Sci ; 48(1): 464-71, 2007 Jan.
Article En | MEDLINE | ID: mdl-17197568

PURPOSE: A prior report showed ornithine cytotoxicity in ornithine-delta-aminotransferase (OAT)-deficient human retinal pigment epithelial (RPE) cells in an in vitro model of gyrate atrophy of the choroid and retina. This study was intended to clarify the mechanism of ornithine cytotoxicity and to determine the responsible amino acid transporters. METHODS: The mRNA expression of amino acid transporters in human telomerase reverse transcriptase (hTERT)-RPE cells was examined by reverse transcription polymerase chain reaction (RT-PCR) and Northern blot analysis. Carrier-mediated ornithine transport via the L-type amino acid transporter (LAT)1, LAT2, cationic amino acid transporter (CAT)-1, and y(+)LAT2 systems was evaluated by short interfering (si)RNA-mediated gene silencing. The cytoprotective effect of CAT-1-specific siRNA on ornithine cytotoxicity was measured using quantitative analysis of cellular adenosine triphosphate (ATP) at 24 hours after treatment with ornithine in OAT-deficient RPE cells. RESULTS: LAT1, LAT2, CAT-1, and y(+)LAT2 mRNA expression was detected by Northern blot analysis, whereas RT-PCR revealed that LAT1, LAT2, y(+)LAT1, y(+)LAT2, CAT-1, and b(0,+)AT mRNAs were expressed together with the heterodimeric glycoproteins 4F2hc and rBAT in hTERT-RPE cells. l-[(14)C]ornithine uptake in hTERT-RPE cells was decreased by 46.6% and 22.0% by CAT-1 and y(+)LAT2 siRNA, respectively, whereas LAT1 and LAT2 siRNA had no significant effect. Further, CAT-1 silencing by siRNA reduced ornithine cytotoxicity in OAT-deficient RPE cells. CONCLUSIONS: The results suggest that ornithine transport via CAT-1 may play a crucial role in ornithine cytotoxicity in hTERT-RPE cells. Reduction of the ornithine transport via CAT-1 may be a new target for treatment of gyrate atrophy.


Cationic Amino Acid Transporter 1/metabolism , Ornithine/metabolism , Ornithine/toxicity , Pigment Epithelium of Eye/metabolism , Adenosine Triphosphate/metabolism , Biological Transport , Blotting, Northern , Cationic Amino Acid Transporter 1/genetics , Cells, Cultured , Gene Silencing , Humans , Leucine/metabolism , Ornithine-Oxo-Acid Transaminase/metabolism , Pigment Epithelium of Eye/drug effects , Plasmids , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Skin Pharmacol Physiol ; 17(6): 283-8, 2004.
Article En | MEDLINE | ID: mdl-15528958

The complex metabolic function of L-ornithine has led to a great interest in studying its pharmacotherapeutic potential. L-Ornithine is known to be crucial for the metabolism of keratinocytes, especially in the synthesis of urea, polyamines and precursors of collagen synthesis. In this study, we investigated in vitro the cytotoxicity of L-ornithine, and its influence on urea synthesis and arginase expression in primary human keratinocytes. L-Ornithine (> or =1 mM) induced a decrease in the de novo urea synthesis of keratinocytes and an increase (> or =10 mM) in the expression of the urea-generating enzyme arginase. Up to 20 mM, L-ornithine showed no cytotoxic potential, whereas higher concentrations induced apoptosis in keratinocytes in a concentration- and time-dependent manner.


Keratinocytes/drug effects , Ornithine/metabolism , Skin/metabolism , Urea/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Keratinocytes/metabolism , Ornithine/toxicity , Skin/drug effects
20.
Invest Ophthalmol Vis Sci ; 44(11): 5023-8, 2003 Nov.
Article En | MEDLINE | ID: mdl-14578430

PURPOSE: To investigate the effect of amino acids on ornithine cytotoxicity in ornithine-delta-aminotransferase (OAT)-deficient human retinal pigment epithelial (RPE) cells as an in vitro model of gyrate atrophy (GA) of the choroid and retina. METHODS: RPE cells were treated with 0.5 mM 5-fluoromethylornithine (5-FMOrn), a specific and irreversible OAT inhibitor. OAT-deficient RPE cells were incubated with 10 mM ornithine in the presence of 20 mM of 1 of 18 amino acids or 10 mM 2-amino-2-norbornane-carboxylic acid (BCH), a conventional inhibitor of the amino acid transporter system L. Ornithine cytotoxicity and cytoprotective effects of each amino acid was evaluated with a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay 72 hours after treatment with ornithine in OAT-deficient RPE cells. Ornithine incorporation into RPE cells was evaluated using DL-[14C]ornithine. RESULTS: An MTT colorimetric assay revealed that small and large zwitterionic amino acids, but not acidic or basic amino acids, decreased ornithine cytotoxicity in OAT-deficient RPE cells. Incorporation of DL-[14C]ornithine by RPE cells decreased to 79% of the control level after incubation for 48 hours with 20 mM leucine, the most effective cytoprotective amino acid. Further, BCH prevented ornithine cytotoxicity in a dose-dependent manner. Both light and heavy chains of L-type amino acid transporter (LAT)-1, LAT2, y+LAT1, and 4F2hc were expressed in RPE cells. CONCLUSIONS: The present results demonstrate that L-type amino acid transporter(s) may be involved in protection against ornithine cytotoxicity in human RPE cells. Thus, amino acid transportation in RPE cells may be a good target for a new therapy for GA as well as other kinds of chorioretinal degeneration.


Amino Acid Transport System y+ , Amino Acids/pharmacology , Cytoprotection/drug effects , Ornithine/analogs & derivatives , Ornithine/toxicity , Pigment Epithelium of Eye/drug effects , Amino Acids, Cyclic/pharmacology , Cell Survival , Cells, Cultured , Colorimetry , Fusion Regulatory Protein 1, Heavy Chain/drug effects , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Light Chains/drug effects , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Gyrate Atrophy/drug therapy , Gyrate Atrophy/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/drug effects , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Ornithine-Oxo-Acid Transaminase/deficiency , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles
...