Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Chem Res Toxicol ; 34(10): 2194-2201, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34609854

ABSTRACT

Monoamine oxidase (MAO) is rapidly gaining appreciation for its pathophysiologic role in cardiac injury and failure. Oxidative deamination of norepinephrine by MAO generates H2O2 and the catecholaldehyde 3,4-dihydroxyphenylglycolaldehyde (DOPEGAL), the latter of which is a highly potent and reactive electrophile that has been linked to cardiotoxicity. However, many questions remain as to whether catecholaldehydes regulate basic physiological processes in the myocardium and the pathways involved. Here, we examined the role of MAO-derived oxidative metabolites in mediating the activation of cardiac fibroblasts in response to norepinephrine. In neonatal murine cardiac fibroblasts, norepinephrine increased reactive oxygen species (ROS), accumulation of catechol-modified protein adducts, expression and secretion of collagens I/III, and other markers of profibrotic activation including STAT3 phosphorylation. These effects were attenuated with MAO inhibitors, the aldehyde-scavenging dipeptide l-carnosine, and FPS-ZM1, an antagonist for the receptor for advanced glycation endproducts (RAGE). Interestingly, treatment of cardiac fibroblasts with a low dose (1 µM) of DOPEGAL-modified albumin phenocopied many of the effects of norepinephrine and also induced an increase in RAGE expression. Higher doses (>10 µM) of DOPEGAL-modified albumin were determined to be toxic to cardiac fibroblasts in a RAGE-dependent manner, which was mitigated by l-carnosine. Collectively, these findings suggest that norepinephrine may influence extracellular matrix remodeling via an adrenergic-independent redox pathway in cardiac fibroblasts involving the MAO-mediated generation of ROS, catecholaldehydes, and RAGE. Furthermore, since elevations in the catecholaminergic tone and oxidative stress in heart disease are linked with cardiac fibrosis, this study illustrates novel drug targets that could potentially mitigate this serious disorder.


Subject(s)
Myofibroblasts/drug effects , Norepinephrine/pharmacology , Norepinephrine/toxicity , Receptor for Advanced Glycation End Products/metabolism , Animals , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Myofibroblasts/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/antagonists & inhibitors
2.
J Cell Mol Med ; 25(15): 7190-7203, 2021 08.
Article in English | MEDLINE | ID: mdl-34169637

ABSTRACT

Curcumin is a natural polyphenol extracted from the rhizome of Curcuma that has an important antitumour effect, but its effect on adverse psychological stress-induced tumour proliferation and invasion has not been reported to date. Here, we found that curcumin not only inhibited the growth of xenografts in chronically stressed nude mice, but also decreased the expression of matrix metalloproteinase (MMP)-2/9 and CD147 in tumour tissues. Exogenous norepinephrine (NE) was used to stimulate glioma cells to simulate the stress environment in vitro, and it was found that curcumin inhibited the NE-induced proliferation and invasion of glioma cells in a dose-dependent manner. Further research found that the effects of NE on glioma cells could lead to the activation of the mitogen-activated protein kinase (MAPK) signalling pathway through ß-adrenergic receptor, while curcumin suppressed the level of extracellular signal-regulated kinase (ERK)1/2 phosphorylation. In addition, blocking ERK1/2 expression with U0126 resulted in the down-regulated expression of CD147, which further led to the decreased expression of MMP-2 and MMP-9. Curcumin could also inhibit the expression of cyclin D1/CDK4/6 and anti-apoptotic protein Bcl-2/Bcl-XL induced by NE, and induced cell cycle changes and increased apoptosis. Therefore, curcumin may be a potential candidate drug for preventing and treating the progression of glioma induced by adverse psychological stress.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation/drug effects , Curcumin/pharmacology , Glioma/metabolism , MAP Kinase Signaling System , Stress, Psychological/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclin D1/metabolism , Down-Regulation , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Norepinephrine/toxicity
3.
Exp Eye Res ; 195: 108048, 2020 06.
Article in English | MEDLINE | ID: mdl-32376471

ABSTRACT

Tissue injury causes the secretion of stress hormone catecholamine and increases susceptibility to opportunistic infection. Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen that is a leading cause of microbial keratitis usually associated with ocular injury or contact lens wear. However, the effect of catecholamine on P. aeruginosa induced corneal infection is unknown. Here, we test if norepinephrine (NE) would promote the progression of P. aeruginosa keratitis in mice. Adult C57BL/6 mouse corneas were scarified and then inoculated with P. aeruginosa. The content of NE was elevated in corneas after scarification and inoculation with P. aeruginosa. Then, exogenous NE was applied to the infected corneas at 24 h after inoculation; control eyes were treated with sterile saline. Topical application of NE aggravated the severity of P. aeruginosa keratitis, accompanied with the increase of clinical score, bacterial load, pathological changes, neutrophils infiltration, bacterial virulence factors and proinflammatory factors levels. In order to further verify the role of NE, N-(2-Chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4), a neurotoxin selected to deplete NE, was injected subconjunctivally 12 h before scarification. Pre-depletion of local NE by DSP-4 significantly alleviated the severity of corneal infection. Moreover, NE was also confirmed to increase the bacterial growth and the expression of virulence factors gene in vitro. Together, these data showed that increased corneal NE content facilitated the progression of P. aeruginosa keratitis in mice by amplifying host excessive inflammatory response and bacterial virulence. Therefore, targeting NE may provide a potential strategy for the treatment of P. aeruginosa keratitis.


Subject(s)
Corneal Ulcer/chemically induced , Epithelium, Corneal/pathology , Eye Infections, Bacterial/pathology , Keratitis/pathology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/physiology , Animals , Bacterial Load , Corneal Ulcer/pathology , Disease Models, Animal , Epithelium, Corneal/drug effects , Epithelium, Corneal/microbiology , Eye Infections, Bacterial/microbiology , Keratitis/microbiology , Mice , Mice, Inbred C57BL , Norepinephrine/toxicity , Pseudomonas Infections/microbiology
4.
Biomed Chromatogr ; 34(7): e4832, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32190911

ABSTRACT

Injectable solutions containing epinephrine (EPI) and norepinephrine (NE) are not stable, and their degradation is favored mainly by the oxidation of catechol moiety. As studies of these drugs under forced degradation conditions are scarce, herein, we report the identification of their degradation products (DP) in anesthetic formulations by the development of stability-indicating HPLC method. Finally, the risk assessment of the major degradation products was evaluated using in silico toxicity approach. HPLC method was developed to obtain a higher selectivity allowing adequate elution for both drugs and their DPs. The optimized conditions were developed using a C18 HPLC column, sodium 1-octanesulfonate, and methanol (80:20, v/v) as mobile phase, with a flow rate of 1.5 mL/min, UV detection at 199 nm. The analysis of standard solutions with these modifications resulted in greater retention time for EPI and NE, which allow the separation of these drugs from their respective DPs. Then, five DPs were identified and analyzed by in silico studies. Most of the DPs showed important alerts as hepatotoxicity and mutagenicity. To the best of our acknowledgment, this is the first report of a stability-indicating HPLC method that can be used with formulations containing catecholamines.


Subject(s)
Anesthetics , Chromatography, High Pressure Liquid/methods , Epinephrine , Norepinephrine , Anesthesia, Dental , Anesthetics/analysis , Anesthetics/chemistry , Anesthetics/toxicity , Animals , Computer Simulation , Drug Stability , Epinephrine/analysis , Epinephrine/chemistry , Epinephrine/toxicity , Limit of Detection , Linear Models , Mice , Norepinephrine/analysis , Norepinephrine/chemistry , Norepinephrine/toxicity , Rats , Reproducibility of Results
5.
Oxid Med Cell Longev ; 2020: 2563764, 2020.
Article in English | MEDLINE | ID: mdl-32104529

ABSTRACT

Norepinephrine (NE) is the naturally occurring adrenergic agonist that is released in response to hypotension, and it is routinely administered in clinical settings to treat moderate to severe hypotension that may occur during general anesthesia and shock states. Although NE has incontestable beneficial effects on blood pressure maintenance during hypotensive conditions, deleterious effects of NE on endothelial cell function may occur. In particular, the role of reactive oxygen species (ROS) and NADPH oxidase (Nox) on the deleterious effects of NE on endothelial cell function have not been fully elucidated. Therefore, we investigated the effects of NE on ROS production in rat lung microvascular endothelial cells (RLMEC) and its contribution to cell death. RLMEC were treated with NE (5 ng/mL) for 24 hours and ROS production was assessed by CellROX and DCFDA fluorescence. Nox activity was assessed by NADPH-stimulated ROS production in isolated membranes and phosphorylation of p47phox; cell death was assessed by flow cytometry and DNA fragmentation. Caspase activation was assessed by fluorescent microscopy. Nox1, Nox2, and Nox4 mRNA expression was assessed by real-time PCR. NE increased ROS production, Nox activity, p47phox phosphorylation, Nox2 and Nox4 mRNA content, caspase-3 activation, and RLMEC death. Phentolamine, an α 1-adrenoreceptor antagonist, inhibited NE-induced ROS production and Nox activity and partly inhibited cell death while ß-blockade had no effect. Apocynin and PEGSOD inhibited NE-induced caspase-3 activation and cell death while direct inhibition of caspase-3 abrogated NE-induced cell death. PEG-CAT inhibited NE-induced cell death but not caspase-3 activation. Collectively, these results indicate that NE induces RLMEC death via activation of Nox by α-adrenergic signaling and caspase-3-dependent pathways. NE has deleterious effects on RLMECs that may be important to its long-term therapeutic use.


Subject(s)
Caspase 3/metabolism , Endothelial Cells/drug effects , Lung/drug effects , NADPH Oxidases/metabolism , Norepinephrine/toxicity , Acetophenones/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Caspase Inhibitors/pharmacology , Cell Death , Endothelial Cells/metabolism , Lung/metabolism , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Phentolamine/pharmacology , Polyethylene Glycols/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/pharmacology
6.
Clin Exp Pharmacol Physiol ; 47(3): 403-411, 2020 03.
Article in English | MEDLINE | ID: mdl-31846086

ABSTRACT

Our group has previously shown in pithed rats that the cardiac sympathetic drive, which produces tachycardic responses, is inhibited by 5-HT via the activation of prejunctional 5-HT1B/1D/5 receptors. Interestingly, when 5-HT2 receptors are chronically blocked with sarpogrelate, the additional role of cardiac sympatho-inhibitory 5-HT1F receptors is unmasked. Although 5-HT2 receptors mediate tachycardia in rats, and the chronic blockade of 5-HT2 receptors unmasked 5-HT7 receptors mediating cardiac vagal inhibition, the role of 5-HT7 receptors in the modulation of the cardiac sympathetic tone remains virtually unexplored. On this basis, male Wistar rats were pretreated during 14 days with sarpogrelate (a 5-HT2 receptor antagonist) in drinking water (30 mg/kg/day; sarpogrelate-pretreated group) or equivalent volumes of drinking water (control group). Subsequently, the rats were pithed to produce increases in heart rate by either electrical preganglionic spinal (C7 -T1 ) stimulation of the cardiac sympathetic drive or iv administration of exogenous noradrenaline. The iv continuous infusion of AS-19 (a 5-HT7 receptor agonist; 10 µg/kg/min) (i) inhibited the tachycardic responses to sympathetic stimulation, but not those to exogenous noradrenaline only in sarpogrelate-pretreated rats. This inhibition was completely reversed by SB258719 (a selective 5-HT7 receptor antagonist; 1 mg/kg, iv) or glibenclamide (an ATP-sensitive K+ channel blocker; 20 mg/kg, iv). These results suggest that chronic 5-HT2 receptor blockade uncovers a cardiac sympatho-inhibitory mechanism mediated by 5-HT7 receptors, involving a hyperpolarization due to the opening of ATP-sensitive K+ channels. Thus, these findings support the role of 5-HT7 receptors in the modulation of the cardiac sympathetic neurotransmission.


Subject(s)
Adrenergic Fibers/physiology , Receptors, Serotonin, 5-HT2/physiology , Receptors, Serotonin/physiology , Serotonin 5-HT2 Receptor Antagonists/therapeutic use , Succinates/therapeutic use , Tachycardia/prevention & control , Adrenergic Fibers/drug effects , Animals , Dose-Response Relationship, Drug , Electric Stimulation/adverse effects , Heart Rate/drug effects , Heart Rate/physiology , Male , Norepinephrine/toxicity , Rats , Rats, Wistar , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Succinates/pharmacology , Sympathomimetics/toxicity , Tachycardia/etiology , Tachycardia/physiopathology
7.
J Cell Physiol ; 233(12): 9750-9762, 2018 12.
Article in English | MEDLINE | ID: mdl-30078229

ABSTRACT

Excessive proliferation, migration, and antiapoptosis of pulmonary artery (PA) smooth muscle cells (PASMCs) underlies the development of pulmonary vascular remodeling. The innervation of the PA is predominantly sympathetic, and increased levels of circulating catecholamines have been detected in pulmonary arterial hypertension (PAH), suggesting that neurotransmitters released by sympathetic overactivation may play an essential role in PAH. However, the responsible mechanism remains unclear. Here, to investigate the effects of norepinephrine (NE) on PASMCs and the related mechanism, we used 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, the proliferating cell nuclear antigen and the cell counting kit-8 assay to evaluate the proliferation of PASMCs, Boyden chamber migration, and wound-healing assays to assess migration and western blot analysis to investigate protein expression. We demonstrated that the phosphorylation level of the protein phosphatase 2A (PP2A) catalytic subunit (Y307) was higher in PAH patients and PAH models than in controls, both in vivo and in vitro. In addition, NE induced the proliferation and migration of PASMCs, which was attenuated by berberine (BBR), a Chinese herbal medicine, and/or PP2A overexpression. PP2A inhibition worsened NE-induced PAH and could not be reversed by BBR. Thus, PP2A is critical in driving PAH, and BBR may alleviate PAH via PP2A signaling pathways, thereby offering a potential therapeutic option for PAH.


Subject(s)
Berberine/administration & dosage , Hypertension, Pulmonary/drug therapy , Protein Phosphatase 2/genetics , Pulmonary Artery/drug effects , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Norepinephrine/toxicity , Pulmonary Artery/pathology , Rats , Signal Transduction/drug effects , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
8.
Food Chem Toxicol ; 118: 726-732, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29913233

ABSTRACT

Epinephrine and norepinephrine have been used in the management of anaphylactic reactions and cardiac resuscitation, along with treatment of asthma and glaucoma extensively, but their toxicological profiles are not yet completed. Based on this circumstance, various toxicological endpoints of epinephrine and norepinephrine were explored. Since there is a paucity of some endpoints' data, readacross was applied to fill the data gaps using analog approach. Along with structural similarity, biological and mechanistic plausibility were also considered in analog selection. The similarity justification and supporting experimental data were provided for uncertainty evaluation. Short term repeated dose toxicity values as NOAEL and LOAEL belonging to epinephrine were used to estimate the repeated dose toxicity of norepinephrine. The in vivo and in vitro mutagenicity tests were considered representative of genotoxicity. Both chemicals are showed to be non-genotoxic. They are experimentally reported to cause developmental and reproductive toxicity. For the carcinogenicity endpoint, a conclusion could not be reached because similar compounds were seen to show conflicting results.


Subject(s)
Epinephrine/toxicity , Norepinephrine/toxicity , Animals , Carcinogens/toxicity , Humans , Mutagens/toxicity , No-Observed-Adverse-Effect Level , Teratogens/toxicity , Toxicity Tests
9.
Aesthet Surg J ; 37(2): 239-245, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27590869

ABSTRACT

BACKGROUND: Lidocaine and epinephrine could potentially decrease adipocyte viability, but these effects have not been substantiated. The phosphorylation status of perilipin in adipocytes may be predictive of cell viability. Perilipin coats lipid droplets and restricts access of lipases; phospho-perilipin lacks this protective function. OBJECTIVES: The authors investigated the effects of tumescent solution containing lidocaine and epinephrine on the phosphorylation status of perilipin in adipocytes. METHODS: In this in vitro study, lipoaspirates were collected before and after tumescence from 15 women who underwent abdominoplasty. Fat samples were fixed, sectioned, and stained for histologic and immunohistochemical analyses. Relative phosphorylation of perilipin was inferred from pixel intensities of immunostained adipocytes observed with confocal microscopy. RESULTS: For adipocytes collected before tumescent infiltration, 10.08% of total perilipin was phosphorylated. In contrast, 30.62% of total perilipin was phosphorylated for adipocytes collected from tumescent tissue (P < .01). CONCLUSIONS: The tumescent technique increases the relative phosphorylation of perilipin in adipocytes, making these cells more vulnerable to lipolysis. Tumescent solution applied for analgesia or hemostasis of the donor site should contain the lowest possible concentrations of lidocaine and epinephrine. LEVEL OF EVIDENCE 5.


Subject(s)
Adipocytes/drug effects , Anesthesia, Local , Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Norepinephrine/pharmacology , Perilipin-1/metabolism , Adipocytes/metabolism , Adult , Anesthesia, Local/adverse effects , Anesthetics, Local/toxicity , Female , Fluorescent Antibody Technique , Humans , Lidocaine/toxicity , Lipolysis/drug effects , Microscopy, Confocal , Middle Aged , Norepinephrine/toxicity , Phosphorylation
10.
Int J Clin Exp Pathol ; 8(6): 6841-6, 2015.
Article in English | MEDLINE | ID: mdl-26261571

ABSTRACT

The present investigation aimed to evaluate the acute and chronic effect of stress (stress hormone) in male albino rat brain. Nor-epinephrine was used for the treatment and saline used for the control. Nor-epinephrine was dissolved in the saline and administered orally to the rats. Following nor-epinephrine administration, the brain was removed surgically at 6 h, 12 h and 45 days. Alanine tansaminase (ALT), aspartate transaminase (AST) and alkaline phosphatase (ALP) were significantly altered in the rats. Lipid peroxidation was measured as malondialdehyde (MDA), showed altered lipid peroxidation. Hematological markers such as packed cell volume (PCV), white blood cells (WBC), neutrophil, lymphocytes and hemoglobin were significantly altered compared to controls. Altered serum biochemical and hematological markers, lipid peroxidation and enzyme activities leads to adverse effect in the cellular metabolism and physiological activities of rats.


Subject(s)
Brain/drug effects , Norepinephrine/toxicity , Stress, Physiological/drug effects , Administration, Oral , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Brain/metabolism , Lipid Peroxidation/drug effects , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Norepinephrine/administration & dosage , Rats , Time Factors
11.
PLoS One ; 10(7): e0133664, 2015.
Article in English | MEDLINE | ID: mdl-26203906

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most common arrhythmias among old people. It causes serious long-term health problems affecting the quality of life. It has been suggested that the autonomic nervous system is involved in the onset and maintenance of AF in human. However, investigation of its pathogenesis and potential treatment has been hampered by the lack of suitable AF models in experimental animals. OBJECTIVES: Our aim was to establish a long-lasting AF model in mice. We also investigated the role of adrenergic receptor (AR) subtypes, which may be involved in the onset and duration of AF. METHODS AND RESULTS: Trans-esophageal atrial burst pacing in mice could induce AF, as previously shown, but with only a short duration (29.0 ± 8.1 sec). We found that adrenergic activation by intraperitoneal norepinephrine (NE) injection strikingly increased the AF duration. It increased the duration to more than 10 minutes, i.e., by more than 20-fold (656.2 ± 104.8 sec; P<0.001). In this model, a prior injection of a specific ß1-AR blocker metoprolol and an α1-AR blocker prazosin both significantly attenuated NE-induced elongation of AF. To further explore the mechanisms underlying these receptors' effects on AF, we assessed the SR Ca(2+) leak, a major trigger of AF, and consequent spontaneous SR Ca(2+) release (SCR) in atrial myocytes. Consistent with the results of our in-vivo experiments, both metoprolol and prazosin significantly inhibited the NE-induced SR Ca(2+) leak and SCR. These findings suggest that both ß1-AR and α1-AR may play important roles in the development of AF. CONCLUSIONS: We have established a long-lasting AF model in mice induced by adrenergic activation, which will be valuable in future AF study using experimental animals, such as transgenic mice. We also revealed the important role of ß1- and α1-AR-mediated signaling in the development of AF through in-vivo and in-vitro experiments.


Subject(s)
Atrial Fibrillation/chemically induced , Disease Models, Animal , Norepinephrine/toxicity , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, beta-1/drug effects , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Atrial Fibrillation/physiopathology , Calcium Signaling/drug effects , Cells, Cultured , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Injections, Intraperitoneal , Male , Metoprolol/pharmacology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Norepinephrine/administration & dosage , Prazosin/pharmacology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta-1/physiology , Sarcoplasmic Reticulum/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology
12.
Redox Biol ; 5: 243-252, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070033

ABSTRACT

Despite recent advances, the role of ROS in mediating hypertrophic and apoptotic responses in cardiac myocytes elicited by norepinephrine (NE) is rather poorly understood. We demonstrate through our experiments that H9c2 cardiac myoblasts treated with 2 µM NE (hypertrophic dose) generate DCFH-DA positive ROS only for 2h; while those treated with 100 µM NE (apoptotic dose) sustains generation for 48 h, followed by apoptosis. Though the levels of DCFH fluorescence were comparable at early time points in the two treatment sets, its quenching by DPI, catalase and MnTmPyP suggested the existence of a different repertoire of ROS. Both doses of NE also induced moderate levels of H2O2 but with different kinetics. Sustained but intermittent generation of highly reactive species detectable by HPF was seen in both treatment sets but no peroxynitrite was generated in either conditions. Sustained generation of hydroxyl radicals with no appreciable differences were noticed in both treatment sets. Nevertheless, despite similar profile of ROS generation between the two conditions, extensive DNA damage as evident from the increase in 8-OH-dG content, formation of γ-H2AX and PARP cleavage was seen only in cells treated with the higher dose of NE. We therefore conclude that hypertrophic and apoptotic doses of NE generate distinct but comparable repertoire of ROS/RNS leading to two very distinct downstream responses.


Subject(s)
Apoptosis/drug effects , Norepinephrine/toxicity , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Catalase/metabolism , Cell Line , DNA Damage/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Enzyme-Linked Immunosorbent Assay , Hydrogen Peroxide/metabolism , Hydroxyl Radical/metabolism , Microscopy, Fluorescence , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Rats , Superoxide Dismutase/metabolism
13.
Article in English | MEDLINE | ID: mdl-25819741

ABSTRACT

Noradrenaline (NA) is the active component of novel antifouling agents and acts by preventing attachment of fouling organisms. The goal of this study was to examine the toxicity of NA to the non-target zooplankton D. magna and C. dubia. Neonates were exposed to one of five concentrations of NA and effects on survival, reproduction and molting were determined. Calculated LC50 values were determined to be 46 and 38 µM in C. dubia and D. magna, respectively. A 10-day C. dubia study found that reproduction metrics were significantly impacted at non-lethal concentrations. In D. magna, concentrations greater than 40 µM significantly impacted molting. A toxicity test was conducted with D. magna using oxidized NA, which yielded similar results. These data indicate that both NA and oxidized NA are toxic to non-target zooplankton. Results obtained from this study can be used to guide future ecological risk assessments of catecholamine-based antifouling agents.


Subject(s)
Cladocera/drug effects , Molting/drug effects , Norepinephrine/toxicity , Pesticides/toxicity , Reproduction, Asexual/drug effects , Water Pollutants, Chemical/toxicity , Zooplankton/drug effects , Animals , Cladocera/growth & development , Cladocera/physiology , Daphnia/drug effects , Daphnia/growth & development , Daphnia/physiology , Drug Resistance , Female , Lethal Dose 50 , Male , Norepinephrine/chemistry , Osmolar Concentration , Oxidation-Reduction , Pesticides/chemistry , Species Specificity , Toxicity Tests, Acute , Toxicity Tests, Chronic , Water Pollutants, Chemical/chemistry , Zooplankton/growth & development , Zooplankton/physiology
14.
Food Funct ; 5(8): 1785-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24910858

ABSTRACT

The purpose of this study was to examine the efficacy of an aqueous wild blueberry extract and five wild blueberry polyphenol fractions on an in vitro model of heart disease. Adult rat cardiomyocytes were pretreated with extract and fractions, and then exposed to norepinephrine (NE). Cardiomyocyte hypertrophy, cell death, oxidative stress, apoptosis and cardiomyocyte contractile function as well as the activities of calpain, superoxide dismutase (SOD) and catalase (CAT) were measured in cardiomyocytes treated with and without NE and blueberry fraction (BF). Four of five blueberry fractions prevented cell death and cardiomyocyte hypertrophy induced by NE. Total phenolic fraction was used for all further analysis. The NE-induced increase in oxidative stress, nuclear condensation, calpain activity and lowering of SOD and CAT activities were prevented upon pretreatment with BF. Reduced contractile function was also significantly improved with BF pretreatment. Blueberry polyphenols prevent NE-induced adult cardiomyocyte hypertrophy and cell death. The protective effects of BF may be in part attributed to a reduction in calpain activity and oxidative stress.


Subject(s)
Blueberry Plants/chemistry , Calpain/metabolism , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Polyphenols/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Catalase/metabolism , Male , Norepinephrine/toxicity , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
15.
Hypertens Res ; 37(1): 26-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23985701

ABSTRACT

Both norepinephrine (NE) and connective tissue growth factor (CTGF) contribute to vascular fibrosis during hypertension. Recent studies indicate that farnesyl pyrophosphate synthase (FPPS) plays an important role in cardiac remodeling in hypertension. However, the role of FPPS in NE-induced fibrotic responses and related molecular mechanisms is unknown. Vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were stimulated with NE. The fibrotic responses were assessed by measuring CTGF, hydroxyproline (hyp), and α-1 procollagen I levels using Western blot, a hydroxyproline test kit, and real-time quantitative PCR assays, respectively. Ras activity was determined by a pull-down assay using a Ras activation assay kit and detected by Western blot. NE dose-dependently increased fibrosis in SHR-VSMCs, and this increase was significantly reduced by ibandronate, an inhibitor of FPPS. The addition of farnesol, but not geranylgeraniol, partially reversed the inhibitory effects of ibandronate. Furthermore, the anti-fibrotic effects of ibandronate could be mimicked by FTI-276 but not by GGTI-286. A pull-down assay showed that ibandronate reduced the NE-induced Ras activation. Moreover, ibandronate inhibited the NE-induced activation of p38, JNK, and ERK1/2. Only SB203580 (specific inhibitor of p38) diminished the NE-induced CTGF production. These results demonstrated that inhibiting FPPS prevents NE-induced fibrotic responses in SHR-VSMCs and that the Ras kinase and p38 pathways were the underlying mechanisms involved in this process.


Subject(s)
Dimethylallyltranstransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Hypertension/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Norepinephrine/antagonists & inhibitors , Norepinephrine/toxicity , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diphosphonates/pharmacology , Enzyme Activation , Fibrosis , Genes, ras/physiology , Hydroxyproline/metabolism , Ibandronic Acid , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/metabolism , Polyisoprenyl Phosphates/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sesquiterpenes/metabolism
16.
J Pharmacol Exp Ther ; 347(3): 681-96, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24071734

ABSTRACT

Twenty-two neuroleptic drugs were studied for interaction with the behavior induced by intravenous injection of apomorphine in rats. All compounds dose-dependently shortened the duration of the apomorphine-induced agitation and-with the exception of clozapine-shortened the onset of the de-arousal grooming that typically occurs immediately after the agitation phase has been terminated. Progressively higher doses were required to antagonize higher levels of apomorphine at earlier time intervals after the intravenous injection. The compounds also decreased palpebral opening, and most of them suppressed grooming behavior at higher doses. Compounds differed considerably in dose increments required for: 1) suppression of grooming, from 0.33 for clozapine to >600 for remoxipride, raclopride, and droperidol; 2) blockade of agitation at 5 minutes after apomorphine, from 2.6 for pimozide to 165 for chlorprothixene and 254 for remoxipride; 3) mild decrease of palpebral opening, from 0.21 for sertindole to 191 for remoxipride; and 4) pronounced decrease of palpebral opening, from 10 for melperone to >820 for raclopride. Only four compounds were able to advance grooming to 15 minutes postapomorphine, but again dose increments varied considerably: droperidol (3.4), pimozide (9.1), raclopride (42), and remoxipride (383). Based on these results obtained in a single animal model, compounds were differentiated in terms of behavioral specificity, incisiveness (the power to counteract the effects of progressively higher apomorphine concentrations), and sedative side-effect liability. Possible explanations for the observed differences and clinical relevance are discussed.


Subject(s)
Antipsychotic Agents/pharmacology , Apomorphine/pharmacology , Behavior, Animal/drug effects , Dopamine Agonists/pharmacology , Receptors, Dopamine/drug effects , Adrenergic alpha-Agonists/toxicity , Animals , Apomorphine/antagonists & inhibitors , Arousal/drug effects , Conjunctiva/drug effects , Dose-Response Relationship, Drug , Grooming/drug effects , Hypnotics and Sedatives , Linear Models , Male , Norepinephrine/toxicity , Psychomotor Agitation/drug therapy , Rats , Rats, Wistar , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Stimulation, Chemical
17.
Stress ; 16(1): 114-21, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22506837

ABSTRACT

Epinephrine and norepinephrine are produced during psychological stress and can directly bind to cells to induce DNA damage. These effects may have more long-lasting consequences such as DNA mutations resulting in an increased potential for cellular transformation and/or tumor progression. This study examined the molecular effects of a chronic (24 h) in vitro exposure to these stress hormones on murine 3T3 cells. Long exposures (24 h) in dose-response experiments with norepinephrine or epinephrine induced significant increases in DNA damage in treated cells compared to that of untreated controls as measured by the alkaline comet assay. Pre-treatment with a blocking agent (the ß-adrenergic receptor antagonist propranolol) eliminated this increase in damage. In addition, both norepinephrine and epinephrine increased cellular transformation, as assessed by growth in soft agar, and 3T3 cells pre-treated with either norepinephrine or epinephrine induced a more rapid onset of tumors and more aggressive tumor growth in nude mice. In summary, incubation of 3T3 cells with catecholamines results in long-term DNA damage as measured by increased transformed phenotypes and tumor progression, indicating that they are important mediators of stress effects on genomic instability and vulnerability to tumor formation.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Epinephrine/toxicity , Norepinephrine/toxicity , Stress, Psychological/physiopathology , Adrenergic beta-Antagonists/pharmacology , Animals , Carcinogenicity Tests , Comet Assay , DNA/drug effects , DNA Damage , Disease Progression , Dose-Response Relationship, Drug , Epinephrine/antagonists & inhibitors , Female , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Norepinephrine/antagonists & inhibitors , Propranolol/pharmacology
18.
J Pharm Pharmacol ; 63(12): 1604-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22060292

ABSTRACT

OBJECTIVES: This study was designed to evaluate the effect of curcumin on H9c2 cardiac cell line and primary rat cardiac myocytes, using purified noradrenaline as a hypertrophy-inducing agent. METHODS: The concentration of curcumin at which cells were treated was determined by MTT (3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay. The effect of this safe dose in preventing noradrenaline-induced cardiac hypertrophy was assessed by biochemical analysis (estimating total protein content), molecular analysis (using RT-PCR to study the expression of fetal genes like ANF), immunological analysis (by determining the nuclear localization of GATA-4) and electrophoretic mobility shift assay (EMSA; to study DNA binding activity of GATA-4). KEY FINDINGS: Curcumin at a concentration of 8 µm was found to suppress the increase in cell size, protein content and enhanced marker gene expression (ANF) caused by noradrenaline. Immunocytochemistry and Western blot analysis showed that curcumin suppressed the localization of transcription factor GATA-4 in the nucleus. It also showed a reduced DNA-binding activity in the presence of noradrenaline as confirmed by EMSA. CONCLUSIONS: These findings suggest that curcumin reduces the hypertrophic marker gene expression by inhibiting nuclear localization and DNA binding activity of GATA-4. Thus it has a great anti-hypertrophic potential.


Subject(s)
Cardiomegaly/prevention & control , Curcumin/pharmacology , Myocytes, Cardiac/ultrastructure , Norepinephrine/antagonists & inhibitors , Animals , Animals, Newborn , Blotting, Western , Cardiomegaly/chemically induced , Cell Nucleus/drug effects , Cell Size/drug effects , Cell Survival , Cells, Cultured , Cytosol/chemistry , Cytosol/metabolism , DNA/metabolism , Electrophoretic Mobility Shift Assay , GATA4 Transcription Factor/metabolism , Gene Expression/drug effects , Heart Ventricles/cytology , Immunohistochemistry , Myocytes, Cardiac/drug effects , Norepinephrine/toxicity , RNA/biosynthesis , RNA/genetics , Rats
19.
Brain Res ; 1324: 64-74, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20153300

ABSTRACT

Degeneration of noradrenergic neurons in the locus coeruleus (LC) and dysfunction of the prefrontal cortex were regarded as playing a specific role in the occurrence of non-motor symptoms in Parkinson's disease. The present study examined the spontaneous firing rate and firing pattern of medial prefrontal cortex (mPFC) pyramidal neurons, and effects of alpha(2)-adrenoceptor agonist UK-14,304 and antagonist yohimbine on the neuronal activity in rats with 6-hydroxydopamine lesions of the LC, medial forebrain bundle (MFB) and with combined MFB and LC lesions. The firing rate of mPFC pyramidal neurons in rats with lesions of the LC and with combine LC and MFB lesions is significantly higher than that of normal and MFB-lesioned rats and the firing pattern of these neurons in rats with lesions of the LC and with combine LC and MFB lesions also changed significantly towards more regular compared with normal and MFB-lesioned rats. The local administration of UK-14,304 in the mPFC inhibited the firing activity of the pyramidal neurons in normal rats and rats with lesions of the LC, MFB and with combined LC and MFB lesions, while yohimbine increased the firing activity of the pyramidal neurons. These results indicate that the lesions of the LC lead to hyperactivity of mPFC pyramidal neurons in normal and MFB-lesioned rats, and the postsynaptic alpha(2)-adrenoceptors may partially mediate the inhibitory effects of LC-noradrenergic system on the firing activity of pyramidal neurons in the mPFC, suggesting that LC-noradrenergic system plays an important role in the functional disorders of mPFC in Parkinson's disease.


Subject(s)
Locus Coeruleus/drug effects , Medial Forebrain Bundle/drug effects , Neurotoxins/toxicity , Norepinephrine/toxicity , Prefrontal Cortex/physiopathology , Pyramidal Cells/physiopathology , Receptors, Adrenergic, alpha-2/metabolism , Action Potentials/drug effects , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Brimonidine Tartrate , Locus Coeruleus/physiopathology , Male , Medial Forebrain Bundle/physiopathology , Oxidopamine/toxicity , Parkinsonian Disorders , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Yohimbine/pharmacology
20.
Neurobiol Aging ; 31(12): 2103-14, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19152986

ABSTRACT

Classical pathological signs of Parkinson's disease (PD) include loss of dopaminergic neurons in substantia nigra (SN) and noradrenergic neurons in locus coeruleus (LC), and deposition of Lewy bodies rich in the presynaptic protein alpha-synuclein (ASYN). Mammalian genetic models based on ASYN overexpression, however, have generally not reproduced the profound dopaminergic deficit of PD and do not display classical PD phenotypes. In the current study we examined these catecholaminergic systems in transgenic (Tg) mice expressing the A53T mutant of human ASYN under the Prion promoter. Surprisingly we detected a substantial reduction in norepinephrine (NE), but not dopamine (DA), levels in spinal cord, olfactory bulb and striatum of aged (15-month-old), but not young (4-month-old) transgenic compared to control mice. In spinal cord and olfactory bulb of 15-month-old Tg mice there was an age-dependent decrease in tyrosine hydroxylase (TH) protein levels, which in spinal cord was accompanied by a decrease in TH-positive terminals detected by immunohistochemistry. There was no difference in the number of TH-positive neuron cell bodies in SN or LC between Tg and control mice. We conclude that aberrant ASYN, expressed in both SN and LC, induces preferential degeneration of noradrenergic terminals. These observations suggest that in mice the NE may be more vulnerable than the DA system to the toxic effects of aberrant alpha-synuclein, and are in line with the major damage to the NE system that occurs in patients with PD.


Subject(s)
Norepinephrine/deficiency , Parkinson Disease/genetics , Parkinson Disease/physiopathology , alpha-Synuclein/genetics , Alanine/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Norepinephrine/biosynthesis , Norepinephrine/toxicity , Parkinson Disease/metabolism , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...