Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Inhal Toxicol ; 35(13-14): 333-349, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38060410

ABSTRACT

INTRODUCTION: Exposures to particulate matter (PM) from combustion sources can exacerbate preexisting asthma. However, the cellular and molecular mechanisms by which PM promotes the exacerbation of asthma remain elusive. We used a house dust mite (HDM)-induced mouse model of asthma to test the hypothesis that inhaled DCB230, which are PM containing environmentally persistent free radicals (EPFRs), will aggravate asthmatic responses. METHODS: Groups of 8-10-week-old C57BL/6 male mice were exposed to either air or DCB230 aerosols at a concentration of 1.5 mg/m3 4 h/day for 10 days with or without prior HDM-induction of asthma. RESULTS: Aerosolized DCB230 particles formed small aggregates (30-150 nm). Mice exposed to DCB230 alone showed significantly reduced lung tidal volume, overexpression of the Muc5ac gene, and dysregulation of 4 inflammation related genes, Ccl11, Ccl24, Il-10, and Tpsb2. This suggests DCB230 particles interacted with the lung epithelium inducing mucous hypersecretion and restricting lung volume. In addition to reduced lung tidal volume, compared to respective controls, the HDM + DCB230-exposed group exhibited significantly increased lung tissue damping and up-regulated expression of Muc5ac, indicating that in this model, mucous hypersecretion may be central to pulmonary dysfunction. This group also showed augmented lung eosinophilic inflammation accompanied by an up-regulation of 36 asthma related genes. Twelve of these genes are part of IL-17 signaling, suggesting that this pathway is critical for DCB230 induced toxicity and adjuvant effects in lungs previously exposed to HDM. CONCLUSION: Our data indicate that inhaled DCB230 can act as an adjuvant, exacerbating asthma through IL-17-mediated responses in a HDM mouse model.


Subject(s)
Asthma , Pneumonia , Mice , Male , Animals , Particulate Matter/toxicity , Pyroglyphidae , Interleukin-17/toxicity , Mice, Inbred C57BL , Asthma/chemically induced , Asthma/genetics , Lung , Free Radicals/toxicity , Disease Models, Animal , Inflammation
2.
Vaccines (Basel) ; 10(9)2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36146588

ABSTRACT

BACKGROUND: Methamphetamine use disorder (MUD) is a growing health concern with no FDA-approved treatment. The present series of studies build upon our previous work developing an anti-methamphetamine (MA) vaccine for MUD. We determined the effects of a formulation that included tetanus-toxoid (TT) conjugated to succinyl-methamphetamine (TT-SMA) adsorbed onto aluminum hydroxide (alum) in combination with the novel Toll-Like Receptor-5 agonist, entolimod. METHODS: Mice were vaccinated (0, 3, 6 weeks) with TT-SMA+alum and various doses of entolimod to determine an optimal dose for enhancing immunogenicity against MA. Functional effects were then assessed using MA-induced locomotor activation in mice. Experiments using passive immunization of antibodies generated by the vaccine tested its ability to attenuate MA-induced cardiovascular effects and alter the reinforcing effects of MA in an MA-induced reinstatement of a drug seeking model of relapse in male and female rats. RESULTS: Antibody levels peaked at 10 weeks following vaccination with TT-SMA+alum combined with entolimod (1, 3 and 10 µg). MA-induced locomotor activation was significantly attenuated in vaccinated vs. unvaccinated mice and antibody levels significantly correlated with ambulation levels. Passive immunization decreased mean arterial pressure following MA dosing in rats of both sexes but did not alter heart rate. Passive immunization also attenuated the ability of MA to reinstate extinguished drug-seeking behavior in male and female rats. Results support further development of this vaccine for relapse prevention for individuals with MUD.

3.
Am J Physiol Heart Circ Physiol ; 321(4): H667-H683, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34415187

ABSTRACT

Airborne particulate matter (PM) is associated with an increased risk for cardiovascular diseases. Although the goal of thermal remediation is to eliminate organic wastes through combustion, when incomplete combustion occurs, organics chemisorb to transition metals to generate PM-containing environmentally persistent free radicals (EPFRs). Similar EPFR species have been detected in PM found in diesel and gasoline exhaust, woodsmoke, and urban air. Prior in vivo studies demonstrated that EPFRs reduce cardiac function secondary to elevations in pulmonary arterial pressures. In vitro studies showed that EPFRs increase ROS and cytokines in pulmonary epithelial cells. We thus hypothesized that EPFR inhalation would promote lung inflammation and oxidative stress, leading to systemic inflammation, vascular endothelial injury, and a decline in vascular function. Mice were exposed to EPFRs for either 4 h or for 4 h/day for 10 days and lung and vascular function were assessed. After a 4-h exposure, plasma nitric oxide (NO) was reduced while endothelin-1 (ET-1) was increased, however lung function was not altered. After 10 day, plasma NO and ET-1 levels were again altered and lung tidal volume was reduced. These time course studies suggested the vasculature may be an early target of injury. To test this hypothesis, an intermediate time point of 3 days was selected. Though the mice exhibited no marked inflammation in either the lung or the blood, we did note significantly reduced endothelial function concurrent with a reduction in lung tidal volume and an elevation in annexin V protein levels in the lung. Although vascular dysfunction was not dependent upon inflammation, it may be associated with an injury at the air-blood interface. Gene expression analysis suggested roles for oxidative stress and aryl hydrocarbon receptor (Ahr) signaling. Studies probing the relationship between pulmonary oxidative stress and AhR signaling at the air-blood interface with vascular dysfunction seem warranted.NEW & NOTEWORTHY Particulate matter (PM) resulting from the combustion of organic matter is known to contribute to cardiopulmonary disease. Despite hypotheses that cardiovascular dysfunction occurring after PM exposures is secondary to lung or systemic inflammation, these studies investigating exposures to PM-containing environmentally persistent free radicals (EPFRs) demonstrate that cardiovascular dysfunction precedes pulmonary inflammation. The cardiopulmonary health consequences of EPFRs have yet to be thoroughly evaluated, especially in healthy, adult mice. Our data suggest the vasculature as a direct target of PM exposure, and our studies aimed to elucidate the mechanisms contributing to EPFR-induced vascular dysfunction.


Subject(s)
Aorta/drug effects , Cardiovascular Diseases/chemically induced , Endothelium, Vascular/drug effects , Environmental Pollutants/toxicity , Free Radicals/toxicity , Lung/drug effects , Particulate Matter/toxicity , Pneumonia/chemically induced , Animals , Aorta/metabolism , Aorta/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Dose-Response Relationship, Drug , Endothelin-1/blood , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Inhalation Exposure , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred C57BL , Nitric Oxide/blood , Oxidative Stress , Pneumonia/genetics , Pneumonia/metabolism , Pneumonia/physiopathology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Tidal Volume/drug effects , Time Factors
4.
Arch Toxicol ; 94(12): 4067-4084, 2020 12.
Article in English | MEDLINE | ID: mdl-32894303

ABSTRACT

Mitoxantrone (MTX) is used to treat several types of cancers and to improve neurological disability in multiple sclerosis. Unfortunately, cardiotoxicity is a severe and common adverse effect in MTX-treated patients. Herein, we aimed to study early and late mechanisms of MTX-induced cardiotoxicity using murine HL-1 cardiomyocytes. Cells were exposed to MTX (0.1, 1 or 10 µM) during short (2, 4, 6, or 12 h) or longer incubation periods (24 or 48 h). At earlier time points, (6 and 12 h) cytotoxicity was already observed for 1 and 10 µM MTX. Proteomic analysis of total protein extracts found 14 proteins with higher expression and 26 with lower expression in the cells exposed for 12 h to MTX (pH gradients 4-7 and 6-11). Of note, the expression of the regulatory protein 14-3-3 protein epsilon was increased by a factor of two and three, after exposure to 1 and 10 µM MTX, respectively. At earlier time-points, 10 µM MTX increased intracellular ATP levels, while decreasing media lactate levels. At later stages (24 and 48 h), MTX-induced cytotoxicity was concentration and time-dependent, according to the MTT reduction and lactate dehydrogenase leakage assays, while caspase-9, -8 and -3 activities increased at 24 h. Regarding cellular redox status, total glutathione increased in 1 µM MTX (24 h), and that increase was dependent on gamma-glutamylcysteine synthetase activity. Meanwhile, for both 1 and 10 µM MTX, oxidized glutathione was significantly higher than control at 48 h. Moreover, MTX was able to significantly decrease proteasomal chymotrypsin-like activity in a concentration and time-independent manner. In summary, MTX significantly altered proteomic, energetic and oxidative stress homeostasis in cardiomyocytes at clinically relevant concentrations and our data clearly demonstrate that MTX causes early cardiotoxicity that needs further study.


Subject(s)
Energy Metabolism/drug effects , Heart Diseases/chemically induced , Mitoxantrone/toxicity , Myocytes, Cardiac/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteome , Proteomics , Animals , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Cell Line , Dose-Response Relationship, Drug , Heart Diseases/metabolism , Mice , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Protein Carbonylation , Time Factors
5.
Med Res Rev ; 38(4): 1332-1403, 2018 07.
Article in English | MEDLINE | ID: mdl-29315692

ABSTRACT

Cardiovascular diseases are a leading cause of morbidity and mortality in most developed countries of the world. Pharmaceuticals, illicit drugs, and toxins can significantly contribute to the overall cardiovascular burden and thus deserve attention. The present article is a systematic overview of drugs that may induce distinct cardiovascular toxicity. The compounds are classified into agents that have significant effects on the heart, blood vessels, or both. The mechanism(s) of toxic action are discussed and treatment modalities are briefly mentioned in relevant cases. Due to the large number of clinically relevant compounds discussed, this article could be of interest to a broad audience including pharmacologists and toxicologists, pharmacists, physicians, and medicinal chemists. Particular emphasis is given to clinically relevant topics including the cardiovascular toxicity of illicit sympathomimetic drugs (e.g., cocaine, amphetamines, cathinones), drugs that prolong the QT interval, antidysrhythmic drugs, digoxin and other cardioactive steroids, beta-blockers, calcium channel blockers, female hormones, nonsteroidal anti-inflammatory, and anticancer compounds encompassing anthracyclines and novel targeted therapy interfering with the HER2 or the vascular endothelial growth factor pathway.


Subject(s)
Adrenergic beta-Antagonists/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular System/drug effects , Steroids/adverse effects , Alkaloids/adverse effects , Amphetamines/adverse effects , Animals , Anti-Arrhythmia Agents/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antineoplastic Agents/adverse effects , Calcium Channel Blockers/adverse effects , Cardiovascular Diseases/drug therapy , Cocaine/adverse effects , Digoxin/adverse effects , Female , Heart Rate/drug effects , Hormones/adverse effects , Humans , Male , Stroke/drug therapy , Vascular Endothelial Growth Factor A
6.
J Am Coll Cardiol ; 70(17): 2139-2153, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29050562

ABSTRACT

BACKGROUND: Sustained sympathetic activation contributes to the progression of myocardial cell injury, cardiac fibrosis, and left ventricular (LV) dysfunction in heart failure (HF). OBJECTIVES: This study investigated the effects of radiofrequency renal nerve denervation (RF-RDN) on the pathobiology of HF and the interaction between the renal sympathetic nerves and natriuretic peptide (NP) metabolism. METHODS: Spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto rats (WKY) were subjected to 45 min of coronary artery ligation and reperfusion for 12 weeks. At 4 weeks post-reperfusion, SHR and WKY underwent either bilateral RF-RDN or sham-RDN. RESULTS: Following RF-RDN in both strains, LV ejection fraction remained significantly above those levels in respective sham-RDN rats, and at the end of the 12-week study, rats in both strains had significantly reduced LV fibrosis and improved vascular function. RF-RDN therapy significantly improved vascular reactivity to endothelium-dependent and -independent vasodilators as well as vascular compliance in the setting of severe HF. Improvements in LV function were accompanied by significant elevations in circulating NP as compared to those associated with sham-RDN. Further investigation into the cause of increased circulating NP levels demonstrated that RF-RDN significantly inhibited renal neprilysin activity in SHR and WKY with HF. Likewise, chronic treatment with the beta1 antagonist bisoprolol inhibited renal neprilysin activity and increased circulation NP levels in WKY with HF. CONCLUSIONS: This study identifies a novel endogenous pathway by which the renal nerves participate in the degradation of cardioprotective NP. Furthermore, removal of the influence of the renal nerves on kidney function attenuates renal neprilysin activity, augments circulating NP levels, reduces myocardial fibrosis, and improves LV function in the setting of HF.


Subject(s)
Heart Failure/therapy , Kidney/innervation , Neprilysin/antagonists & inhibitors , Sympathectomy , Aminobutyrates/pharmacology , Angiotensin II/blood , Animals , Biphenyl Compounds , Bisoprolol/pharmacology , Blood Pressure , Drug Combinations , Echocardiography , Myocardium/chemistry , Myocardium/pathology , Neprilysin/physiology , Nitrites/analysis , Norepinephrine/blood , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Renal Artery/innervation , Renin/blood , Reperfusion Injury/physiopathology , Tetrazoles/pharmacology , Valsartan , Ventricular Function, Left/physiology
7.
Peptides ; 95: 25-32, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28720396

ABSTRACT

Mitoxantrone (MXT) is an androstenedione that is used to treat cancers and progressive forms of multiple sclerosis; however, its use is limited by its cardiotoxicity. Pituitary adenylate cyclase activating polypeptide (PACAP) is a member of the secretin/growth hormone-releasing hormone/vasoactive intestinal peptide family and has many functions, including cytoprotection and immunosuppression. We tested the hypothesis that PACAP can protect against MXT-induced cardiotoxicity in mice. Female BALB/c mice were treated once weekly for 4 weeks with saline (n=14) or MXT (3mg/kg, i.p.; n=14). Half of the mice in each group received PACAP (10µg, i.p.) 1h before and 24 and 48h after MXT, while the remaining mice received injections of saline on the same schedule. Echocardiography was used to assess cardiac structure and function. In mice treated with MXT and saline, body weight was significantly reduced after the third dose of MXT. PACAP significantly attenuated the reduction in body weight; however, the weights did not return to control level. Compared to controls, MXT-treated mice had significantly increased left ventricular (LV) diameter and LV volume and decreased LV posterior wall thickness. Fractional shortening (FS) and ejection fraction (EF) were also significantly decreased. Treatment with PACAP prevented MXT-induced LV dilation and significantly attenuated the reductions in FS and EF, although FS and EF did not return to control level. PACAP38 did not prevent MXT-induced decreases in LV posterior wall thickness. MXT dose-dependently decreased the viability of cultured U937 (human leukemia) cells; PACAP did not protect cultured U937 cells from MXT-mediated cell death. In conclusion, PACAP can attenuate MXT-mediated LV dilation and dysfunction in mice.


Subject(s)
Heart Injuries/drug therapy , Mitoxantrone/adverse effects , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Ventricular Dysfunction, Left/drug therapy , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/pathology , Cell Line, Tumor , Disease Models, Animal , Heart Injuries/chemically induced , Heart Injuries/pathology , Humans , Mice , Mitoxantrone/therapeutic use , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/pathology , Protective Agents/administration & dosage , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/pathology
8.
Cardiovasc Toxicol ; 17(2): 140-149, 2017 04.
Article in English | MEDLINE | ID: mdl-27052339

ABSTRACT

Samples of environmental particulate matter contain environmentally persistent free radicals (EPFRs) capable of sustained generation of oxygen radicals. While exposure to EPFRs produces cardiac toxicity and oxidative stress in experimental animals, the underlying mechanisms are largely unknown. To determine whether EPFRs could directly damage cardiomyocytes, cultured mouse cardiomyocytes (HL-1) and primary rat adult left ventricular myocytes (ALVM) were incubated with an EPFR consisting of 1,2-dichlorobenzene chemisorbed to CuO-coated silica beads (DCB230). Treatment with DCB230 killed both HL-1 and ALVM in a dose- and time-dependent manner. The cytotoxic effects of DCB230 were significantly attenuated by treatment with α-tocopherol. One to 2 h after exposure to DCB230, there were significant reductions in mitochondrial membrane potential and significant increases in cleaved caspase-9, but no significant increases in DNA damage or cell death. After 8 h of treatment, there were significant increases in caspase-3, caspase-9, DNA damage and PARP cleavage associated with significant cell death. Together, these data indicate that DCB230 kills HL-1 myocytes by inducing oxidative stress that initiates apoptosis, with the intrinsic or mitochondrial pathway acting early in the apoptotic signaling process.


Subject(s)
Apoptosis/drug effects , Environmental Pollutants/toxicity , Free Radicals/toxicity , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Particulate Matter/toxicity , Animals , Antioxidants/pharmacology , Caspases/metabolism , Cell Line , Dose-Response Relationship, Drug , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
9.
Am J Physiol Heart Circ Physiol ; 308(9): H998-H1006, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25681431

ABSTRACT

Increases in airborne particulate matter (PM) are linked to increased mortality from myocardial ischemia. PM contains environmentally persistent free radicals (EPFRs) that form as halogenated hydrocarbons chemisorb to transition metal oxide-coated particles, and are capable of sustained redox cycling. We hypothesized that exposure to the EPFR DCB230 would increase cardiac vulnerability to subsequent myocardial ischemia-reperfusion (MI/R) injury. Rats were exposed to DCB230 or vehicle via nose-only inhalation (230 µg max/day) over 30 min/day for 7 days. MI/R or sham MI/R (sham) was initiated 24 h after the final exposure. Following 1 or 7 days of reperfusion, left ventricular (LV) function was assessed and infarct size measured. In vehicle-exposed rats, MI/R injury did not significantly reduce cardiac output (CO), stroke volume (SV), stroke work (SW), end-diastolic volume (EDV), or end-systolic volume (ESV) after 1 day of reperfusion, despite significant reductions in end-systolic pressure (ESP). Preload-recruitable SW (PRSW; contractility) was elevated, presumably to maintain LV function. MI/R 1-day rats exposed to DCB230 also had similarly reduced ESP. Compared with vehicle controls, CO, SV, and SW were significantly reduced in DCB230-exposed MI/R 1-day rats; moreover, PRSW did not increase. DCB230's effects on LV function dissipated within 8 days of exposure. These data show that inhalation of EPFRs can exacerbate the deficits in LV function produced by subsequent MI/R injury. Infarct size was not different between the MI/R groups. We conclude that inhalation of EPFRs can compromise cardiac function during MI/R injury and may help to explain the link between PM and MI/R-related mortality.


Subject(s)
Free Radicals/toxicity , Myocardial Infarction/etiology , Myocardial Reperfusion Injury/etiology , Particulate Matter/toxicity , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/drug effects , Animals , Disease Models, Animal , Inhalation Exposure , Male , Myocardial Infarction/chemically induced , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Particle Size , Rats, Sprague-Dawley , Risk Assessment , Risk Factors , Stroke Volume/drug effects , Time Factors , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
10.
Chem Res Toxicol ; 26(12): 1862-71, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24224526

ABSTRACT

Particulate matter (PM) is emitted during thermal decomposition of waste. During this process, aromatic compounds chemisorb to the surface of metal-oxide-containing PM, forming a surface-stabilized environmentally persistent free radical (EPFR). We hypothesized that EPFR-containing PM redox cycle to produce ROS and that this redox cycle is maintained in biological environments. To test our hypothesis, we incubated model EPFRs with the fluorescent probe dihydrorhodamine (DHR). Marked increases in DHR fluorescence were observed. Using a more specific assay, hydroxyl radicals ((•)OH) were also detected, and their level was further increased by cotreatment with thiols or ascorbic acid (AA), known components of epithelial lining fluid. Next, we incubated our model EPFR in bronchoalveolar lavage fluid (BALF) or serum. Detection of EPFRs and (•)OH verified that PM generate ROS in biological fluids. Moreover, incubation of pulmonary epithelial cells with EPFR-containing PM increased (•)OH levels compared to those in PM lacking EPFRs. Finally, measurements of oxidant injury in neonatal rats exposed to EPFRs by inhalation suggested that EPFRs induce an oxidant injury within the lung lining fluid and that the lung responds by increasing antioxidant levels. In summary, our EPFR-containing PM redox cycle to produce ROS, and these ROS are maintained in biological fluids and environments. Moreover, these ROS may modulate toxic responses of PM in biological tissues such as the lung.


Subject(s)
Free Radicals/metabolism , Models, Biological , Particulate Matter/chemistry , Particulate Matter/metabolism , Reactive Oxygen Species/metabolism , Animals , Free Radicals/chemistry , Humans , Oxidation-Reduction , Rats , Rats, Inbred BN
11.
Psychopharmacology (Berl) ; 225(3): 675-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22972412

ABSTRACT

RATIONALE: Exceedingly little experimental research exists on the popular recreational drug mephedrone (4-methylmethcathinone) despite clinical reports concerning its behavioral and cardiovascular toxicity. OBJECTIVES: To characterize mephedrone preclinically by examining its capacity to (1) serve as a discriminative stimulus, (2) disrupt the acquisition of response sequences, and (3) disrupt mean arterial pressure (MAP) and heart rate (HR). METHODS AND RESULTS: In one group of subjects that reliably discriminated 3.2 mg/kg of mephedrone from saline (n = 9), substitution tests indicated that stimulants (cocaine, MDMA, and methamphetamine) more closely approximated the mephedrone discriminative stimulus than non-stimulants (fenfluramine, morphine, and phencyclidine), although none fully substituted. In a second group (n = 6), mephedrone (0.56-10 mg/kg, i.p.) dose-dependently decreased response rate and increased errors in both components of a procedure in which subjects either acquired a new response sequence each session (repeated acquisition) or completed the same response sequence each session (performance). Finally, in a third group (n = 12), radio telemetry probes were used to measure the changes in MAP and HR elicited by mephedrone and then compared them to a known stimulant, methamphetamine. In these studies, mephedrone (0.01-9 mg/kg, i.v.) elicited increases in MAP and HR that were very similar to those elicited by methamphetamine (0.01-9 mg/kg, i.v.). The tachycardia and pressor responses to mephedrone (3 mg/kg) were blocked by the ß-blocker atenolol (1 mg/kg, i.v.) and the α1, α2-blocker phentolamine (3 mg/kg, i.v.), respectively. CONCLUSIONS: Mephedrone produces behavioral and cardiovascular responses that are similar to other stimulants; however, differences from the classical stimulants were also apparent.


Subject(s)
Behavior, Animal/drug effects , Blood Pressure/drug effects , Heart Rate/drug effects , Illicit Drugs/toxicity , Methamphetamine/analogs & derivatives , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , Male , Methamphetamine/toxicity , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
12.
PLoS One ; 7(11): e48910, 2012.
Article in English | MEDLINE | ID: mdl-23155428

ABSTRACT

Angiotensin-converting enzyme type 2 (ACE2) has been shown to be an important member of the renin angiotensin system. Previously, we observed that central ACE2 reduces the development of hypertension following chronic angiotensin II (Ang-II) infusion in syn-hACE2 transgenic (SA) mice, in which the human ACE2 transgene is selectively targeted to neurons. To study the physiological consequences of central ACE2 over-expression on cardiac function and cardiac hypertrophy, SA and non-transgenic (NT) mice were infused with Ang-II (600 ng/kg/min, sc) for 14 days, and cardiac function was assessed by echocardiography. Blood pressure (BP), hemodynamic parameters, left ventricle (LV) mass/tibia length, relative ventricle wall thickness (2PW/LVD), cardiomyocyte diameters and collagen deposition were similar (P>0.05) between NT and SA mice during saline infusion. After a 2-week infusion, BP was elevated in NT but not in SA mice. Although ejection fraction and fractional shortening were not altered, Ang-II infusion increased 2PW/LVD compared to saline infusion in NT mice. Interestingly, the 2PW/LVD and LV mass/tibia ratios were significantly lower in SA compared to NT mice at the end of infusion. Moreover, Ang-II infusion significantly increased arterial collagen deposition and cardiomyocytes diameter in NT mice but not in transgenic animals (P<0.05). More importantly, ACE2 over expression significantly reduced the Ang-II-mediated increase in urine norepinephrine levels in SA compared to NT mice. The protective effect of ACE2 appears to involve reductions in Ang-II-mediated hypertension and sympathetic nerve activity.


Subject(s)
Brain/metabolism , Cardiomegaly/genetics , Heart/physiopathology , Neurons/metabolism , Peptidyl-Dipeptidase A/genetics , Angiotensin II , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Hemodynamics/drug effects , Hemodynamics/physiology , Mice , Norepinephrine/urine , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
13.
Am J Physiol Heart Circ Physiol ; 303(9): H1135-42, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22942180

ABSTRACT

Epidemiological studies have consistently linked inhalation of particulate matter (PM) to increased cardiac morbidity and mortality, especially in at risk populations. However, few studies have examined the effect of PM on baseline cardiac function in otherwise healthy individuals. In addition, airborne PM contain environmentally persistent free radicals (EPFR) capable of redox cycling in biological systems. The purpose of this study was to determine whether nose-only inhalation of EPFRs (20 min/day for 7 days) could decrease baseline left ventricular function in healthy male Sprague-Dawley rats. The model EPFR tested was 1,2-dichlorobenzene chemisorbed to 0.2-µm-diameter silica/CuO particles at 230°C (DCB230). Inhalation of vehicle or silica particles served as controls. Twenty-four hours after the last exposure, rats were anesthetized (isoflurane) and ventilated (3 l/min), and left ventricular function was assessed using pressure-volume catheters. Compared with controls, inhalation of DCB230 significantly decreased baseline stroke volume, cardiac output, and stroke work. End-diastolic volume and end-diastolic pressure were also significantly reduced; however, ventricular contractility and relaxation were not changed. DCB230 also significantly increased pulmonary arterial pressure and produced hyperplasia in small pulmonary arteries. Plasma levels of C-reactive protein were significantly increased by exposure to DCB230, as were levels of heme oxygenase-1 and SOD2 in the left ventricle. Together, these data show that inhalation of EPFRs, but not silica particles, decreases baseline cardiac function in healthy rats by decreasing cardiac filling, secondary to increased pulmonary resistance. These EPFRs also produced systemic inflammation and increased oxidative stress markers in the left ventricle.


Subject(s)
Blood Pressure/drug effects , Chlorobenzenes/pharmacology , Free Radicals/pharmacology , Heart/drug effects , Pulmonary Artery/drug effects , Administration, Inhalation , Animals , Blood Pressure/physiology , C-Reactive Protein/metabolism , Cardiac Output/drug effects , Cardiac Output/physiology , Chlorobenzenes/administration & dosage , Free Radicals/administration & dosage , Heart/physiology , Male , Models, Animal , Particulate Matter/administration & dosage , Particulate Matter/pharmacology , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Stroke Volume/drug effects , Stroke Volume/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
14.
PLoS One ; 6(8): e23337, 2011.
Article in English | MEDLINE | ID: mdl-21829729

ABSTRACT

OBJECTIVE: Resistance vessel remodeling is controlled by myriad of hemodynamic and neurohormonal factors. This study characterized structural and molecular remodeling in mesenteric resistance arteries (MRAs) in diabetic (db/db) and control (Db/db) mice. METHODS: Structural properties were assessed in isolated MRAs from 12 and 16 wk-old db/db and Db/db mice by pressure myography. Matrix regulatory proteins were measured by Western blot analysis. Mean arterial pressure and superior mesenteric blood flow were measured in 12 wk-old mice by telemetry and a Doppler flow nanoprobe, respectively. RESULTS: Blood pressure was similar between groups. Lumen diameter and medial cross-sectional area were significantly increased in 16 wk-old db/db MRA compared to control, indicating outward hypertrophic remodeling. Moreover, wall stress and cross-sectional compliance were significantly larger in diabetic arteries. These remodeling indices were associated with increased expression of matrix regulatory proteins matrix metalloproteinase (MMP)-9, MMP-12, tissue inhibitors of matrix metalloproteinase (TIMP)-1, TIMP-2, and plasminogen activator inhibitor-1 (PAI-1) in db/db arteries. Finally, superior mesenteric artery blood flow was increased by 46% in 12 wk-old db/db mice, a finding that preceded mesenteric resistance artery remodeling. CONCLUSIONS: These data suggest that flow-induced hemodynamic changes may supersede the local neurohormonal and metabolic milieu to culminate in hypertrophic outward remodeling of type 2 DM mesenteric resistance arteries.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Mesenteric Arteries/pathology , Animals , Blood Glucose/metabolism , Blood Pressure , Blotting, Western , Male , Mice , Mice, Inbred C57BL
15.
Basic Res Cardiol ; 106(6): 1123-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21744279

ABSTRACT

Little is known about the impact of type 2 diabetes mellitus (DM) on coronary arteriole remodeling. The aim of this study was to determine the mechanisms that underlie coronary arteriole structural remodeling in type 2 diabetic (db/db) mice. Passive structural properties of septal coronary arterioles isolated from 12- to 16-week-old diabetic db/db and control mice were assessed by pressure myography. Coronary arterioles from 12-week-old db/db mice were structurally similar to age-matched controls. By 16 weeks of age, coronary wall thickness was increased in db/db arterioles (p < 0.01), while luminal diameter was reduced (control: 118 ± 5 µm; db/db: 102 ± 4 µm, p < 0.05), augmenting the wall-to-lumen ratio by 58% (control: 5.9 ± 0.6; db/db: 9.5 ± 0.4, p < 0.001). Inward hypertrophic remodeling was accompanied by a 56% decrease in incremental elastic modulus (p < 0.05, indicating decreased vessel coronary wall stiffness) and a ~30% reduction in coronary flow reserve (CFR) in diabetic mice. Interestingly, aortic pulse wave velocity and femoral artery incremental elastic modulus were increased (p < 0.05) in db/db mice, indicating macrovascular stiffness. Molecular tissue analysis revealed increased elastin-to-collagen ratio in diabetic coronaries when compared to control and a decrease in the same ratio in the diabetic aortas. These data show that coronary arterioles isolated from type 2 diabetic mice undergo inward hypertrophic remodeling associated with decreased stiffness and increased elastin-to-collagen ratio which results in a decreased CFR. This study suggests that coronary microvessels undergo a different pattern of remodeling from macrovessels in type 2 DM.


Subject(s)
Arterioles/pathology , Coronary Vessels/pathology , Diabetes Mellitus, Type 2/pathology , Elasticity/physiology , Animals , Arterioles/chemistry , Arterioles/metabolism , Collagen Type I , Coronary Vessels/chemistry , Coronary Vessels/metabolism , Diabetes Mellitus, Type 2/metabolism , Elastin/analysis , Elastin/metabolism , Male , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction
16.
Cardiovasc Res ; 87(1): 111-8, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20139112

ABSTRACT

AIMS: Our aim was to test the hypothesis that the repeated, binge administration of methamphetamine would produce oxidative stress in the myocardium leading to structural remodeling and impaired left ventricular function. METHODS AND RESULTS: Echocardiography and Millar pressure-volume catheters were used to monitor left ventricular structure and function in rats subjected to four methamphetamine binges (3 mg/kg, iv for 4 days, separated by a 10-day drug-free period). Hearts from treated and control rats were used for histological or proteomic analysis. When compared with saline treatment, four methamphetamine binges produced eccentric left ventricular hypertrophy. The drug also significantly impaired systolic function (decreased fractional shortening, ejection fraction, and adjusted maximal power) and produced significant diastolic dysfunction (increased -dP/dt and tau). Dihydroethedium staining showed that methamphetamine significantly increased (285%) the levels of reactive oxygen species in the left ventricle. Treatment with methamphetamine also resulted in the tyrosine nitration of myofilament (desmin, myosin light chain) and mitochondrial (ATP synthase, NADH dehydrogenase, cytochrome c oxidase, prohibitin) proteins. Treatment with the superoxide dismutase mimetic, tempol in the drinking water prevented methamphetamine-induced left ventricular dilation and systolic dysfunction; however, tempol (2.5 mM) did not prevent the diastolic dysfunction. Tempol significantly reduced, but did not eliminate dihydroethedium staining in the left ventricle, nor did it prevent the tyrosine nitration of mitochondrial and contractile proteins. CONCLUSION: This study shows that oxidative stress plays a significant role in mediating methamphetamine-induced eccentric left ventricular dilation and systolic dysfunction.


Subject(s)
Myocardium/metabolism , Oxidative Stress , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Ventricular Remodeling , Actin Cytoskeleton/metabolism , Animals , Antioxidants/pharmacology , Cardiac Catheterization , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Male , Methamphetamine , Mitochondrial Proteins/metabolism , Myocardial Contraction , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Spin Labels , Stroke Volume , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Ultrasonography , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Ventricular Pressure , Ventricular Remodeling/drug effects
17.
Cardiovasc Toxicol ; 9(1): 30-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19333788

ABSTRACT

Repeated administration of 3,4-methylenedioxymethamphetamine (MDMA) (ecstasy) produces eccentric left ventricular (LV) dilation and diastolic dysfunction. While the mechanism(s) underlying this toxicity are unknown, oxidative stress plays an important role. MDMA is metabolized into redox cycling metabolites that produce superoxide. In this study, we demonstrated that metabolites of MDMA induce oxidative stress and contractile dysfunction in adult rat left ventricular myocytes. Metabolites of MDMA used in this study included alpha-methyl dopamine, N-methyl alpha-methyl dopamine and 2,5-bis(glutathion-S-yl)-alpha-MeDA. Dihydroethidium was used to detect drug-induced increases in reactive oxygen species (ROS) production in ventricular myocytes. Contractile function and changes in intracellular calcium transients were measured in paced (1 Hz), Fura-2 AM loaded, myocytes using the IonOptix system. Production of ROS in ventricular myocytes treated with MDMA was not different from control. In contrast, all three metabolites of MDMA exhibited time- and concentration-dependent increases in ROS that were prevented by N-acetyl-cysteine (NAC). The metabolites of MDMA, but not MDMA alone, significantly decreased contractility and impaired relaxation in myocytes stimulated at 1 Hz. These effects were prevented by NAC. Together, these data suggest that MDMA-induced oxidative stress in the left ventricle can be due, at least in part, to the metabolism of MDMA to redox active metabolites.


Subject(s)
Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Deoxyepinephrine/analogs & derivatives , Deoxyepinephrine/toxicity , Dose-Response Relationship, Drug , Glutathione/analogs & derivatives , Glutathione/toxicity , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Myocytes, Cardiac/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors
18.
J Recept Signal Transduct Res ; 28(4): 429-36, 2008.
Article in English | MEDLINE | ID: mdl-18702013

ABSTRACT

Gamma-hydroxybutyrate (GHB) and its metabolic precursor, 1,4-butanediol (BDL), are widely used recreational drugs. Although most commonly described as CNS depressants, GHB and BDL elicit significant sympathomimetic cardiovascular responses [increases in mean arterial pressure (MAP) and heart rate] when administered parenterally. Given that humans most commonly ingest both drugs orally, we examined the dose-response relationships for intragastrically administered GHB and BDL on MAP and heart rate in conscious rats using radiotelemetry. The intragastric administration of GHB increased MAP. BDL increased both MAP and heart rate and was approximately 10-fold more potent as a cardiovascular stimulant than GHB when administered intragastrically. Pretreatment with ethanol prevented the lethality of BDL. These data indicate that 1) both GHB and BDL produce cardiovascular responses when administered intragastrically and 2) BDL is more potent and potentially more dangerous than GHB when administered via this route.


Subject(s)
Blood Pressure/drug effects , Butylene Glycols/pharmacology , Heart Rate/drug effects , Sodium Oxybate/pharmacology , Animals , Butylene Glycols/administration & dosage , Drug Administration Routes , Rats , Rats, Sprague-Dawley , Sodium Oxybate/administration & dosage
19.
Cardiovasc Res ; 79(4): 662-70, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18495670

ABSTRACT

AIMS: Our aim was to determine whether the repeated, binge administration of 3,4-methylenedioxymethamphetamine (ecstasy; MDMA) produces structural and/or functional changes in the myocardium that are associated with oxidative stress. METHODS AND RESULTS: Echocardiography and pressure-volume conductance catheters were used to assess left ventricular (LV) structure and function in rats subjected to four ecstasy binges (9 mg/kg i.v. for 4 days, separated by a 10 day drug-free period). Hearts from treated and control rats were used for either biochemical and proteomic analysis or the isolation of adult LV myocytes. After the fourth binge, treated hearts showed eccentric LV dilation and diastolic dysfunction. Systolic function was not altered in vivo; however, the magnitude of the contractile responses to electrical stimulation was significantly smaller in myocytes from rats treated in vivo with ecstasy compared with myocytes from control rats. The magnitude of the peak increase in intracellular calcium (measured by Fura-2) was also significantly smaller in myocytes from ecstasy-treated vs. control rats. The relaxation kinetics of the intracellular calcium transients were significantly longer in myocytes from ecstasy-treated rats. Ecstasy significantly increased nitrotyrosine content in the left ventricle. Proteomic analysis revealed increased nitration of contractile proteins (troponin-T, tropomyosin alpha-1 chain, myosin light polypeptide, and myosin regulatory light chain), mitochondrial proteins (Ub-cytochrome-c reductase and ATP synthase), and sarcoplasmic reticulum calcium ATPase. CONCLUSION: The repeated binge administration of ecstasy produces eccentric LV dilation and dysfunction that is accompanied by oxidative stress. These functional responses may result from the redox modification of proteins involved in excitation-contraction coupling and/or mitochondrial energy production. Together, these results indicate that ecstasy has the potential to produce serious cardiac toxicity and ventricular dysfunction.


Subject(s)
Hallucinogens/toxicity , Myocytes, Cardiac/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Oxidative Stress/drug effects , Ventricular Dysfunction, Left/chemically induced , Animals , Calcium Signaling/drug effects , Cells, Cultured , Diastole , Dilatation, Pathologic , Electric Stimulation , Hallucinogens/administration & dosage , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Injections, Intravenous , Kinetics , Male , Muscle Proteins/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Rats , Rats, Sprague-Dawley , Systole , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
20.
Endocrinology ; 149(8): 3842-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450958

ABSTRACT

Activator of G protein signaling (AGS)-3 plays functional roles in cell division, synaptic plasticity, addictive behavior, and neuronal development. As part of a broad effort to define the extent of functional diversity of AGS3-regulated-events in vivo, we generated AGS3 null mice. Surprisingly, AGS3 null adult mice exhibited unexpected alterations in cardiovascular and metabolic functions without any obvious changes in motor skills, basic behavioral traits, and brain morphology. AGS3 null mice exhibited a lean phenotype, reduced fat mass, and increased nocturnal energy expenditure. AGS3 null mice also exhibited altered blood pressure control mechanisms. These studies expand the functional repertoire for AGS3 and other G protein regulatory proteins providing unexpected mechanisms by which G protein systems may be targeted to influence obesity and cardiovascular function.


Subject(s)
Cardiovascular Physiological Phenomena , Carrier Proteins/genetics , Carrier Proteins/physiology , Metabolism/genetics , Adipose Tissue/metabolism , Animals , Body Composition/genetics , Brain/anatomy & histology , COS Cells , Chlorocebus aethiops , Crosses, Genetic , Energy Metabolism/genetics , Female , Guanine Nucleotide Dissociation Inhibitors , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...