Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Chem Biol Interact ; 366: 110112, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36029803

ABSTRACT

Abuse of anabolic-androgenic steroids (AAS) is associated with neurological and cognitive problems in athletes. The Purpose of this study was to investigate the simultaneous effect of resistance training (RT) and spirulina supplementation (Sp) on the function of the antioxidant system with emphasis on mir125b, mir146a and cognitive function in Stanazolol (S)-induced neurotoxicity in rats. This experimental animal model study was performed with a post-test design with a control group. 45 male Sprague-Dawley rats were divided into six groups of 9 animals including (Althobaiti et al., 2022) [1]: sham (Sh/normal saline intake) (Havnes et al., 2019) [2], 25 mg/kg/wk of stanazolol (S) (Albano et al., 2021) [3], S + 100 mg/kg of Sp + (S + Sp) (Bjørnebekk et al., 2021) [4], RT (six weeks with an intensity of 50-100% of body weight) + S (S + RT) (Kanayama et al., 2013) [5] S + Sp + RT. Levels of superoxide dismutase (SOD), glutathione peroxidase (GPx), total antioxidant capacity (TAC), malondialdehyde (MDA), percentage of healthy cells in the C1 and C3 regions of hippocampus, miR125b, miR146a, step-through latency (STL), time spent in dark compartment (TDC), repeated entry in dark compartment (RDC) and percentage of alternation (PA%) were measured in the post-test. Results showed that the Sp, RT and SP + RT increased levels of SOD, GPx and percentage of healthy cells in C1 region, decreased MDA, mir125b, mir146a in hippocampal tissue and decreased TDC levels in S-exposed rats (P ≤ 0.05). Sp + RT decreased RDC and increased SOD levels; on the other hand, RT decreased RDC levels in S-exposed rats (P ≤ 0.05). Levels of TAC in the Sp groups were significantly higher than the S group (P ≤ 0.05). Also, the effect of Sp + RT in reducing miR125b, miR146a, and STL levels was much higher than the effect of Sp and RT alone (P ≤ 0.05). It seems that applying resistance training and spirulina supplementation both separately and interactively is effective in improving the antioxidant system as well as memory and learning in cognitive impairment caused by stanazolol. However, more studies on microRNAs are needed.


Subject(s)
MicroRNAs , Neurotoxicity Syndromes , Resistance Training , Spirulina , Animals , Antioxidants/metabolism , Cognition , Glutathione Peroxidase/metabolism , Humans , Male , Malondialdehyde , MicroRNAs/genetics , MicroRNAs/pharmacology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Saline Solution/pharmacology , Spirulina/metabolism , Stanozolol/pharmacology , Superoxide Dismutase/metabolism
2.
Steroids ; 182: 109000, 2022 06.
Article in English | MEDLINE | ID: mdl-35283118

ABSTRACT

Androgenic-Anabolic Steroids (AAS) consumption may have irreversible effects on athletes' hearts. The beneficial effects of Tribulus Terrestris (TT) have been shown to reduce cardiovascular risks through disruption in apoptosome complex construction. Therefore, this study aimed to investigate the effect of eight weeks of resistance training (RT) with TT consumption in the heart tissue of rats exposed to Stanozolol. Thirty-five male rats were divided into seven groups, Control group, Stanozolol (ST), ST + 100 mg/kg TT, ST + 50 mg/kg TT, RT + ST, RT + ST + 100 mg/kg TT, and RT + ST + 50 mg/kg TT. Differential genes expression was measured by q-RT-PCR. Artificial intelligence highlighted apoptosis pathways as a vital process in cardiovascular risks. Hence, we estimated the binding affinity of chemical and bioactive molecules on the cut point hub gene by pharmacophore modeling and molecular docking. Moreover, ST increased IL-6, Cat, Aif-1, and Caspase-9. 100 mg/kg TT has a more favorable effect than 50 mg/kg T. Also, RT with TT had interactive effects on reducing IL-6, Cat, Aif-1, and Caspase-9. RT and TT consumption seemed to synergistically reduce the apoptotic pathway markers in the heart tissue of rats exposed to the supra-physiologic dose of ST. Moreover, TT could be added to supplements and sports drink to increase an athlete's performance.


Subject(s)
Resistance Training , Tribulus , Animals , Artificial Intelligence , Caspase 9 , Humans , Interleukin-6 , Male , Molecular Docking Simulation , Plant Extracts/pharmacology , Rats , Stanozolol/pharmacology , Tribulus/chemistry
3.
J Steroid Biochem Mol Biol ; 210: 105863, 2021 06.
Article in English | MEDLINE | ID: mdl-33677017

ABSTRACT

The illicit use of anabolic androgenic steroids (AAS) among adolescents and young adults is a major concern due to the unknown and unpredictable impact of AAS on the developing brain and the consequences of this on mental health, cognitive function and behaviour. The present study aimed to investigate the effects of supra-physiological doses of four structurally different AAS (testosterone, nandrolone, stanozolol and trenbolone) on neurite development and cell viability using an in vitro model of immature primary rat cortical cell cultures. A high-throughput screening image-based approach, measuring the neurite length and number of neurons, was used for the analysis of neurite outgrowth. In addition, cell viability and expression of the Tubb3 gene (encoding the protein beta-III tubulin) were investigated. Testosterone, nandrolone, and trenbolone elicited adverse effects on neurite outgrowth as deduced from an observed reduced neurite length per neuron. Trenbolone was the only AAS that reduced the cell viability as indicated by a decreased number of neurons and declined mitochondrial function. Moreover, trenbolone downregulated the Tubb3 mRNA expression. The adverse impact on neurite development was neither inhibited nor supressed by the selective androgen receptor (AR) antagonist, flutamide, suggesting that the observed effects result from another mechanism or mechanisms of action that are operating apart from AR activation. The results demonstrate a possible AAS-induced detrimental effect on neuronal development and regenerative functions. An impact on these events, that are essential mechanisms for maintaining normal brain function, could possibly contribute to behavioural alterations seen in AAS users.


Subject(s)
Anabolic Agents/chemistry , Anabolic Agents/pharmacology , Cerebral Cortex/cytology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Cell Survival/drug effects , Cerebral Cortex/embryology , Dose-Response Relationship, Drug , Female , Nandrolone/chemistry , Nandrolone/pharmacology , Neurons/metabolism , Primary Cell Culture , Rats, Wistar , Receptors, Androgen/metabolism , Stanozolol/chemistry , Stanozolol/pharmacology , Testosterone/chemistry , Testosterone/pharmacology , Trenbolone Acetate/chemistry , Trenbolone Acetate/pharmacology , Tubulin/genetics
4.
Steroids ; 161: 108670, 2020 09.
Article in English | MEDLINE | ID: mdl-32473164

ABSTRACT

We investigated the adverse effects of the anabolic androgenic steroids (AAS) boldenone (BOL) and stanazolol (ST) on the enzymatic antioxidant systems of the rat liver. Male Wistar rats were divided in three protocols (P): PI, 5 mg/kg BOL or ST once a week for 4 weeks; PII, 2.5 mg/kg BOL or ST once a week for 8 weeks; PIII, 1.25 mg/kg BOL or ST once a week for 12 weeks. AAS were administered intramuscularly (0.2 ml, olive oil vehicle) once a week in all protocols. Activities of the enzymes glutathione peroxidase (GPx), glutathione S-transferase (GST), and glutathione reductase (GR), superoxide dismutase (SOD), catalase (CAT), were investigated. We assessed the content of hydrogen peroxide (H2O2), glycogen and lactate; and enzyme markers of neutrophils (myeloperoxidase, MPO) and macrophages (NAGase). PI and PII altered the SOD and CAT activities and increased the H2O2 content. PI led to increases in the MPO and NAGase activities. In contrast, changes in GPx, GST and, GR were observed under PII and, to a greater extend, under PIII. Following PIII, GPx, GR, and GST exhibited reduced activities. All protocols altered the glycogen and lactate content. The use of high doses of AAS for a short duration first alters SOD/CAT activity. In contrast, at lower doses of AAS for long periods is associated with changes in the glutathione system. Protocols with high doses of AAS for a short duration exert the most deleterious effects on redox status, markers of cellular infiltration, and the metabolic functioning of hepatic tissues.


Subject(s)
Antioxidants/metabolism , Glycogen/metabolism , Lactic Acid/metabolism , Liver/drug effects , Liver/enzymology , Stanozolol/pharmacology , Testosterone/analogs & derivatives , Acetylglucosaminidase/metabolism , Animals , Dose-Response Relationship, Drug , Liver/metabolism , Peroxidase/metabolism , Rats , Rats, Wistar , Testosterone/pharmacology , Time Factors
5.
Steroids ; 145: 19-22, 2019 05.
Article in English | MEDLINE | ID: mdl-30772437

ABSTRACT

Testosterone cypionate and Stanozolol are Anabolic-Androgenic Steroids (AAS) which are synthetic substances that possess functions similar to testosterone. The use of these substances has increased considerably among youngsters and sports practitioners aiming better performance of with aesthetic purposes. The major concern is the effects caused by the inappropriate use of the substances, such as hypertension, myocardial ischemia, and left ventricle hypertrophy. The objective of the present research was to measure the diameter of the left ventricle lumen and the thickness of the left ventricle myocardium in mice submitted to supraphysiological doses of AAS. A total of 30 female Swiss mice were used in the experiments. The animals received supraphysiological doses of the AAS for 30 days, and during the treatment period, they were put to swim in intercalated days. After treatment animals were euthanized and slides were made from the hearts for measurements. Results demonstrated that both AAS changed significantly the heart morphology: Testosterone cypionate led to an increase in the ventricular lumen and stanozolol increased left ventricle myocardium thickness. In conclusion, the use of AAS in supraphysiological doses can change the heart morphology and can lead to serious health consequences.


Subject(s)
Heart/drug effects , Physical Conditioning, Animal , Stanozolol/pharmacology , Testosterone/analogs & derivatives , Animals , Female , Heart/physiology , Mice , Testosterone/pharmacology
6.
J. appl. oral sci ; 27: e20180014, 2019. graf
Article in English | LILACS, BBO - Dentistry | ID: biblio-975888

ABSTRACT

Abstract Stanozolol (ST) is a synthetic androgen with high anabolic potential. Although it is known that androgens play a positive role in bone metabolism, ST action on bone cells has not been sufficiently tested to support its clinical use for bone augmentation procedures. Objective: This study aimed to assess the effects of ST on osteogenic activity and gene expression in SaOS-2 cells. Material and Methods: SaOS-2 deposition of mineralizing matrix in response to increasing doses of ST (0-1000 nM) was evaluated through Alizarin Red S and Calcein Green staining techniques at 6, 12 and 24 days. Gene expression of runt-related transcription factor 2 (RUNX2), vitamin D receptor (VDR), osteopontin (SPP1) and osteonectin (ON) was analyzed by RT-PCR. Results: ST significantly influenced SaOS-2 osteogenic activity: stainings showed the presence of rounded calcified nodules, which increased both in number and in size over time and depending on ST dose. RT-PCR highlighted ST modulation of genes related to osteogenic differentiation. Conclusions: This study provided encouraging results, showing ST promoted the osteogenic commitment of SaOS-2 cells. Further studies are required to validate these data in primary osteoblasts and to investigate ST molecular pathway of action.


Subject(s)
Humans , Osteogenesis/drug effects , Stanozolol/pharmacology , Gene Expression/drug effects , Anabolic Agents/pharmacology , Osteoblasts/drug effects , Time Factors , Calcification, Physiologic/drug effects , Linear Models , Osteonectin/analysis , Osteonectin/drug effects , Reproducibility of Results , Analysis of Variance , Receptors, Calcitriol/analysis , Receptors, Calcitriol/drug effects , Cell Line, Tumor/drug effects , Core Binding Factor Alpha 1 Subunit/analysis , Core Binding Factor Alpha 1 Subunit/drug effects , Osteopontin/analysis , Osteopontin/drug effects , Real-Time Polymerase Chain Reaction
7.
J Appl Oral Sci ; 27: e20180014, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30427473

ABSTRACT

Stanozolol (ST) is a synthetic androgen with high anabolic potential. Although it is known that androgens play a positive role in bone metabolism, ST action on bone cells has not been sufficiently tested to support its clinical use for bone augmentation procedures. OBJECTIVE: This study aimed to assess the effects of ST on osteogenic activity and gene expression in SaOS-2 cells. MATERIAL AND METHODS: SaOS-2 deposition of mineralizing matrix in response to increasing doses of ST (0-1000 nM) was evaluated through Alizarin Red S and Calcein Green staining techniques at 6, 12 and 24 days. Gene expression of runt-related transcription factor 2 (RUNX2), vitamin D receptor (VDR), osteopontin (SPP1) and osteonectin (ON) was analyzed by RT-PCR. RESULTS: ST significantly influenced SaOS-2 osteogenic activity: stainings showed the presence of rounded calcified nodules, which increased both in number and in size over time and depending on ST dose. RT-PCR highlighted ST modulation of genes related to osteogenic differentiation. CONCLUSIONS: This study provided encouraging results, showing ST promoted the osteogenic commitment of SaOS-2 cells. Further studies are required to validate these data in primary osteoblasts and to investigate ST molecular pathway of action.


Subject(s)
Anabolic Agents/pharmacology , Gene Expression/drug effects , Osteogenesis/drug effects , Stanozolol/pharmacology , Analysis of Variance , Calcification, Physiologic/drug effects , Cell Line, Tumor/drug effects , Core Binding Factor Alpha 1 Subunit/analysis , Core Binding Factor Alpha 1 Subunit/drug effects , Humans , Linear Models , Osteoblasts/drug effects , Osteonectin/analysis , Osteonectin/drug effects , Osteopontin/analysis , Osteopontin/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Calcitriol/analysis , Receptors, Calcitriol/drug effects , Reproducibility of Results , Time Factors
8.
J Basic Clin Physiol Pharmacol ; 30(1): 29-36, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30074896

ABSTRACT

Background Testosterone, nandrolone, and stanozolol are among the highly consumed anabolic androgenic steroids (AASs). Although the desired effects of AAS are being achieved by the abusers, unfortunately, this leads to numerous physical and physiological side effects. The present study was designed to investigate and determine whether early pubertal exposure to AAS treatment had detrimental effects on blood testosterone and estradiol concentrations, mating behavior, and pregnancy outcome during the pubertal period in male rats. Materials Early pubertal rats (PND41) were given two doses (2.5 mg/kg and 5 mg/kg) each of testosterone, nandrolone, and stanozolol subcutaneously for 6 weeks. Upon completion, three rats with the highest weight were chosen from each group for mating with the females, in a ratio of one male to two females for 10 days. After 10 days, all male rats were sacrificed to obtain the testes for weight recording, and blood samples were collected for testosterone and estradiol quantitation. Pregnant females were housed separately until birth, and the number of offsprings produced was counted. Results The results clearly show a reduction in reproductive hormonal and behavioral parameters between testosterone and nandrolone, and testosterone and stanozolol. Stanozolol administration exhibited suppressing effects in all parameters including testicular weight deterioration, serum testosterone and estradiol reduction, low mating preferences, and declined pregnancy outcome. Conclusions AAS exposure during the onset of puberty results in reproductive detrimental effects, which are postulated to affect the pregnancy rate.


Subject(s)
Androgens/pharmacology , Estradiol/blood , Pregnancy Outcome , Sexual Behavior, Animal/drug effects , Sexual Maturation/drug effects , Testosterone/blood , Animals , Female , Male , Nandrolone/pharmacology , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Sexual Behavior, Animal/physiology , Sexual Maturation/physiology , Stanozolol/pharmacology , Testosterone Congeners/pharmacology
9.
Int J Mol Med ; 42(1): 405-413, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29717770

ABSTRACT

Anabolic agents are doping substances which are commonly used in sports. Stanozolol, a 17α­alkylated derivative of testosterone, has a widespread use among athletes and bodybuilders. Several medical and behavioral adverse effects are associated with anabolic androgenic steroids (AAS) abuse, while the liver remains the most well recognized target organ. In the present study, the hepatic effects of stanozolol administration in rats at high doses resembling those used for doping purposes were investigated, in the presence or absence of exercise. Stanozolol and its metabolites, 16­ß­hydroxystanozolol and 3'­hydroxystanozolol, were detected in rat livers using liquid chromatography­mass spectrometry (LC­MS). Telomerase activity, which is involved in cellular aging and tumorigenesis, was detected by examining telomerase reverse transcriptase (TERT) and phosphatase and tensin homolog (PTEN) expression levels in the livers of stanozolol­treated rats. Stanozolol induced telomerase activity at the molecular level in the liver tissue of rats and exercise reversed this induction, reflecting possible premature liver tissue aging. PTEN gene expression in the rat livers was practically unaffected either by exercise or by stanozolol administration.


Subject(s)
Aging/physiology , Liver/physiology , Physical Conditioning, Animal , Stanozolol/administration & dosage , Stanozolol/pharmacology , Telomerase/metabolism , Animals , Gene Expression Regulation/drug effects , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Male , Metabolome/drug effects , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Rats, Sprague-Dawley , Stanozolol/analogs & derivatives , Telomere/metabolism
10.
BMC Vet Res ; 14(1): 103, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29554899

ABSTRACT

BACKGROUND: Intra-articular administration of stanozolol has shown promising results by improving the clinical management of lameness associated with naturally-occurring osteoarthritis (OA) in horses, and by decreasing osteophyte formation and subchondral bone reaction in sheep following surgically induced OA. However, there is limited evidence on the anti-inflammatory and modulatory properties of stanozolol on articular tissues. The objective of the current study was to evaluate the effects of stanozolol on chondrocyte viability and gene expression in normal equine chondrocytes and an inflammatory in vitro system of OA (interleukin-1ß (IL-1ß) treated chondrocytes). RESULTS: Chondrocytes from normal metacarpophalangeal joints of skeletally mature horses were exposed to four treatment groups: (1) media only (2) media+IL-1ß (3) media+IL-1ß + stanozolol (4) media+stanozolol. Following exposure, chondrocyte viability and the expression of catabolic, anabolic and structural genes were determined. General linear models with Dunnet's comparisons with Bonferroni's adjustment were performed. Cell viability was similar in all groups. Stanozolol treatment reduced gene expression of MMP-13, MMP-1, IL-6 and COX-2 in both normal and IL-1ß treated chondrocytes. Stanozolol treatment reduced ADAMTS4 gene expression in normal chondrocytes. Stanozolol reduced the expression of COL2A1. CONCLUSIONS: The current study demonstrates stanozolol has chondroprotective effects through downregulation of genes for pro-inflammatory/catabolic cytokines and enzymes associated with OA. However, there is no evidence of increased cartilage stimulation through upregulation of the anabolic and structural genes tested.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Interleukin-1beta/pharmacology , Stanozolol/pharmacology , Animals , Horse Diseases/drug therapy , Horses , In Vitro Techniques , Lameness, Animal/drug therapy , Osteoarthritis/drug therapy , Osteoarthritis/veterinary
11.
Steroids ; 134: 96-100, 2018 06.
Article in English | MEDLINE | ID: mdl-29477345

ABSTRACT

Stanozolol is a widely used 17α-alkylated anabolic androgenic steroid (AAS) derivative. Despite stanozolol's adverse effects, its effect on oxidative stress parameters and mitochondrial apoptosis pathway is not clearly defined. In our study, thirty four male Sprague-Dawley rats were divided into 5 groups as control (C), vehicle control (VC), steroid (ST), vehicle control-exercise (VCE), and steroid-exercise (STE). Animals were subcutaneously administered stanozolol 5 mg/kg in steroid groups and propylene glycol 1 ml/kg in the vehicle-control groups. On the 28th day-after sacrification, oxidative stress (MDA, GSH, PC, SOD, CAT) and apoptosis parameters (TUNEL, Cytochrome-c) in cardiac tissue were evaluated. Also, blood vessel morphology of cardiac tissue was evaluated with Verhoeff-van Giesen staining. It has been demonstrated that stanozolol administration triggers apoptosis by using TUNEL assay and cytochrome-c immunohistochemical staining intensity, while this effect is significantly reduced in the presence of exercise. In conclusion, the present study demonstrated that stanozolol administration induces apoptosis with increasing PC and CAT levels, while GSH, MDA and SOD parameters do not reveal any significant change. Exercise has a protective role in stanozolol induced oxidative stress and apoptosis. According to Verhoeff-van Giesen staining results for blood vessel morphology assessment, it has been seen that exercise has a protective role on cardiac blood vessels. This mechanism needs further investigations with long term exposure studies for clarifying possible pathways.


Subject(s)
Apoptosis/drug effects , Myocardium/cytology , Myocardium/metabolism , Stanozolol/pharmacology , Animals , Biomarkers/metabolism , Blood Vessels/drug effects , Blood Vessels/metabolism , Male , Oxidative Stress , Physical Conditioning, Animal , Rats , Rats, Sprague-Dawley
12.
J Steroid Biochem Mol Biol ; 174: 265-275, 2017 11.
Article in English | MEDLINE | ID: mdl-29030156

ABSTRACT

A robust LC-MS/MS method was developed to quantify a large number of phase I and phase II steroids in urine. The decision limit is for most compounds lower than 1ngml-1 with a measurement uncertainty smaller than 30%. The method is fully validated and was applied to assess the influence of administered synthetic steroids and beta-agonists on the steroidogenesis. From three animal experiments, clenbuterol, diethylstilbestrol and stanozolol, the steroid profiles in urine of bovine animals were compared before and after treatment. It was demonstrated that the steroid profiles were altered due to these treatments. A predictive multivariate model was built to identify deviations from normal population steroid profiles. The abuse of synthetic steroids can be detected in urine samples from bovine animals using this model. The samples from the animal experiments were randomly analysed using this method and predictive model. It was shown that these samples were predicted correctly in the exogenous steroids group.


Subject(s)
Anabolic Agents/pharmacology , Clenbuterol/pharmacology , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Stanozolol/pharmacology , Steroids/urine , Animals , Cattle/urine , Chromatography, High Pressure Liquid , Male , Tandem Mass Spectrometry
13.
PLoS One ; 12(6): e0177623, 2017.
Article in English | MEDLINE | ID: mdl-28594925

ABSTRACT

The aim of this study was to assess if the dose and exposure duration of the anabolic androgenic steroids (AAS) boldenone (BOL) and stanazolol (ST) affected memory, anxiety, and social interaction, as well as acetylcholinesterase (AChE) activity and oxidative stress in the cerebral cortex (CC) and hippocampus (HC). Male Wistar rats (90 animals) were randomly assigned to three treatment protocols: (I) 5 mg/kg BOL or ST, once a week for 4 weeks; (II) 2.5 mg/kg BOL or ST, once a week for 8 weeks; and (III) 1.25 mg/kg BOL or ST, once a week for 12 weeks. Each treatment protocol included a control group that received an olive oil injection (vehicle control) and AAS were administered intramuscularly (a total volume of 0.2 ml) once a week in all three treatment protocols. In the BOL and ST groups, a higher anxiety level was observed only for Protocol I. BOL and ST significantly affected social interaction in all protocols. Memory deficits and increased AChE activity in the CC and HC were found in the BOL groups treated according to Protocol III only. In addition, BOL and ST significantly increased oxidative stress in both the CC and HC in the groups treated according to Protocol I and III. In conclusion, our findings show that the impact of BOL and ST on memory, anxiety, and social interaction depends on the dose and exposure duration of these AAS.


Subject(s)
Acetylcholinesterase/metabolism , Anabolic Agents/pharmacology , Androgens/pharmacology , Behavior, Animal/drug effects , Oxidative Stress/drug effects , Aggression/drug effects , Anabolic Agents/administration & dosage , Androgens/administration & dosage , Animals , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/pathology , Male , Rats, Wistar , Stanozolol/administration & dosage , Stanozolol/pharmacology , Territoriality , Testosterone/administration & dosage , Testosterone/analogs & derivatives , Testosterone/pharmacology
14.
Int J Sports Med ; 36(12): 984-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26252547

ABSTRACT

We analyzed the effects of high-intensity exercise (HIE) and anabolic androgenic steroids (AAS) on brain redox status. 40 male Wistar rats were randomly distributed in 4 experimental groups (n=10) with or without HIE and with or without weekly Stanozolol administration. Thiobarbituric acid-reactive substances (TBARs) and protein carbonyl content (PCC) were assessed. Total superoxide dismutase (tSOD), manganese superoxide dismutase (Mn-SOD), copper/zinc superoxide dismutase (CuZn-SOD) and catalase (CAT) activities were measured. Finally, protein expression levels of glutathione peroxidase (GPx), NAD(P)H dehydrogenase, Quinone 1 (NQO1), NF-E2-Related Factor 2 (Nrf2), glial fibrillary acidic protein (GFAP), nuclear factor kappa ß p65 (NF-κß) and signal transducer and activator of transcription 3 were determined. Brain PCC concentrations were lower in the HIE groups compared to the untrained controls, whereas CAT activity was higher (both, p<0.01). Both HIE and AAS groups exhibited higher expression levels of GFAP and GPx, but lower NQO1 levels (all, p<0.05). There were increased expression levels of NF-κß in the AAS groups (p<0.01). In addition, there was increased expression of Nrf2 in the HIE groups (p<0.001). HIE*AAS interactions were found on TBARs content and GFAP expression, with HIE downregulating and upregulating AAS-mediated increases in TBARs and GFAP, respectively (p<0.05). Overall, HIE appeared to reduce the AAS-mediated negative effect on brain redox status.


Subject(s)
Anabolic Agents/pharmacology , Brain/metabolism , Oxidative Stress/drug effects , Physical Conditioning, Animal/methods , Stanozolol/pharmacology , Animals , Biomarkers/metabolism , Body Weight , Brain/anatomy & histology , Brain/enzymology , Eating , Male , Organ Size , Protein Carbonylation , Random Allocation , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
15.
PLoS One ; 9(2): e87106, 2014.
Article in English | MEDLINE | ID: mdl-24533053

ABSTRACT

Elevated concentrations of testosterone and its synthetic analogs may induce changes in cardiovascular function. However, the effects of the combination of anabolic/androgenic steroid (AAS) treatment and exercise training on systolic and diastolic cardiac function are poorly understood. In the present study, we aimed to investigate the effects of low-dose steroid treatment (stanozolol) on cardiac contractile parameters when this steroid treatment was combined with exercise training in rats and the effects of chronic steroid treatment on the Frank-Starling (length-tension curves) relationship. Male Wistar rats were randomly assigned to one of four groups: U (untrained), US (untrained and treated with stanozolol 5 mg/kg/week), T (trained, 16 m/min/1 h) and TS (trained and treated with stanozolol 5 mg/kg/week). Continuous exercise training was conducted 5 days/week for 8 consecutive weeks. The speed of the treadmill was gradually increased to a final setting of 16 m/min/1 h. Experiments were divided into two independent series: 1) central hemodynamic analysis for mean arterial blood pressure (MAP) and cardiac output (CO) measurements and 2) isolated papillary muscle preparation in Krebs solution. Stanozolol treatment significantly increased the MAP and the heart size in untrained and trained rats (U 113±2; T 106±2; US 138±8 and TS 130±7 mmHg). Furthermore, stanozolol significantly decreased developed tension and dT/dt (maximal and minimal) in U rats. However, the developed tension was completely restored by training. The Frank/Starling relationship was impaired in rats treated with stanozolol; however, again, training completely restored diastolic function. Taken together, the present data suggest that AAS treatment is able to decrease cardiac performance (systolic and diastolic functions). The combination of stanozolol and physical training improved cardiac performance, including diastolic and systolic functions, independent of changes in central hemodynamic parameters. Therefore, changes in ventricular myocyte calcium transients may play a cardioprotective role.


Subject(s)
Anabolic Agents/pharmacology , Exercise Test/methods , Heart/drug effects , Myocardium/pathology , Stanozolol/pharmacology , Animals , Blood Pressure , Cardiac Output , Heart Rate/drug effects , Heart Ventricles/drug effects , Hematocrit , Hemodynamics/drug effects , Male , Myocardial Contraction/drug effects , Organ Size/drug effects , Physical Conditioning, Animal , Rats , Rats, Wistar , Steroids/pharmacology
16.
Mol Cell Endocrinol ; 363(1-2): 100-10, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-22906881

ABSTRACT

Several doping agents, such as anabolic androgenic steroids (AAS) and peptide hormones like insulin-like growth factor-I (IGF-I), are employed without considering the potential deleterious effects that they can cause. In addition, androgens are used in postmenopausal women as replacement therapy. However, there are no clear guidelines regarding the optimal therapeutic doses of androgens or long-term safety data. In this study we aimed to determine if two commonly used AAS, nandrolone and stanozolol, alone or in combination with IGF-I, could activate signaling involved in breast cancer cell proliferation. Using a human breast cancer cell line, MCF-7, as an experimental model we found that both nandrolone and stanozolol caused a dose-dependent induction of aromatase expression and, consequently, estradiol production. Moreover, when nandrolone and stanozolol were combined with IGF-I, higher induction in aromatase expression was observed. This increase involved phosphatidylinositol 3-kinase (PI3K)/AKT and phospholipase C (PLC)/protein kinase C (PKC), which are part of IGF-I transductional pathways. Specifically, both AAS were able to activate membrane rapid signaling involving IGF-I receptor, extracellular regulated protein kinases 1/2 (ERK1/2) and AKT, after binding to estrogen receptor (ER), as confirmed by the ability of the ER antagonist ICI182, 780 to block such activation. The estrogenic activity of nandrolone and stanozolol was further confirmed by their capacity to induce the expression of the ER-regulated gene, CCND1 encoding for the cell cycle regulator cyclin D1, which represents a key protein for the control of breast cancer cell proliferation. In fact, when nandrolone and stanozolol were combined with IGF-I, they increased cell proliferation to levels higher than those elicited by the single factors. Taken together these data clearly indicate that the use of high doses of AAS, as occurs in doping practice, may increase the risk of breast cancer. This potential risk is higher when AAS are used in association with IGF-I. To our knowledge this is the first report directly associating AAS with this type of cancer.


Subject(s)
Anabolic Agents/pharmacology , Androgens/pharmacology , Aromatase/metabolism , Cell Proliferation/drug effects , Enzyme Induction/drug effects , Insulin-Like Growth Factor I/physiology , Nandrolone/pharmacology , Stanozolol/pharmacology , Aromatase/genetics , Breast Neoplasms , Cell Survival/drug effects , Drug Synergism , Estradiol/biosynthesis , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Genes, Reporter , Humans , Insulin-Like Growth Factor I/pharmacology , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism , Up-Regulation/drug effects
17.
J Steroid Biochem Mol Biol ; 129(3-5): 172-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22133647

ABSTRACT

The abuse of anabolic-androgenic steroids (AAS) for improved physical performance is associated with many deleterious effects. The present study aims to evaluate the short-term effect of an AAS compound stanozolol, on lipoprotein profile, granulopoiesis and immune response in adult female mice. The mice were assigned to five experimental groups and different doses of stanozolol (low - 0.05 mg, medium - 0.5 mg, high - 5 mg and highest dose - 7.5 mg/kg bwt or only vehicle respectively) were administered s.c. for 15 days. A decrease in high density lipoprotein cholesterol (HDL-c) as well as total cholesterol (TC) in all the stanozolol treated groups and an increase in low density lipoprotein (LDL-c) in high and the highest dose treated groups indicate that stanozolol alters serum lipoprotein profile. A significant increase in the percentage of myelocytes, metamyelocytes and neutrophils in all the treated mice unveils the stimulation of granulopoiesis through the acceleration of neutrophil precursors' maturation in the bone marrow of mice. The stimulation of erythropoiesis was also noted in all the treated groups. The flow cytometry analysis of lymphocyte subpopulations (CD3(+) and CD4(+)) revealed immunoenhancing response of stanozolol at optimum physiological dose, however, it is immunosuppressive at supraphysiologic level. We conclude that stanozolol accelerates granulopoiesis and stimulates immune response (at physiologic level only), though it alters the lipoprotein profile in mice.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Anabolic Agents/administration & dosage , Anabolic Agents/pharmacology , Lipoproteins/blood , Stanozolol/administration & dosage , Stanozolol/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow/ultrastructure , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Female , Granulocytes/cytology , Granulocytes/drug effects , Lipids/blood , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice
18.
J Cell Physiol ; 227(5): 2079-88, 2012 May.
Article in English | MEDLINE | ID: mdl-21769864

ABSTRACT

Several substances such as anabolic androgenic steroids (AAS), peptide hormones like insulin-like growth factor-I (IGF-I), aromatase inhibitors and estrogen antagonists are offered via the Internet, and are assumed without considering the potential deleterious effects that can be caused by their administration. In this study we aimed to determine if nandrolone and stanozolol, two commonly used AAS, could have an effect on Leydig cell tumor proliferation and if their effects could be potentiated by the concomitant use of IGF-I. Using a rat Leydig tumor cell line, R2C cells, as experimental model we found that nandrolone and stanozolol caused a dose-dependent induction of aromatase expression and estradiol (E2) production. When used in combination with IGF-I they were more effective than single molecules in inducing aromatase expression. AAS exhibited estrogenic activity and induced rapid estrogen receptor (ER)-dependent pathways involving IGF1R, AKT, and ERK1/2 phosphorylation. Inhibitors for these kinases decreased AAS-dependent aromatase expression. Up-regulated aromatase levels and related E2 production increased cell proliferation as a consequence of increased cyclin E expression. The observation that ER antagonist ICI182,780 was also able to significantly reduce ASS- and AAS + IGF-induced cell proliferation, confirmed a role for estrogens in AAS-dependent proliferative effects. Taken together these data clearly indicate that the use of high doses of AAS, as it occurs in doping practice, enhances Leydig cell proliferation, increasing the risk of tumor development. This risk is higher when AAS are used in association with IGF-I. To our knowledge this is the first report directly associating AAS and testicular cancer.


Subject(s)
Androgens/pharmacology , Cell Proliferation/drug effects , Estrogens/metabolism , Insulin-Like Growth Factor I/pharmacology , Leydig Cell Tumor/pathology , Nandrolone/pharmacology , Stanozolol/pharmacology , Testicular Neoplasms/pathology , Anabolic Agents/adverse effects , Anabolic Agents/pharmacology , Androgens/adverse effects , Animals , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Leydig Cell Tumor/chemically induced , Male , Nandrolone/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Stanozolol/adverse effects , Testicular Neoplasms/chemically induced
19.
Steroids ; 77(3): 269-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22197661

ABSTRACT

An extensive literature has documented adverse effects on mental health in anabolic androgenic steroids (AAS) abusers. Depression seems a common adverse reaction in AAS abusers. Recently it has been reported that in a rat model of AAS abuse stanozolol induces behavioural and biochemical changes related to the pathophysiology of major depressive disorder. In the present study, we used the model of AAS abuse to examine possible changes in the monoaminergic system, a neurobiological substrate of depression, in different brain areas of stanozolol-treated animals. Wistar rats received repeated injections of stanozolol (5mg/kg, s.c.), or vehicle (propylene glycol, 1ml/kg) once daily for 4weeks. Twenty-four hours after last injection, changes of dopamine (DA) and relative metabolite levels, homovanilic acid (HVA) and 3,4-dihydroxy phenylacetic acid (DOPAC), serotonin (5-HT) and its metabolite levels, 5-hydroxy indolacetic acid (5-HIAA), and noradrenaline (NA) amount were investigated in prefrontal cortex (PFC), nucleus accumbens (NAC), striatum (STR) and hippocampus (HIPP). The analysis of data showed that after chronic stanozolol, DA levels were increased in the HIPP and decreased in the PFC. No significant changes were observed in the STR or in the NAC. 5-HT and 5-HIAA levels were decreased in all brain areas investigated after stanozolol exposure; however, the 5-HIAA/5-HT ratio was not altered. Taken together, our data indicate that chronic use of stanozolol significantly affects brain monoamines leading to neurochemical modifications possibly involved in depression and stress-related states.


Subject(s)
Hippocampus/drug effects , Prefrontal Cortex/drug effects , Stanozolol/pharmacology , 3,4-Dihydroxyphenylacetic Acid/analysis , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Depression/chemically induced , Depression/physiopathology , Dopamine/analysis , Hippocampus/metabolism , Homovanillic Acid/analysis , Hydroxyindoleacetic Acid/analysis , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Serotonin/analysis , Stanozolol/administration & dosage , Stanozolol/adverse effects , Substance-Related Disorders/physiopathology
20.
J Pediatr Endocrinol Metab ; 24(5-6): 275-81, 2011.
Article in English | MEDLINE | ID: mdl-21823523

ABSTRACT

Improving the final adult height is one of the most important aims for treatment of central precocious puberty. Stanozolol (ST) is a synthetic derivative of androgen. In this study, we investigated the effects and the mechanisms of ST on the proliferation of growth plate chondrocytes isolated from adolescent rats treated with gonadotropin-releasing hormone analogue (GnRHa). Treatment with ST resulted in time- and concentration-dependent effects on proliferation as determined by MTT and proliferating cell nuclear antigen (PCNA) assays. Western blotting showed that ST increased the phosphorylation level of the estrogen receptor alpha (ERalpha), but not the androgen receptor (AR). Pharmacological inhibition of ERalpha and mitogen-activated protein kinase (MAPK) attenuated the effects of ST on the proliferation of growth plate chondrocytes. A molecular dynamics simulation showed hydrophobic interactions between ST and ERalpha. These results suggested that ERalpha, but not AR, partially mediates the ST-driven proliferation of growth plate chondrocytes, and that multiple pathways may be involved in the mechanism of action of ST.


Subject(s)
Chondrocytes/drug effects , Estrogen Receptor alpha/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Stanozolol/pharmacology , Animals , Binding Sites , Body Height/drug effects , Cell Proliferation/drug effects , Child , Chondrocytes/cytology , Chondrocytes/metabolism , Disease Models, Animal , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/chemistry , Female , Gonadotropin-Releasing Hormone/pharmacology , Growth Plate/cytology , Growth Plate/drug effects , Growth Plate/metabolism , Humans , MAP Kinase Signaling System/drug effects , Male , Molecular Dynamics Simulation , Phosphorylation , Puberty, Precocious/drug therapy , Puberty, Precocious/metabolism , Puberty, Precocious/pathology , Rats , Receptors, Androgen/metabolism , Stanozolol/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...