Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Environ Sci Pollut Res Int ; 30(9): 22913-22928, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36307569

RESUMEN

Contaminants of emerging concern have been increasingly associated with the modulation of the epigenome, leading to potentially inherited and persistent impacts on apical endpoints. Here, we address the performance of the OECD Test No. 236 FET (fish embryo acute toxicity) in the identification of chemicals able to modulate the epigenome. Using zebrafish (Danio rerio) embryos, acute and chronic exposures were performed with the pharmaceutical, simvastatin (SIM), a widely prescribed hypocholesterolemic drug reported to induce inter and transgenerational effects. In the present study, the epigenetic effects of environmentally relevant concentrations of SIM (from 8 ng/L to 2000 ng/L) were addressed following (1) an acute embryo assay based on OECD Test No. 236 FET, (2) a chronic partial life-cycle exposure using adult zebrafish (90 days), and (3) F1 embryos obtained from parental exposed animals. Simvastatin induced significant effects in gene expression of key epigenetic biomarkers (DNA methylation and histone acetylation/deacetylation) in the gonads of exposed adult zebrafish and in 80 hpf zebrafish embryos (acute and chronic parental intergenerational exposure), albeit with distinct effect profiles between biological samples. In the chronic exposure, SIM impacted particularly DNA methyltransferase genes in males and female gonads, whereas in F1 embryos SIM affected mostly genes associated with histone acetylation/deacetylation. In the embryo acute direct exposure, SIM modulated the expression of both genes involved in DNA methylation and histone deacetylase. These findings further support the use of epigenetic biomarkers in zebrafish embryos in a high throughput approach to identify and prioritize epigenome-modulating chemicals.


Asunto(s)
Simvastatina , Contaminantes Químicos del Agua , Masculino , Animales , Femenino , Simvastatina/toxicidad , Pez Cebra/genética , Epigenoma , Histonas , Bioensayo , Contaminantes Químicos del Agua/toxicidad , Embrión no Mamífero
2.
Aquat Toxicol ; 244: 106095, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35121565

RESUMEN

The primary focus of environmental toxicological studies is to address the direct effects of chemicals on exposed organisms (parental generation - F0), mostly overlooking effects on subsequent non-exposed generations (F1 and F2 - intergenerational and F3 transgenerational, respectively). Here, we addressed the effects of simvastatin (SIM), one of the most widely prescribed human pharmaceuticals for the primary treatment of hypercholesterolemia, using the keystone crustacean Gammarus locusta. We demonstrate that SIM, at environmentally relevant concentrations, has significant inter and transgenerational (F1 and F3) effects in key signaling pathways involved in crustaceans' neuroendocrine regulation (Ecdysteroids, Catecholamines, NO/cGMP/PKG, GABAergic and Cholinergic signaling pathways), concomitantly with changes in apical endpoints, such as depressed reproduction and growth. These findings are an essential step to improve hazard and risk assessment of biological active compounds, such as SIM, and highlight the importance of studying the transgenerational effects of environmental chemicals in animals' neuroendocrine regulation.


Asunto(s)
Anfípodos , Contaminantes Químicos del Agua , Animales , Reproducción , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad
3.
J Proteomics ; 254: 104477, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34990819

RESUMEN

Statin-associated muscle symptoms (SAMS) are the main side effects of statins. Currently, there are no effective biomarkers for accurate clinical diagnosis. Urine is not subject to homeostatic control and therefore accumulates early changes, making it an ideal biomarker source. We therefore examined urine proteome changes associated with SAMS. Here, we established a SAMS rat model by intragastric intubation with simvastatin (80 mg/kg). Biochemical analyses and hematoxylin and eosin staining were used to evaluate the degree of muscle injury. The urine proteome on days 3, 6, 9 and 14 was profiled using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Differential proteins on day 14 of SAMS were mainly associated with glycolysis/gluconeogenesis, pyruvate metabolism, metabolism of reactive oxygen species and apoptosis, which were associated with the pathological mechanism of SAMS. Among the 14 differential proteins on day 3, Fibrinogen gamma chain (FIBG), Osteopontin (OSTP) and C-reactive protein (CRP) were associated with muscle damage, while EH domain-containing protein 1(EHD1), Cubilin (CUBN) and Fibronectin (FINC) were associated with the pathogenic mechanisms of SAMS. Our preliminary results indicated that the urine proteome can reflect early changes in the SAMS rat model, providing the potential for monitoring drug side effects in future clinical research. SIGNIFICANCE: This study demonstrate that the early muscle damage caused by simvastatin can be reflected in urinary proteins. The urine proteome also has the potential to reflect the pharmacology and toxicology of drugs in future clinical research.


Asunto(s)
Proteoma , Simvastatina , Animales , Biomarcadores , Cromatografía Liquida , Músculo Esquelético/química , Proteoma/análisis , Ratas , Simvastatina/toxicidad , Espectrometría de Masas en Tándem , Proteínas de Transporte Vesicular
4.
Pak J Pharm Sci ; 34(5(Supplementary)): 1939-1944, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34836863

RESUMEN

Type 2 Diabetes Mellitus (T2DM) patients are at high risk of Coronary Heart Disease (CHD) and need a global therapeutic intervention. A fixed-dose combination prescription medication containing anti-diabetic drug (Sitagliptin) and lipid lowering (Simvastatin) has recently been approved. Present study was designed to explore the potential synergistic toxic effects of sitagliptin and simvastatin at cellular level. MTT assay revealed the potential synergistic cytotoxic effect whereas Comet assay spotlighted the genotoxicity. MTT assay conducted on Vero cell lines revealed no significant change in proliferative activity upon treatment with simvastatin but cell survival percentage (CSP) decreased upon treatment with sitagliptin (51% at 1000µg/mL). However, combination of both drugs exhibited a better survival percentage except highest dose combination (1000:500µg/mL) which augmented antiproliferative effects rendering CSP 71.6%. The genotoxic assay spotted that Simvastatin produced less damage to DNA with the threshold of 500µg/ml whereas Sitagliptin significantly damage above the 250µg/mL, However, combination of drugs produced lesser damage than Sitagliptin alone. The findings concluded a non-genotoxic combination of sitagliptin and simvastatin which possess a least cytotoxic potential suggesting the safe use of the combination both in T2DM and CHD.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Hipoglucemiantes/toxicidad , Mutágenos/toxicidad , Simvastatina/toxicidad , Fosfato de Sitagliptina/toxicidad , Animales , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Ensayo Cometa , Daño del ADN , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Combinación de Medicamentos , Interacciones Farmacológicas , Sinergismo Farmacológico , Células Vero
5.
Aquat Toxicol ; 239: 105951, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34467877

RESUMEN

Simvastatin (SV) is a common hypolipidemic drug in clinical medicine that can reduce endogenous cholesterol biosynthesis by inhibiting hydroxyl-methyl-glutaryl coenzyme A reductase. SV took a large market share in the lipid-lowering drugs and it is frequently detected in various water bodies due to its increasing consumption in past years. In the present investigation, we selected a native fish species in the Pearl River Basin in China, Mugilogobius abei (M. abei), to study the effects of SV on non-target aquatic organisms. Results showed that a significant decrease in the volume of adipocytes under SV exposure were observed on oil red O section, and the expression of HMG-CoAR decreased significantly. The mRNA and protein expression of PPARα were significantly up-regulated, the expressions of other genes related to lipid metabolism were up-regulated to varying degrees as well. There was a positive correlation between the concentrations of SV and the protein expressions of plasma phospholipid transfer protein (PLTP) and cholesterolester transfer protein (CETP). In addition, the frozen sections showed that SV led to ROS accumulation in liver in a time and concentration dependent manner. The mRNA and protein expressions of Nrf2 were significantly up-regulated after 24 hours of SV exposure. Some biomarkers associated with antioxidant such as Trx2, TrxR and MDA content were positively correlated with the exposure concentration and time, while the content of GSH decreased sharply. It is noteworthy that the environmentally relevant concentration (0.5 µg/L) of SV exposure caused delayed embryonic development and deformations, decreased hatching rates. We conclude that SV promotes fat metabolism, gives rise to oxidative stress and has significant toxicity on embryo development in M. abei.


Asunto(s)
Simvastatina , Contaminantes Químicos del Agua , Animales , Desarrollo Embrionario , Estrés Oxidativo , PPAR alfa/genética , Transducción de Señal , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad
6.
Biochem Pharmacol ; 192: 114750, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34461118

RESUMEN

Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.


Asunto(s)
Diana Mecanicista del Complejo 2 de la Rapamicina/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Prenilación/efectos de los fármacos , Simvastatina/toxicidad , Proteínas de Unión al GTP rap1/antagonistas & inhibidores , Animales , Línea Celular , Sistemas de Liberación de Medicamentos/métodos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Prenilación/fisiología , Simvastatina/administración & dosificación , Proteínas de Unión al GTP rap1/metabolismo
7.
Ecotoxicol Environ Saf ; 209: 111849, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33387775

RESUMEN

The adverse effects of certain environmental chemicals have been recently associated with the modulation of the epigenome. Although changes in the epigenetic signature have yet to be integrated into hazard and risk assessment, they are interesting candidates to link environmental exposures and altered phenotypes, since these changes may be passed across multiple non-exposed generations. Here, we addressed the effects of simvastatin (SIM), one of the most prescribed pharmaceuticals in the world, on epigenetic regulation using the amphipod Gammarus locusta as a proxy, to support its integration into hazard and environmental risk assessment. SIM is a known modulator of the epigenome in mammalian cell lines and has been reported to impact G. locusta ecological endpoints at environmentally relevant levels. G. locusta juveniles were exposed to three SIM environmentally relevant concentrations (0.32, 1.6 and 8 µg L-1) for 15 days. Gene transcription levels of selected epigenetic regulators, i.e., dnmt1, dmap1, usp7, kat5 and uhrf1 were assessed, along with the quantification of DNA methylation levels and evaluation of key ecological endpoints: survival and growth. Exposure to 0.32 and 8 µg L-1 SIM induced significant downregulation of DNA methyltransferase 1 (dnmt1), concomitant with global DNA hypomethylation and growth impacts. Overall, this work is the first to validate the basal expression of key epigenetic regulators in a keystone marine crustacean, supporting the integration of epigenetic biomarkers into hazard assessment frameworks.


Asunto(s)
Anfípodos/fisiología , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Anfípodos/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/farmacología , Metilación de ADN , Exposición a Riesgos Ambientales , Epigénesis Genética , Preparaciones Farmacéuticas , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/farmacología , Peptidasa Específica de Ubiquitina 7/metabolismo
8.
Environ Toxicol Pharmacol ; 81: 103522, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33144098

RESUMEN

Due to their wide use, pharmaceuticals can be discarded, metabolized and excreted into the environment, potentially affecting aquatic organisms. Lipid-regulating drugs are among the most prescribed medications around the world, to control human cholesterol levels, in more than 20 million patients. Despite this massive use of lipid-regulating drugs, particularly simvastatin, the role of these drugs is not fully characterized and understood in terms of its potential toxicological effects at the environmental level. This work intended to characterize the toxicity of an acute (120 h post-fertilization) and chronic (60 days) exposure to the antihyperlipidemic drug simvastatin (in concentrations of 92.45, 184.9, 369.8, 739.6 and 1479.2 ng L-1), in the freshwater species zebrafish (Danio rerio). The concentrations hereby mentioned were implemented in both exposures, and were based on levels found in wastewater treatment plant influents (11.7 ± 3.2 µg L-1), effluents (2.65 ± 0.8 µg L-1) and Apies River (1.585 ± 0.3 µg L-1), located in Pretoria, South Africa and, particularly in the maximum levels found in effluents from wastewater treatment plants in Portugal (369.8 ng L-1). The acute effects were analysed focusing on behavioural endpoints (erratic and purposeful swimming), total distance travelled and swimming time), biomarkers of oxidative stress (the activities of the enzymes superoxide dismutase, catalase, glutathione peroxidase), biotransformation (the activity of glutathione S-transferases) and lipid peroxidation (levels of thiobarbituric acid reactive substances). Animals chronically exposed were also histologically analysed for sex determination and gonadal developmental stages identification. In terms of acute exposure, significant alterations were reported in terms of behavioural alterations (hyperactivity), followed by a general reduction in all tested biomarkers. Also, the analysis of chronically exposed fish evidenced no alterations in sex ratio and maturation stages. In addition, the analysis of chronically exposed fish evidenced no alterations in terms of sexual characteristics, suggesting that the chronic exposure of Danio rerio to simvastatin does not alter the sex ratio and maturation stages of individuals. This assumption suggests that simvastatin did not act as an endocrine disruptor. Moreover, the metabolism, neuronal interactions and the antioxidant properties of SIM seem to have modulated the hereby-mentioned results of toxicity. Results from this assay allow inferring that simvastatin can have an ecologically relevant impact in living organisms.


Asunto(s)
Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Embrión no Mamífero , Desarrollo Embrionario , Femenino , Masculino , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Estrés Oxidativo/efectos de los fármacos , Razón de Masculinidad , Espermatogénesis/efectos de los fármacos , Natación , Pez Cebra
9.
Chemosphere ; 269: 128725, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33153852

RESUMEN

Simvastatin (SV) is a typical lipid-lowering agent detected widely in waters, so its latent toxic effects to fish are deserved of concern. The purposes of this study aim at revealing the responses of antioxidant system in mosquitofish (Gambusia affinis) under SV exposure. Transcriptional expressions of oxidative stress-related key transcriptional factor Nrf2 and its downstream genes in mosquitofish were determined under SV exposure for different time. Partly related enzymatic activities, Nrf2 and MAPK protein expressions were also addressed in the same conditions, and histological changes in liver tissues were investigated too. Results showed that Nrf2 mRNA increased with the rising SV concentrations at 3 d and 7 d, displaying typical dose-dependent relationship, and Nrf2 protein by WB showed consistency with transcriptional changes to some degree. Comparatively, responses of gene expressions were more sensitive than enzymatic changes. The histological changes in the mosquitofish liver exposed to SV for 7 d indicated the potential adverse effects of statins. This work demonstrated that SV in aquatic environment could affect the transcriptional expression of antioxidant system, partly related enzymatic activity, and hepatic structure in the mosquitofish, revealing its potential risk on non-target organisms and environmental safety.


Asunto(s)
Ciprinodontiformes , Factor 2 Relacionado con NF-E2 , Animales , Ciprinodontiformes/genética , Hígado , Factor 2 Relacionado con NF-E2/genética , Transducción de Señal , Simvastatina/toxicidad
10.
Environ Int ; 144: 106020, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32861161

RESUMEN

The hypothesis that exposure to certain environmental chemicals during early life stages may disrupt reproduction across multiple non-exposed generations has significant implications for understanding disease etiology and adverse outcomes. We demonstrate here reproductive multi and transgenerational effects, at environmentally relevant levels, of one of the most prescribed human pharmaceuticals, simvastatin, in a keystone species, the amphipod Gammarus locusta. The transgenerational findings has major implications for hazard and risk assessment of pharmaceuticals and other contaminants of emerging concern given that transgenerational effects of environmental chemicals are not addressed in current hazard and risk assessment schemes. Considering that the mevalonate synthesis, one of the key metabolic pathways targeted by simvastatin, is highly conserved among metazoans, these results may also shed light on the potential transgenerational effects of simvastatin on other animals, including humans.


Asunto(s)
Anfípodos , Simvastatina , Animales , Epigénesis Genética , Humanos , Reproducción , Simvastatina/toxicidad
11.
Toxicol Appl Pharmacol ; 401: 115076, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32479918

RESUMEN

Statin induced myopathy (SIM) is a main deleterious effect leading to the poor treatment compliance, while the preventive or therapeutic treatments are absent. Mounting evidences demonstrated that vitamin D plays a vital role in muscle as a direct modulator. The deficiency of vitamin D was considered as a cause of muscle dysfunction, whereas the supplementation resulted in a remission. However, there is no causal proof that vitamin D supplementation rescues SIM. Here, using the mice model of simvastatin-induced myopathy, we investigated the role of vitamin D supplementation and the mechanisms associated with mitochondria. Results indicated that simvastatin administration (80 mg/kg) impaired skeletal muscle with the increased serum creatine kinase (CK) level and the declined grip strength, which were alleviated by vitamin D supplementation. Moreover, vitamin D supplementation rescued the energy metabolism dysfunction in simvastatin-treated mice gastrocnemius by reducing the abnormal aggregation of muscular glycogen and lactic acid. Mitochondrial homeostasis plays a key role in the process of energy metabolism. Thus, the mitochondrial dysfunction is a mortal damage for the highly energy-requiring tissue. In our study, the mitochondrial cristae observed under transmission electron microscope (TEM) were lytic in simvastatin-treated gastrocnemius. Interestingly, vitamin D supplementation improved the mitochondrial cristae shape by regulating the expression of mitofusin-1/2 (MFN1/2), optic atrophy 1 (OPA1) and dynamin-related protein 1 (Drp1). As expected, the mitochondrial dysfunction and oxidative stress was mitigated by vitamin D supplementation. In conclusion, these findings suggested that moderate vitamin D supplementation rescued simvastatin induced myopathy via improving the mitochondrial cristae shape and function.


Asunto(s)
Suplementos Dietéticos , Mitocondrias/efectos de los fármacos , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/tratamiento farmacológico , Simvastatina/toxicidad , Vitamina D/administración & dosificación , Animales , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/patología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Enfermedades Musculares/metabolismo , Distribución Aleatoria
12.
Ecotoxicology ; 29(7): 1062-1071, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32588236

RESUMEN

High consumption of drugs, combined with their presence in the environment, raises concerns about its consequences. Even though researches are often engaged in analyzing substances separately, that is not the environmental reality. Therefore, the aim of this study was to investigate the acute toxicity of the pharmaceuticals simvastatin, metformin, omeprazole and diazepam, and all possible mixtures between them, to the organism Aliivibrio fischeri, verifying possible synergistic or antagonistic effects and assessing byproducts formation. In terms of individual toxicity, omeprazole is the most toxic of the active ingredients, followed by simvastatin, diazepam and, finally, metformin. When the toxicity of mixtures was tested, synergism, antagonism and hormesis were perceived, most probably generated due to byproducts formation. Moreover, it was observed that even when compounds are at concentrations below the non-observed effect concentration (NOEC), there may be toxicity to the mixture. Hence, this work points to the urgent need for more studies involving mixtures, since chemicals are subject to interactions and modifications, can mix, and potentiate or nullify the toxic effect of each other.


Asunto(s)
Aliivibrio fischeri/efectos de los fármacos , Diazepam/toxicidad , Metformina/toxicidad , Omeprazol/toxicidad , Simvastatina/toxicidad , Pruebas de Toxicidad Aguda
13.
J Toxicol Environ Health A ; 83(3): 113-125, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32116137

RESUMEN

Simvastatin (SIM), a hypocholesterolaemic drug belonging to the statins group, is a widely prescribed pharmaceutical for prevention of cardiovascular diseases. Several studies showed that lipophilic statins, as SIM, cross the blood-brain barrier and interfere with the energy metabolism of the central nervous system in humans and mammalian models. In fish and other aquatic organisms, the effects of SIM on the brain energy metabolism are unknown, particularly following exposure to low environmentally relevant concentrations. Therefore, the present study aimed at investigating the influence of SIM on gene signaling pathways involved in brain energy metabolism of adult zebrafish (Danio rerio) following chronic exposure (90 days) to environmentally relevant SIM concentrations ranging from 8 ng/L to 1000 ng/L. Real-time PCR was used to determine the transcript levels of several genes involved in different pathways of the brain energy metabolism (glut1b, gapdh, acadm, accα, fasn, idh3a, cox4i1, and cox5aa). The findings here reported integrated well with ecological and biochemical responses obtained in a parallel study. Data demonstrated that SIM modulates transcription of key genes involved in the mitochondrial electron transport chain, in glucose transport and metabolism, in fatty acid synthesis and ß-oxidation. Further, SIM exposure led to a sex-dependent transcription profile for some of the studied genes. Overall, the present study demonstrated, for the first time, that SIM modulates gene regulation of key pathways involved in the energy metabolism in fish brain at environmentally relevant concentrations.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/toxicidad , Bioensayo , Esquema de Medicación , Femenino , Humanos , Masculino , Simvastatina/administración & dosificación , Contaminantes Químicos del Agua/administración & dosificación , Pez Cebra
14.
Anim Biotechnol ; 31(5): 391-396, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31060421

RESUMEN

Simvastatin (SIM) is a widely used anticholesterolemic drug that blocks the biosynthesis of cholesterol. However, SIM also has pleiotropic effects on 3-hydroxy-3-methyglutary-CoA reductase (HMGR), cholesteryl ester transfer protein (CETP), and lipoprotein lipase (LPL), which are important genes in the cholesterol biosynthesis and transport processes. We investigated the effects of different concentrations of SIM on the mRNA expression of these genes in bovine intramuscular and subcutaneous adipocytes from the longissimus dorsi muscle and subcutaneous fat tissues of Luxi Yellow cattle. The results showed that SIM treatment showed dose-dependent toxicity on normal adipose cells, but no effect on cell proliferation. SIM decreased HMGR expression in a dose-dependent manner but showed no significant effect on CETP and LPL expression. Thus, SIM may lower the cholesterol content by decreasing the HMGR expression level, but CETP and LPL may be regulated through other mechanisms, which require further investigation.


Asunto(s)
Adipocitos , Proliferación Celular/efectos de los fármacos , Músculo Esquelético , Simvastatina/toxicidad , Grasa Subcutánea , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Bovinos , Colesterol/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Grasa Subcutánea/citología , Grasa Subcutánea/efectos de los fármacos
15.
Ecotoxicol Environ Saf ; 182: 109389, 2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31272027

RESUMEN

Simvastatin (SV), as an hypocholesterolaemic drug, has been detected in various aquatic environment. However, limited information is available on the effects of SV on freshwater invertebrates. In the present study, we investigated the toxic effects of SV on Daphnia. magna (D. magna) through measuring the physiological changes (e.g., survival, growth rate, and reproduction) in a 21-d chronic toxicity test We also determined the expression of seven detoxification and reproduction-related genes (i.e. HR96, P-gp, CYP360A8, GST, CYP314, EcR and Vtg) and several enzymes (i.e. APND, ERND, GST and CAT) in a acute test (24 h). Results showed that high concentration (e.g. 50 µg L-1) of SV for short time exposure (e.g. 24 h) significantly induced the expression of HR96 and P-gp (e.g. up to 2.5 folds)and enzymes (e.g. increasing 4.0 folds for ERND and GST activity) in D. magna.. The long-term chronic exposure (21 days) may cause the changes of life history parameters such as decreasing total egg production number per individual and intrinsic growth rates etc. SV may act as a potential endocrine disruptor to D. magna and the reproduction parameters were more sensitive endpoints than the survival and growth for evaluating SV exposure.


Asunto(s)
Daphnia , Disruptores Endocrinos/toxicidad , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Daphnia/efectos de los fármacos , Daphnia/enzimología , Daphnia/genética , Inactivación Metabólica/efectos de los fármacos , Inactivación Metabólica/genética , Reproducción/efectos de los fármacos , Reproducción/genética , Pruebas de Toxicidad Crónica
16.
Sci Rep ; 9(1): 7409, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092879

RESUMEN

Simvastatin is an inhibitor of the 3-hydroxy-3-methylglutaryl-CoA reductase used for decreasing low density lipoprotein (LDL)-cholesterol in patients. It is well-tolerated but can cause myopathy. Our aims were to enlarge our knowledge regarding mechanisms and effects of insulin on simvastatin-associated myotoxicity in C2C12 myotubes. Simvastatin (10 µM) reduced membrane integrity and ATP content in myotubes treated for 24 hours, which could be prevented and partially reversed concentration- and time-dependently by insulin. Furthermore, simvastatin impaired the phosphorylation of Akt (Protein Kinase B) mainly at Ser473 and less at Thr308, indicating impaired activity of the mammalian Target of Rapamycin Complex 2 (mTORC2). Impaired activation of Akt increased mRNA expression of the muscle atrophy F-Box (MAFbx), decreased activation of the mammalian Target of Rapamycin Complex 1 (mTORC1) and stimulated apoptosis by impairing the Ser9 phosphorylation of glycogen synthase kinase 3ß. Decreased phosphorylation of Akt at both phosphorylation sites and of downstream substrates as well as apoptosis were prevented concentration-dependently by insulin. In addition, simvastatin caused accumulation of the insulin receptor ß-chain in the endoplasmic reticulum (ER) and increased cleavage of procaspase-12, indicating ER stress. Insulin reduced the expression of the insulin receptor ß-chain but increased procaspase-12 activation in the presence of simvastatin. In conclusion, simvastatin impaired activation of Akt Ser473 most likely as a consequence of reduced activity of mTORC2. Insulin could prevent the effects of simvastatin on the insulin signaling pathway and on apoptosis, but not on the endoplasmic reticulum (ER) stress induction.


Asunto(s)
Insulina/farmacología , Células Musculares/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Simvastatina/toxicidad , Animales , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/citología , Receptor de Insulina/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Simvastatina/antagonistas & inhibidores
17.
Toxicol Sci ; 169(2): 543-552, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30859212

RESUMEN

Plasma membrane ion channels and mitochondrial electron transport complexes (mETC) are recognized "off targets" for certain drugs. Simvastatin is one such drug, a lipophilic statin used to treat hypercholesterolemia, but which is also associated with adverse effects like myopathy and increased risk of glucose intolerance. Such myopathy is thought to arise through adverse actions of simvastatin on skeletal muscle mETC and mitochondrial respiration. In this study, we investigated whether the glucose intolerance associated with simvastatin is also mediated via adverse effects on mETC in pancreatic beta-cells because mitochondrial respiration underlies insulin secretion from these cells, an effect in part mediated by promotion of Ca2+ influx via opening of voltage-gated Ca2+ channels (VGCCs). We used murine pancreatic beta-cells to investigate these ideas. Mitochondrial membrane potential, oxygen consumption, and ATP-sensitive-K+-channel activity were monitored as markers of mETC activity, respiration, and cellular ATP/ADP ratio respectively; Ca2+ channel activity and Ca2+ influx were also measured. In intact beta-cells, simvastatin inhibited oxidative respiration (IC50 approximately 3 µM) and mETC (1 < IC50 < 10 µM), effects expected to impair VGCC opening. Consistent with this idea simvastatin > 0.1 µM reversed activation of VGCCs by glucose but had no significant effect in the sugar's absence. The VGCC effects were mimicked by rotenone which also decreased respiration and ATP/ADP. This study demonstrates modulation of beta-cell VGCC activity by mitochondrial respiration and their sensitivity to mETC inhibitors. This reveals a novel outcome for the action of drugs like simvastatin for which mETC is an "off target".


Asunto(s)
Bloqueadores de los Canales de Calcio/toxicidad , Canales de Calcio Tipo L/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Simvastatina/toxicidad , Animales , Calcio/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Mitocondrias/fisiología , Fosforilación Oxidativa/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Rotenona/farmacología
18.
Biochem Pharmacol ; 164: 23-33, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30796916

RESUMEN

Statins inhibit cholesterol biosynthesis and lower serum LDL-cholesterol levels. They are generally well tolerated, but can cause insulin resistance in patients. Therefore, we investigated the mechanisms underlying the statin-induced insulin resistance. We used mice and C2C12 myotubes (murine cell line): mice (n = 10) were treated with oral simvastatin (5 mg/kg/day) or water (control) for 21 days and C2C12 cells were exposed to 10 µM simvastatin for 24 h. After intraperitoneal glucose application (2 g/kg), simvastatin-treated mice had higher glucose but equal insulin plasma concentrations than controls and lower glucose transport into skeletal muscle. Similarly, glucose uptake by C2C12 myotubes exposed to 10 µM simvastatin for 24 h was impaired compared to control cells. In simvastatin-treated C2C12 myotubes, mRNA and protein expression of the insulin receptor (IR) ß-chain was increased, but the phosphorylation (Tyr1361) was impaired. Simvastatin decreased numerically Akt/PKB Thr308 phosphorylation (via insulin signaling pathway) and significantly Akt/PKB Ser473 phosphorylation (via mTORC2), which was explained by impaired phosphorylation of mTOR Ser2448. Reduced phosphorylation of Akt/PKB impaired downstream phosphorylation of GSK3ß, leading to impaired translocation of GLUT4 into plasma membranes of C2C12 myotubes. In contrast, reduced phosphorylation of AS160 could be excluded as a reason for impaired GLUT4 translocation. In conclusion, simvastatin caused insulin resistance in mice and impaired glucose uptake in C2C12 myotubes. The findings in myotubes can be explained by diminished activation of Akt/PKB by mTORC2 and downstream effects on GSK3ß, impairing the translocation of GLUT4 and the uptake of glucose.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Resistencia a la Insulina/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Simvastatina/toxicidad , Animales , Línea Celular , Glucosa/antagonistas & inhibidores , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distribución Aleatoria
19.
Toxicol Appl Pharmacol ; 365: 112-123, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30639414

RESUMEN

We showed previously that in utero exposure to the cholesterol-lowering drug simvastatin (SMV) during sex differentiation lowers fetal lipids and testicular testosterone production (T Prod) in Hsd:SD rats. Here, the effects of SMV on fetal lipids and T Prod in Crl:CD(SD) rats were correlated with postnatal alterations in F1 males. The current study was conducted in two parts: 1) a prenatal assessment to confirm and further characterize the dose response relationship among previously reported alterations of SMV on fetal T Prod and the fetal lipid profile and 2) a postnatal assessment to determine the effects of SMV exposure during the periods of major organogenesis and/or sexual differentiation on F1 offspring growth and development. We hypothesized that SMV would have adverse effects on postnatal development and sexual differentiation as a consequence of the disruptions of fetal lipid levels and testicular T Prod since fetal cholesterol is essential for normal intrauterine growth and development and steroid synthesis. In the prenatal assessment, SMV was administered orally at 0, 15.6, 31.25, 62.5, 80, 90, 100, and 110 mg SMV/kg/d from GD 14-18, the period that cover the critical window of sex differentiation in the male rat fetus. T Prod was maximally reduced by ~40% at 62.5 mg/kg/d, and higher doses induced overt maternal and toxicity. In the postnatal assessment, SMV was administered at 0, 15.6, 31.25, and 62.5 mg/kg/d from GD 8-18 to determine if it altered postnatal development. We found that exposure during this time frame to 62.5 mg SMV/kg/d reduced pup viability by 92%, decreased neonatal anogenital distance, and altered testis histology and morphology in 17% of the F1 males. In another group, SMV was administered only during the masculinizing window (GD14-18) at 62.5 mg/kg/d to determine if male rat sexual differentiation and postnatal reproductive development were altered. SMV-exposed F1 males displayed female-like areolae/nipples, delayed puberty, and reduced seminal vesicle and levator ani-bulbocavernosus weights. Together, these results demonstrate that in utero exposure to SMV reduces offspring viability and permanently disrupts reproductive tract development in the male offspring. While the effects of high dose, short term in utero exposure to SMV in the adult male are likely androgen-dependent and consistent with the 40% reduction in T Prod in the fetal testes, long-term, lower dose administration induced some effects that were likely not mediated by decreased T Prod.


Asunto(s)
Feto/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Simvastatina/toxicidad , Testículo/efectos de los fármacos , Testosterona/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Femenino , Feto/metabolismo , Edad Gestacional , Masculino , Técnicas de Cultivo de Órganos , Organogénesis/efectos de los fármacos , Embarazo , Ratas Sprague-Dawley , Medición de Riesgo , Diferenciación Sexual/efectos de los fármacos , Desarrollo Sexual/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testículo/metabolismo
20.
Sci Total Environ ; 651(Pt 1): 399-409, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30240922

RESUMEN

Simvastatin is one of the most commonly cholesterol-lowering prescribed drugs all over the world. With the increase of consumption of these pharmaceuticals and subsequent their discharge into the aquatic environment in recent years, they are present at detectable levels in most sewage effluents. Unfortunately, limited information is provided about their potential impacts on aquatic organisms, especially on the detoxification-related metabolism in fish. In the present study, one local native benthic fish (Mugilogobius abei) in southern China was employed as test species and exposed to SV (0.5 µg L-1, 5 µg L-1, 50 µg L-1 and 500 µg L-1) for 72 h. The transcriptional expression of nucleus transcriptional factor pregnane X receptor (PXR) and its downstream targeted genes including multixenobiotics resistance protein or permeability glycoprotein (P-gp), cytochrome 1A (CYP1A), cytochrome P450 3A (CYP3A), glutathione-S-transferase (GST) and the expression of associated microRNA such as miR-27, miR-34 and miR-148 in Mugilogobius abei were investigated. Result showed that the expressions of P-gp, CYP 1A, CYP 3A, GST and PXR were induced to some extend under simvastatin exposure for 72 h. A positive correlation was observed between PXR and CYP1A, CYP3A and P-gp. While for microRNA, a negative relationship was found between miR-34a and CYP3A, CYP1A. The expression of miR-148a was significantly induced under the exposure of SV (50 µg L-1), which was positive related to the transcriptional expression of PXR. For enzyme activity, erythromycin N-demethylase (ERND) significantly increased at 24 h and the activity of catalase (CAT) and superoxide dismutase (SOD) exhibited different trends. CAT was slightly inhibited at 24 h exposure but SOD was significantly induced in high concentration. Glutathione-S-transferase (GST) activity was significant inhibited after 72 h exposure. The reductive small molecule glutathione (GSH) content showed obvious decrease, while the quantity of malondialdehyde (MDA) increased significantly in high concentrations of SV exposure. GSH and MDA showed a typical negative correlation to some degree. Moreover, simvastatin caused histological changes in the liver tissues of M. abei, especially the size of adipocyte significantly decreased. The present study indicated that environmentally relevant concentration SV may affect the PXR signaling pathway in M. abei and pose potential ecological risks to non-target organisms like fish.


Asunto(s)
Anticolesterolemiantes/toxicidad , Proteínas de Peces/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Perciformes/genética , Simvastatina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Proteínas de Peces/metabolismo , Hígado/patología , Perciformes/metabolismo , Receptor X de Pregnano/genética , Receptor X de Pregnano/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA