Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Reprod Toxicol ; 108: 1-9, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34974146

RESUMEN

Tamoxifen, a selective non-steroidal estrogen receptor modulator, is the standard adjuvant endocrine treatment for breast cancer. Since information on the risk of using tamoxifen during pregnancy is still scarce, this study evaluated whether the in utero and lactational treatment with this drug could compromise reproductive and behavioural parameters in male offspring. Pregnant Wistar rats were exposed to three doses of tamoxifen (0.12; 0.6; 3 µg/kg), by gavage, from gestational day 15 to lactational day 20. Tamoxifen exposure did not alter the anogenital distance in the male offspring; however, there was a significant increase in the body weight in the 0.12 µg/kg dose and a decrease in the 0.6 µg/kg dose. The male offspring treated with the highest dose exhibited a delay in the onset of puberty, evidenced by an increase in the age of preputial separation. Regarding sperm parameters, there was an increase in the sperm count in the cauda epididymis in the intermediate and highest dose groups, in addition to an increase in the number of static sperm and a decrease in the progressive sperm in the same groups. Moreover, an increase in the number of hyperplasia of the epithelial clear cells was observed in the epididymis. In conclusion, the present study demonstrated that maternal exposure to tamoxifen compromised the installation of puberty of the male offspring and the maturation of the epididymis, affecting sperm storage and motility in the adult life.


Asunto(s)
Conducta Animal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Espermatozoides/efectos de los fármacos , Tamoxifeno/toxicidad , Animales , Epidídimo/efectos de los fármacos , Epidídimo/crecimiento & desarrollo , Femenino , Hipotálamo/citología , Lactancia , Masculino , Intercambio Materno-Fetal , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Ratas Wistar , Receptores Androgénicos/metabolismo , Maduración Sexual/efectos de los fármacos , Recuento de Espermatozoides , Motilidad Espermática/efectos de los fármacos , Espermatozoides/fisiología
2.
Gastroenterology ; 158(6): 1650-1666.e15, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32032583

RESUMEN

BACKGROUND & AIMS: Gastric chief cells, a mature cell type that secretes digestive enzymes, have been proposed to be the origin of metaplasia and cancer through dedifferentiation or transdifferentiation. However, studies supporting this claim have had technical limitations, including issues with the specificity of chief cell markers and the toxicity of drugs used. We therefore sought to identify genes expressed specifically in chief cells and establish a model to trace these cells. METHODS: We performed transcriptome analysis of Mist1-CreERT-traced cells, with or without chief cell depletion. Gpr30-rtTA mice were generated and crossed to TetO-Cre mice, and lineage tracing was performed after crosses to R26-TdTomato mice. Additional lineage tracing experiments were performed using Mist1-CreERT, Kitl-CreERT, Tff1-Cre, and Tff2-Cre mice crossed to reporter mice. Mice were given high-dose tamoxifen or DMP-777 or were infected with Helicobacter pylori to induce gastric metaplasia. We studied mice that expressed mutant forms of Ras in gastric cells, using TetO-KrasG12D, LSL-KrasG12D, and LSL-HrasG12V mice. We analyzed stomach tissues from GPR30-knockout mice. Mice were given dichloroacetate to inhibit pyruvate dehydrogenase kinase (PDK)-dependent cell competition. RESULTS: We identified GPR30, the G-protein-coupled form of the estrogen receptor, as a cell-specific marker of chief cells in gastric epithelium of mice. Gpr30-rtTA mice crossed to TetO-Cre;R26-TdTomato mice had specific expression of GPR30 in chief cells, with no expression noted in isthmus stem cells or lineage tracing of glands. Expression of mutant Kras in GPR30+ chief cells did not lead to the development of metaplasia or dysplasia but, instead, led to a reduction in labeled numbers of chief cells and a compensatory expansion of neck lineage, which was derived from upper Kitl+ clones. Administration of high-dose tamoxifen, DMP-777, or H pylori decreased the number of labeled chief cells. Chief cells were eliminated from epithelia via GPR30- and PDK-dependent cell competition after metaplastic stimuli, whereas loss of GRP30 or inhibition of PDK activity preserved chief cell numbers and attenuated neck lineage cell expansion. CONCLUSIONS: In tracing studies of mice, we found that most chief cells are lost during metaplasia and therefore are unlikely to contribute to gastric carcinogenesis. Expansion of cells that coexpress neck and chief lineage markers, known as spasmolytic polypeptide-expressing metaplasia, does not occur via dedifferentiation from chief cells but, rather, through a compensatory response from neck progenitors to replace the eliminated chief cells.


Asunto(s)
Células Principales Gástricas/fisiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Azetidinas/toxicidad , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Ácido Dicloroacético/administración & dosificación , Modelos Animales de Enfermedad , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Infecciones por Helicobacter/microbiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaplasia/inducido químicamente , Metaplasia/microbiología , Metaplasia/patología , Ratones , Ratones Noqueados , Piperazinas/toxicidad , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/fisiología , Tamoxifeno/toxicidad
3.
Toxicol Mech Methods ; 30(2): 115-123, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31532279

RESUMEN

Tamoxifen (TAM) is used in breast cancer chemotherapy since its approval by the Food and Drug Administration in 1977. However, TAM therapy is accompanied with hepatotoxicity - a source of worry to clinicians. Oxidative stress and inflammation are the major implicated mechanisms contributing to TAM hepatotoxicity. In this study, we explored whether zinc (Zn) supplementation could prevent TAM-induced hepatotoxicity in female Wistar rats. Rats were subjected to oral pretreatment of Zn (100 mg/kg body weight (b.w.)/day) for 14 days against hepatic toxicity induced by single intraperitoneal administration of TAM (50 mg/kg b.w.) on day 13. TAM markedly elevated serum liver enzymes, whereas total protein and albumin considerably reduced. TAM caused prominent depletion of hepatic-reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activity. Also, TAM significantly increased malondialdehyde (MDA) level. Further, it raised liver levels of tumor necrosis factor-α (TNF-α), interleukin-1ß, (IL-1ß), interleukin-6 (IL-6), and nitric oxide (NO) confirmed by the liver histopathological alterations. The mechanistic inflammatory expression of inducible nitric oxide synthase (iNOS) and nuclear factor-kappa B (NF-ĸB), and expression of caspase-3 protein prominently increased. Zinc supplementation significantly modulated serum liver function markers, antioxidant enzymes, and GSH and MDA levels. Zinc downregulated the expression of cytokines, NO, iNOS, NF-ĸB and caspase-3, and ameliorated histopathological changes. Zinc protects against TAM-induced hepatotoxicity; it may serve as an adjuvant supplement for female patients undergoing TAM chemotherapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cloruros/farmacología , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Tamoxifeno/toxicidad , Compuestos de Zinc/farmacología , Animales , Antioxidantes/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Cloruros/administración & dosificación , Citocinas/metabolismo , Suplementos Dietéticos , Femenino , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Sustancias Protectoras/administración & dosificación , Ratas , Ratas Wistar , Transducción de Señal , Compuestos de Zinc/administración & dosificación
4.
J Pathol ; 248(2): 179-190, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30689202

RESUMEN

In the mammalian stomach, the isthmus has been considered as a stem cell zone. However, various locations and proliferative activities of gastric stem cells have been reported. We focused here on the stem cell marker Bmi1, a polycomb group protein, aiming to elucidate the characteristics of Bmi1-expressing cells in the stomach and to examine their stem cell potential. We investigated the Bmi1-expressing cell lineage in Bmi1-CreERT; Rosa26-YFP, LacZ or Rosa26-Confetti mice. We examined the in vivo and ex vivo effects of Bmi1-expressing cell ablation by using Bmi1-CreERT; Rosa26-iDTR mice. The Bmi1 lineage was also traced during regeneration after high-dose tamoxifen-, irradiation- and acetic acid-induced mucosal injuries. In the lineage-tracing experiments using low-dose tamoxifen, Bmi1-expressing cells in the isthmus of the gastric antrum and corpus provided progeny bidirectionally, towards both the luminal and basal sides over 6 months. In gastric organoids, Bmi1-expressing cells also provided progeny. Ablation of Bmi1-expressing cells resulted in impaired gastric epithelium in both mouse stomach and organoids. After high-dose tamoxifen-induced gastric mucosal injury, Bmi1-expressing cell lineages expanded and fully occupied all gastric glands of the antrum and the corpus within 7 days after tamoxifen injection. After irradiation- and acetic acid-induced gastric mucosal injuries, Bmi1-expressing cells also contributed to regeneration. In conclusion, Bmi1 is a gastric stem cell marker expressed in the isthmus of the antrum and corpus. Bmi1-expressing cells have stem cell potentials, both under physiological conditions and during regeneration after gastric mucosal injuries. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Antro Pilórico/metabolismo , Células Madre/metabolismo , Ácido Acético , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Modelos Animales de Enfermedad , Fluorouracilo/toxicidad , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones Transgénicos , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Antro Pilórico/efectos de los fármacos , Antro Pilórico/embriología , Antro Pilórico/efectos de la radiación , Regeneración , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/efectos de la radiación , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/metabolismo , Úlcera Gástrica/patología , Tamoxifeno/toxicidad
5.
Ecotoxicol Environ Saf ; 169: 18-27, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30412894

RESUMEN

The objective was to investigate endocrine-disrupting effects of polar compounds from oxidized frying oil. Estrogenicity of polar compounds was tested with a rat uterotrophic bioassay. Dietary oxidized frying oil (containing 51% polar compounds) or polar compounds isolated from it were incorporated into feed (in lieu of fresh soybean oil) and fed to ovariectomized rats, with or without treatment with exogenous ethynyl estradiol. Exogenous estrogen restored uterine weight, and caused histological abnormalities (stratified epithelia and conglomerate glands) as well as proliferation of uterine epithelial cells. However, tamoxifen or polar compounds reduced these effects. Furthermore, tamoxifen or polar compounds down-regulated uterine mRNA expression of estrogen receptor (ER)-target genes, implicating reduced ER activity in this hypo-uterotrophic effect. Inhibition of ER signaling and mitosis by polar compounds were attributed to reduced MAPK and AKT activation, as well as a reduced ligand binding domain-transactivity of ERα/ß. We concluded polar compounds from frying oil are potential endocrine-disrupting chemicals, with implications for food and environmental safety.


Asunto(s)
Disruptores Endocrinos/toxicidad , Antagonistas de Estrógenos/toxicidad , Animales , Culinaria , Dieta , Estrógenos/farmacología , Etinilestradiol/farmacología , Femenino , Oxidación-Reducción , Ratas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Aceite de Soja , Tamoxifeno/toxicidad , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología
6.
Acta Pharmacol Sin ; 40(5): 608-619, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30315252

RESUMEN

Tamoxifen (TMX) is an antiestrogen drug that is used in the treatment and prevention of all stages of estrogen-dependent breast cancer. Adverse effects of TMX include hepatotoxicity. In this study, we investigated the therapeutic effects of osthole, isolated from medicinal plants especially Fructus Cnidii, on TMX-induced acute liver injury in mice. Mice were injected with osthole (100 mg/kg, ip) or vehicle, followed by TMX (90 mg/kg, ip) 24 h later. We showed that a single injection of TMX-induced liver injury and oxidative stress. Pretreatment with osthole attenuated TMX-induced liver injury evidenced by dose-dependent reduction of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities. Pretreatment with osthole also blunted TMX-induced oxidative stress, evidenced by significant increase of reduced glutathione (GSH) as well as reduction of malondialdehyde (MDA) and hydrogen peroxide (H2O2). Consistently, osthole significantly enhanced the expressions of antioxidant genes (GPX1, SOD2, GCL-c, and G6pdh), but suppressed those of pro-oxidant genes (NOX2 and ACOX). Furthermore, osthole inhibited the production of inflammatory cytokines, reduced the metabolic activation of TMX, and promoted its clearance. We further revealed that osthole elevated hepatic cAMP and cGMP levels, but inhibition of PKA or PKG failed to abolish the hepatoprotective effect of osthole. Meanwhile, prominent phosphorylation of p38 was observed in liver in response to TMX, which was significantly inhibited by osthole. Pretreatment with SB203580, a p38 inhibitor, significantly attenuated TMX-induced increase of ALT and AST activities, reduced oxidative stress, and reversed the alterations of gene expression caused by TMX. Moreover, pretreatment with L-buthionine sulfoximine (BSO), an inhibitor of GSH synthesis, partly reversed the effect of osthole on TMX-induced liver injury. Consistently, pretreatment with N-acetyl-L-cysteine (NAC) significantly attenuated TMX-induced increase in ALT and AST activities. Notably, both BSO and NAC had no detectable effect on the phosphorylation levels of p38. Collectively, our results suggest that osthole prevents TMX hepatotoxicity by suppressing p38 activation and subsequently reducing TMX-induced oxidative damage.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cumarinas/uso terapéutico , Moduladores de los Receptores de Estrógeno/toxicidad , Tamoxifeno/toxicidad , Alanina Transaminasa/sangre , Alanina Transaminasa/metabolismo , Animales , Antioxidantes/administración & dosificación , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/metabolismo , Cumarinas/administración & dosificación , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Moduladores de los Receptores de Estrógeno/administración & dosificación , Peróxido de Hidrógeno/metabolismo , Inflamación/prevención & control , Hígado/patología , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Tamoxifeno/administración & dosificación
7.
J Appl Toxicol ; 37(8): 943-953, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28138993

RESUMEN

Lipid profiling has emerged as an effective approach to not only screen disease and drug toxicity biomarkers but also understand their underlying mechanisms of action. Tamoxifen, a widely used antiestrogenic agent for adjuvant therapy against estrogen-positive breast cancer, possesses side effects such as hepatic steatosis and phospholipidosis (PLD). In the present study, we administered tamoxifen to Sprague-Dawley rats and used lipidomics to reveal tamoxifen-induced alteration of the hepatic lipid profile and its association with the plasma lipid profile. Treatment with tamoxifen for 28 days caused hepatic PLD in rats. We compared the plasma and liver lipid profiles in treated vs. untreated rats using a multivariate analysis to determine differences between the two groups. In total, 25 plasma and 45 liver lipids were identified and altered in the tamoxifen-treated group. Of these lipids, arachidonic acid (AA)-containing phosphatidylcholines (PCs), such as PC (17:0/20:4) and PC (18:1/20:4), were commonly reduced in both plasma and liver. Conversely, tamoxifen increased other phosphoglycerolipids in the liver, such as phosphatidylethanolamine (18:1/18:1) and phosphatidylinositol (18:0/18:2). We also examined alteration of AA-containing PCs and some phosphoglycerolipids in the pre-PLD stage and found that these lipid alterations were initiated before pathological alteration in the liver. In addition, changes in plasma and liver levels of AA-containing PCs were linearly associated. Moreover, levels of free AA and mRNA levels of AA-synthesizing enzymes, such as fatty acid desaturase 1 and 2, were decreased by tamoxifen treatment. Therefore, our study demonstrated that AA-containing PCs might have potential utility as novel and predictive biomarkers for tamoxifen-induced PLD. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Ácido Araquidónico/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Lipidosis/metabolismo , Hígado/metabolismo , Fosfatidilcolinas/sangre , Tamoxifeno/toxicidad , Animales , Ácido Araquidónico/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Relación Dosis-Respuesta a Droga , Metabolismo de los Lípidos/efectos de los fármacos , Lipidosis/inducido químicamente , Hígado/efectos de los fármacos , Masculino , Fosfatidilcolinas/metabolismo , Ratas Sprague-Dawley
8.
Chem Biol Interact ; 255: 12-22, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26718876

RESUMEN

A possible risk factor for drug-induced hepatotoxicity is drug metabolizing enzyme activity, which is known to vary among individuals due to genetic (genetic polymorphism) and environmental factors (environmental pollutants, foods, and medications that are inhibitors or inducers of drug metabolizing enzymes). We hypothesize that hepatic cytochrome P450-dependent monooxygenase (CYP) activity is one of the key risk factors for drug induced liver injuries (DILI) in the human population, especially for drugs that are metabolically activated to cytotoxic/reactive metabolites. Human hepatocytes from 19 donors were evaluated for the activities of 8 major P450 isoforms: CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4. Extensive individual variations were observed, consistent with what is known to be in the human population. As CYP3A4 is known to be one of the most important P450 isoforms for drug metabolism, studies were performed to evaluate the relationship between the in vitro cytotoxicity of hepatotoxic drugs and CYP3A4 activity. In a proof of concept study, hepatocytes from six donors (lots) representing the observed range of CYP3A4 activities were chosen for the evaluation of in vitro hepatotoxicity of four drugs known to be associated with acute liver failure: acetaminophen, cyclophosphamide, ketoconazole, and tamoxifen. The hepatocytes were cultured in collagen-coated plates and treated with the hepatotoxicants for approximately 24 h, followed by viability determination based on cellular adenosine triphosphate (ATP) contents. HH1023, the lot of hepatocytes with the highest CYP3A4 activity, was found to be the most sensitive to the cytotoxicity of all 4 hepatotoxic drugs, thereby suggesting that high CYP3A4 activity may be a risk factor. To further validate the relationship, a second study was performed with hepatocytes from 16 donors. In this study, the hepatocytes were quantified for CYP3A4 activity at the time of treatment. Results of the second study show confirm the correlation between with high CYP3A4 activity and sensitivity to hepatotoxic drugs. Our results with primary cultured hepatocytes from multiple donors support the hypothesis that elevated P450 activity may be a risk factor for drug-induced liver injuries.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Hepatocitos/efectos de los fármacos , Acetaminofén/toxicidad , Adolescente , Adulto , Anciano , Analgésicos no Narcóticos/toxicidad , Antifúngicos/toxicidad , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Niño , Ciclofosfamida/toxicidad , Citocromo P-450 CYP3A/metabolismo , Antagonistas de Estrógenos/toxicidad , Femenino , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunosupresores/toxicidad , Cetoconazol/toxicidad , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Isoformas de Proteínas/metabolismo , Tamoxifeno/toxicidad , Adulto Joven
9.
ACS Chem Biol ; 11(1): 251-62, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26569462

RESUMEN

Tamoxifen is not only considered a very potent chemotherapeutic adjuvant for estrogen receptor positive breast cancers but also a very good chemo-preventive drug. Recently, there has been a rising amount of evidence for a nongenomic cytotoxicity of tamoxifen, even in estrogen receptor negative cells, which has greatly confounded researchers. Clinically, the side effects of tamoxifen can be very serious, ranging from liver steatosis to cirrhosis, tumorigenesis, or onset of porphyrias. Herein, we deciphered the nongenomic, mitochondrial cytotoxicity of tamoxifen in estrogen receptor positive MCF7 versus triple-negative MDA-MB-231 cells, employing the mitochondrial complex III quinoloxidizing-center inhibitor myxothiazol. We showed a role for hydroxyl-radical-mediated lipid peroxidation, catalyzed by iron, stemming from the redox interactions of tamoxifen quinoid metabolites with complex III, resulting in Fenton-capable reduced quinones. The role of tamoxifen semiquinone species in mitochondrial toxicity was also shown together with evidence of mitochondrial DNA damage. Tamoxifen caused an overall metabolic (respiratory and glycolytic) rate decrease in the Pasteur type MCF cells, while in the Warburg type MDA-MB-231 cells the respiratory rate was not significantly affected and the glycolytiv rate was significantly boosted. The nongenomic cytotoxicity of tamoxifens was hence associated with the metabolic phenotype and redox activity of the cells, as in the present paradigm of Pasteur MCF7s versus Warburg MDA-MB-231 cells. Our present findings call for caution in the use of the drugs, especially as a chemopreventive and/or in cases of iron overload diseases.


Asunto(s)
Mitocondrias/efectos de los fármacos , Tamoxifeno/toxicidad , Antineoplásicos/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Peroxidación de Lípido/efectos de los fármacos , Células MCF-7 , Microscopía Confocal , Estructura Molecular , Oxidación-Reducción/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas
10.
Pharm Biol ; 52(8): 1009-14, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24646302

RESUMEN

CONTEXT: Tamoxifen (TAM) is widely used for treatment of hormone-dependent breast cancer; however, it may be accompanied with hepatic injury. Allicin is the most abundant thiosulfinate molecule from garlic with the potential to provide beneficial effects on various diseases. OBJECTIVE: To elucidate the effect of commercially available allicin on both antitumor activity and liver injury of TAM. MATERIALS AND METHODS: The cytotoxicity of TAM and/or allicin was evaluated in vitro using cultured Ehrlich ascites carcinoma (EAC) cells and in vivo against murine tumor (solid) model of EAC. TAM induced liver injury in rats by intraperitoneally (i.p.) injection at a dose of 45 mg/kg, for 7 successive days. RESULTS: TAM at a dose of 3 µM (IC50) significantly decreased percent survival of EAC to 52%. TAM combination with allicin (5 or 10 µM) showed a significant cytotoxic effect compared with the TAM-treated group as manifested by a decrease in percent survival of EAC to 35% and 29%, respectively. Allicin (10 mg/kg, orally) enhanced the efficacy of TAM (1 mg/kg, i.p.) in mice as manifested by a significant increase in solid tumor growth inhibition by 82% compared with 70% in the TAM group. In rats, TAM intoxication resulted in a significant decline in SOD, GSH, and total protein with significant elevation in TBARS, ALT and AST, ALP, LDH, total bilirubin, γGT, and TNF-α levels. These changes are abrogated by allicin treatment. DISCUSSION AND CONCLUSION: The results suggest the beneficial role of allicin as an adjuvant to TAM in cancer treatment by alleviating liver injury.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Antioxidantes/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Ácidos Sulfínicos/uso terapéutico , Tamoxifeno/toxicidad , Animales , Carcinoma de Ehrlich/tratamiento farmacológico , Carcinoma de Ehrlich/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Disulfuros , Femenino , Ratones , Distribución Aleatoria , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Glia ; 61(10): 1735-47, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918524

RESUMEN

NG2-glia are known to proliferate in the adult brain, however the extent of their mitotic and regenerative capacity and particularly their adult origin is uncertain. By employing a paradigm of mitotic blockade in conjunction with genetic fate tracing we demonstrate that intracerebroventricular mitotic blocker infusion leads to wide-spread and complete ablation of NG2-glial cells in the hypothalamus and other periventricular brain regions. However, despite the extensive glia loss, parenchymal NG2-glia coverage is fully restored to pretreatment levels within two weeks. We further reveal that in response to mitotic blocker treatment, NG2-glia bordering the ablated territories start to express the stem cell marker nestin, divide and migrate to replace the lost cells. Importantly, the migration front of repopulating NG2-glia invariably proceeds from the distal parenchyma towards the ventricles, ruling out contributions of the subventricular zone neurogenic niche or the corresponding area of the third ventricle as source of new NG2-glia. NG2-CreER-based fate tracing further substantiates that NG2-glia which have been spared from mitotic blockade are the sole source of regenerating NG2-glia. Collectively, our data reveals that all adult NG2-glia retain the ability to divide and that they are capable of fully restoring parenchymal NG2-glia coverage after wide-spread NG2 cell loss, indicating complete self-sufficiency in maintaining NG2-glia population levels in the adult brain.


Asunto(s)
Antígenos/metabolismo , Diferenciación Celular/fisiología , Neuroglía/fisiología , Proteoglicanos/metabolismo , Regeneración/fisiología , Animales , Antígenos/genética , Bromodesoxiuridina , Antígenos CD13/metabolismo , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/efectos de los fármacos , Citarabina/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Nestina/metabolismo , Proteoglicanos/genética , ARN Mensajero/metabolismo , Regeneración/efectos de los fármacos , Regeneración/genética , Tamoxifeno/toxicidad , Factores de Tiempo
12.
Chem Biol Interact ; 199(1): 1-8, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22640811

RESUMEN

The integrated discrete multiple organ co-culture system (IdMOC) allows the co-culturing of multiple cell types as physically separated cells interconnected by a common overlying medium. We report here the application of IdMOC with two cell types: the metabolically competent primary human hepatocytes, and a metabolically incompetent cell line, mouse 3T3 fibroblasts, in the definition of the role of hepatic metabolism on the cytotoxicity of three model toxicants: cyclophosphamide (CPA), aflatoxin B1 (AFB) and tamoxifen (TMX). The presence of hepatic metabolism in IdMOC with human hepatocytes was demonstrated by the metabolism of the P450 isoform 3A4 substrate, luciferin-IPA. The three model toxicants showed three distinct patterns of cytotoxic profile: TMX was cytotoxic to 3T3 cells in the absence of hepatocytes, with slightly lower cytotoxicity towards both 3T3 cells and hepatocytes in the IdMOC. AFB was selective toxic towards the human hepatocytes and relatively noncytotoxic towards 3T3 cells both in the presence and absence of the hepatocytes. CPA cytotoxicity to the 3T3 cells was found to be significantly enhanced by the presence of the hepatocytes, with the cytotoxicity dependent of the number of hepatocytes, and with the cytotoxicity attenuated by the presence of a non-specific P450 inhibitor, 1-aminobenzotriazole. We propose here the following classification of toxicants based on the role of hepatic metabolism as defined by the human hepatocyte-3T3 cell IdMOC assay: type I: direct-acting cytotoxicants represented by TMX as indicated by cytotoxicity in 3T3 cells in the absence of hepatocytes; type II: metabolism-dependent cytotoxicity represented by AFB1 with effects localized within the site of metabolic activation (i. e. hepatocytes); and type III: metabolism-dependent cytotoxicity with metabolites that can diffuse out of the hepatocytes to cause toxicity in cells distal from the site of metabolism, as exemplified by CPA.


Asunto(s)
Aflatoxina B1/toxicidad , Ciclofosfamida/toxicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Tamoxifeno/toxicidad , Xenobióticos/toxicidad , Células 3T3/efectos de los fármacos , Acetales/metabolismo , Aflatoxina B1/farmacocinética , Animales , Técnicas de Cocultivo , Ciclofosfamida/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Luciferina de Luciérnaga/análogos & derivados , Luciferina de Luciérnaga/metabolismo , Hepatocitos/citología , Humanos , Inactivación Metabólica , Concentración 50 Inhibidora , Ratones , Tamoxifeno/farmacocinética , Pruebas de Toxicidad/métodos , Triazoles/farmacología , Xenobióticos/farmacocinética
13.
J Obstet Gynaecol Res ; 37(7): 696-708, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21651672

RESUMEN

Emerging evidence suggests that adenomyosis, like endometriosis, may also be an epigenetic disease. In this study, we evaluated the effect of valproic acid (VPA) in ICR mice with adenomyosis, induced by neonatal dosing with tamoxifen. For all mice, we evaluated the bodyweight and the response to thermal stimuli by hotplate and tail-flick tests 4, 8, and 12 weeks after dosing, respectively, and then treated mice with low- and high-dose of VPA, progesterone (P4), P4 + VPA, or vehicle only. Three weeks after treatment, both bodyweight and thermal response tests were evaluated again before sacrifice, and the depth of myometrial infiltration was evaluated. We found that: (i) the induction of adenomyosis resulted in progressive generalized hyperalgesia as measured by hotplate and tail-flick tests, along with decreased bodyweight; (ii) treatment with VPA, P4, or a combination was efficacious in improving generalized hyperalgesia; and (iii) drug treatment appeared to reduce the myometrial infiltration, but the difference did not reach statistical significance. Thus, VPA seems to be a promising therapeutics for treating adenomyosis, as reported recently in some case series in humans.


Asunto(s)
Analgésicos no Narcóticos/uso terapéutico , Endometriosis/tratamiento farmacológico , Hiperalgesia/prevención & control , Tocolíticos/uso terapéutico , Ácido Valproico/uso terapéutico , Analgésicos no Narcóticos/administración & dosificación , Animales , Animales Recién Nacidos , Endometriosis/patología , Endometriosis/fisiopatología , Femenino , Hiperalgesia/etiología , Ratones , Ratones Endogámicos ICR , Miometrio/efectos de los fármacos , Miometrio/patología , Distribución Aleatoria , Tamoxifeno/toxicidad , Tocolíticos/administración & dosificación , Ácido Valproico/administración & dosificación
14.
Indian J Exp Biol ; 48(3): 280-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21046982

RESUMEN

Liver injury was induced in female rats using tamoxifen (TAM). Grape seeds (Vitis vinifera) extract (GSE), black seed (Nigella sativa) extract (NSE), curcumin (CUR) or silymarin (SYL) were orally administered to TAM-intoxicated rats. Liver histopathology of TAM-intoxicated:rats showed pathological changes. TAM-intoxication elicited declines in liver antioxidant enzymes levels (glutathione peroxidase, glutathione reductase, superoxide dismutase and catalase), reduced glutathione (GSH) and GSH/GSSG ratio plus the hepatic elevations in lipid peroxides, oxidized glutathione (GSSG), tumor necrosis factor-alpha (TNF-alpha) and serum liver enzymes; alanine transaminase, aspartate transaminase, alkaline phosphatase, lactate dehydrogenase and gamma glutamyl transferase levels. Oral intake of NSE, GSE, CUR or SYL to TAM-intoxicated rats, attenuated histopathological changes and corrected all parameters mentioned above. Improvements were prominent in case of NSE (similarly SYL) > CUR > GSE. Data indicated that NSE, GSE or CUR act as free radicals scavengers and protect TAM-induced liver injury in rats.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Curcumina/farmacología , Extractos Vegetales/farmacología , Semillas/química , Administración Oral , Alanina Transaminasa/sangre , Animales , Antiinflamatorios no Esteroideos/farmacología , Aspartato Aminotransferasas/sangre , Catalasa/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Curcumina/administración & dosificación , Femenino , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/metabolismo , Extracto de Semillas de Uva/administración & dosificación , Extracto de Semillas de Uva/farmacología , Peróxidos Lipídicos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Nigella sativa/química , Extractos Vegetales/administración & dosificación , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Tamoxifeno/toxicidad , Factor de Necrosis Tumoral alfa/sangre , Vitis/química
15.
Toxicol Lett ; 199(3): 416-24, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20937368

RESUMEN

Tamoxifen is an antiestrogenic agent used widely in the treatment of estrogen receptor-positive breast cancer. However, hepatic steatosis has been reported during clinical trials of tamoxifen. To explore the mechanism responsible for this tamoxifen-induced hepatic steatosis, we used microarray analysis to profile the gene expression pattern of mouse liver after tamoxifen treatment. Tamoxifen was administered orally as a single dose of 10mg/kg (low dose), 50mg/kg (medium dose), or 100mg/kg (high dose) to C57BL/6 mice, and the livers were removed 2h, 4h, 8h, and 24h later. From microarray data obtained from the liver samples, 414 genes were selected as tamoxifen-responsive genes (P<0.05, two-way ANOVA; cutoff ≥ 1.5-fold response). These genes were classified into three groups: 308 of the 414 genes showed a time-dependent response, nine genes showed a dose-dependent response, and 97 genes showed a time- and dose-dependent response. Most of the 308 time-dependent-responsive genes were associated predominantly with the biological processes involved in lipid metabolism. Overrepresented transcription factor binding site analysis showed that the following nuclear receptors that are important in lipid and carbohydrate metabolism were overrepresented: the androgen receptor (AR), nuclear receptor subfamily 2 group F member 1 (NR2F1), hepatocyte nuclear factor 4α (HNF4α), and retinoic acid receptor-related orphan receptor alpha 1 (RORα1). Reporter gene analysis further revealed that tamoxifen repressed the 5α-dihydrotestosterone-induced activation of the AR and the intrinsic transactivation function of RORα1, HNF4α, and NR2F1. Taken together, these data provide a better understanding of the molecular mechanism underlying tamoxifen-induced steatogenic hepatotoxicity and useful information for predicting steatogenic hepatotoxicity.


Asunto(s)
Hígado Graso/inducido químicamente , Perfilación de la Expresión Génica , Tamoxifeno/toxicidad , Animales , Colesterol/biosíntesis , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/genética , Activación Transcripcional/efectos de los fármacos
16.
Food Chem Toxicol ; 47(7): 1689-95, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19394397

RESUMEN

Tamoxifen (TAM) is widely used in the treatment and prevention of breast cancer. Adverse effects of TAM include hepatotoxicity. Caffeic acid phenethyl ester (CAPE), an active component of propolis, has been used in folk medicine for diverse ailments. In the current study, the protective effects of CAPE against TAM-induced hepatotoxicity in female rats were evaluated. TAM (45 mg/kg/day, i.p., for 10 consecutive days) resulted in an elevation of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP), depletion of liver reduced glutathione (GSH) and accumulation of oxidized glutathione (GSSG) and lipid peroxidation (LPO). Also, TAM treatment resulted in inhibition of hepatic activity of glutathione reductase (GR), glutathione peroxidase (GPx), superoxide dismutase (SOD) and catalase (CAT). Further, it raised liver tumor necrosis factor-alpha (TNF-alpha) level and induced histopathological changes. Pretreatment with CAPE (2.84 mg/kg/day; i.p., for 20 consecutive days, starting 10 days before TAM injection) significantly prevented the elevation in serum activity of the assessed enzymes. CAPE significantly inhibited TAM-induced hepatic GSH depletion and GSSG and LPO accumulation. Consistently, CAPE normalized the activity of GR, GPx, SOD and CAT, inhibited the rise in TNF-alpha and ameliorated the histopathological changes. In conclusion, CAPE protects against TAM-induced hepatotoxicity.


Asunto(s)
Antineoplásicos Hormonales/antagonistas & inhibidores , Antineoplásicos Hormonales/toxicidad , Ácidos Cafeicos/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Alcohol Feniletílico/análogos & derivados , Tamoxifeno/antagonistas & inhibidores , Tamoxifeno/toxicidad , Animales , Antioxidantes/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Pruebas de Función Hepática , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Alcohol Feniletílico/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa/metabolismo , Xantina Oxidasa/metabolismo
17.
Toxicology ; 225(2-3): 109-18, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16797820

RESUMEN

Natural antioxidants like catechin are now known to have a modulatory role on physiological functions and biotransformation reactions involved in the detoxification process, thereby affording protection from toxic metabolic actions of xenobiotics. Reactive oxygen intermediates have been demonstrated to play an etiological role in anticancer drug-induced toxicity. This study was performed to explore the modulatory and protective effect of catechin on the toxicity of an anticancer drug, tamoxifen (TAM) with special reference to protection against disruption of glutathione metabolizing and antioxidant enzymes. TAM treatment resulted in a significant increase in the lipid peroxidation (LPO), H(2)O(2) generation and protein carbonyl (PC) contents in the liver and kidney as compared to controls while catechin+TAM-treated group showed significant decrease in LPO levels, H(2)O(2) generation and PC contents in liver and kidney when compared with TAM-treated group. Non-enzymatic antioxidants like reduced glutathione (GSH) and low molecular antioxidants like ascorbic acid (AsA) also showed normalcy due to exogenous catechin administration. Catechin pre-treatment showed restoration in the level of cytochrome P450 (CYP) content and in the activities of glutathione metabolizing enzymes, viz., glutathione-S-transferase (GST), glutathione reductase (GR) and glutathione peroxidase (GPx) and other antioxidant enzymes such as, glucose-6-phosphate dehydrogenase (G6-PD), catalase (CAT) and superoxide dismutase (SOD) in both liver and kidney when compared to TAM-treated animals. The results of the study show that catechin supplementation might be helpful in abrogation of TAM toxicity during chemotherapy. Additionally, it makes it a prophylactic and preventive agent of anticancer drug-induced oxidative stress.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Antioxidantes/farmacología , Catequina/farmacología , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Tamoxifeno/toxicidad , Animales , Ácido Ascórbico/metabolismo , Antagonismo de Drogas , Quimioterapia Combinada , Enzimas/metabolismo , Glutatión/metabolismo , Peróxido de Hidrógeno/metabolismo , Riñón/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Carbonilación Proteica/efectos de los fármacos
18.
J Biochem Mol Biol ; 38(5): 563-70, 2005 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16202236

RESUMEN

Tamoxifen citrate (TAM), is widely used for treatment of breast cancer. It showed a degree of hepatic carcinogenesis. The purpose of this study was to elucidate the antioxidant capacity of green tea (Camellia sinensis) extract (GTE) against TAM-induced liver injury. A model of liver injury in female rats was done by intraperitoneal injection of TAM in a dose of 45mg Kg(-1) day(-1), i.p. for 7 successive days. GTE in the concentration of 1.5 %, was orally administered 4 days prior and 14 days after TAM-intoxication as a sole source of drinking water. The antioxidant flavonoid; epicatechin (a component of green tea) was not detectable in liver and blood of rats in either normal control or TAM-intoxicated group, however, TAM intoxication resulted in a significant decrease of its level in liver homogenate of tamoxifenintoxicated rats. The model of TAM-intoxication elicited significant declines in the antioxidant enzymes (glutathione-S-transferase,glutathione peroxidase, superoxide dismutase and catalase) and reduced glutathione concomitant with significant elevations in TBARS (thiobarbituric acid reactive substance) and liver transaminases; sGPT (serum glutamate pyruvate transaminase) and sGOT (serum glutamate oxaloacetate transaminase) levels. The oral administration of 1.5 % GTE to TAM-intoxicated rats, produced significant increments in the antioxidant enzymes and reduced glutathione concomitant with significant decrements in TBARS and liver transaminases levels. The data obtained from this study speculated that 1.5 % GTE has the capacity to scavenge free radical and can protect against oxidative stress induced by TAM intoxication. Supplementation of GTE could be useful in alleviating tamoxifen-induced liver injury in rats.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , Hígado/patología , Extractos Vegetales/farmacología , Tamoxifeno/toxicidad , Té/química , Animales , Antioxidantes/metabolismo , Camellia sinensis/química , Catequina/análisis , Enfermedad Hepática Inducida por Sustancias y Drogas , Femenino , Hígado/química , Hígado/enzimología , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Distribución Aleatoria , Ratas , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
19.
Arch Toxicol ; 77(9): 487-99, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12802581

RESUMEN

The main objective of this 28-day oral gavage toxicity study in the rat was to investigate which of the current and/or additional parameters of the OECD Test Guideline 407 would reliably and sensitively detect the endocrine-mediated effects of the nonsteroidal antiestrogen tamoxifen. In addition, as this study was performed using two subgroups of five animals of each sex run concurrently, it enabled an assessment of the intralaboratory reproducibility while also assessing the potential value of using ten animals of each sex per group instead of using the standard five animals of each sex per group stipulated by the current guideline. Tamoxifen was administered daily by gavage to groups of 7-week-old Wistar rats for at least 28 days at dose levels of 5, 30, or 200 microg/kg body weight. Additional parameters specified in the enhanced OECD Test Guideline 407 were spermatozoa enumeration and morphology of the cauda epididymis, hormonal analysis of the thyroid-stimulating hormone (TSH), triiodothyronine (T3) and thyroxine (T4) levels, monitoring of the estrous cycle during week 4 of treatment to ensure females were in diestrus on the day of terminal sacrifice, organ weight of ovary, uterus, thyroid gland, prostate gland (ventral and dorsolateral parts), seminal vesicles with coagulation glands and pituitary gland, and microscopic investigation of the pituitary gland, vagina, mammary gland, seminal vesicles with coagulating glands, epididymis, and prostate gland (ventral and dorsolateral parts). Overall, 200 microg/kg per day was considered to be the Maximum Tolerated Dose (MTD) in both sexes, resulting in a marked reduction of body weight gain, together with slight effects on clinical signs, hematology, plasma chemistry, and microscopic changes in some endocrine tissues. Five micrograms per kilogram per day represented the No Observed Adverse Effect Level (NOAEL) in males and the No Observed Effect Level (NOEL) in females. At the intermediate dose level (30 microg/kg per day), the current OECD Test Guideline 407 was appropriate to detect the specific endocrine-related changes induced by tamoxifen in females, based on the histopathology findings observed in the ovary and the uterus. The additional parameters which were found to be changed in females (thyroid hormone levels, ovary and uterus weights, and histopathology of vagina) provided supplementary information further confirming tamoxifen-mediated endocrine effects. In males, when data from the current Test Guideline 407 were considered at the intermediate dose level, specific endocrine effects were only indicated on the basis of the histopathology findings observed in the prostate gland. The additional parameters examined which were found to be changed (prostate gland and seminal vesicle weights, and histopathology of seminal vesicle and mammary gland) were necessary to confirm the specific tamoxifen-mediated endocrine effects. Hence, amongst the additional parameters contained in the enhanced OECD Test Guideline 407, organ weights and histopathological examination of endocrine-related organs were the most helpful in confirming the detection of tamoxifen-mediated endocrine effects. The reproducibility evaluation showed that a group size of five animals of each sex consistently allowed the detection of endocrine effects with the current Test Guideline in both sexes at the high dose level and in females at the intermediate dose level. Doubling the animal number from five to ten of each sex per dose level did not notably increase the sensitivity of detection of endocrine-mediated effects.


Asunto(s)
Sistema Endocrino/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/toxicidad , Tamoxifeno/toxicidad , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Femenino , Guías como Asunto , Masculino , Tamaño de los Órganos/efectos de los fármacos , Tiempo de Protrombina , Ratas , Ratas Wistar , Espermatozoides/efectos de los fármacos , Hormonas Tiroideas/sangre , Pruebas de Toxicidad Crónica
20.
Toxicology ; 179(3): 221-32, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12270594

RESUMEN

The use of tamoxifen (TAM) has been questioned on the chemotherapy and chemoprevention of breast cancer due to several estrogen receptor-independent cytotoxic effects. As an alternative, its more active metabolite 4-hydroxytamoxifen (OHTAM) has been proposed with presumed lower side effects. In this work, the potential OHTAM toxicity on rat liver mitochondrial bioenergetics in relation to the multiple deleterious effects of TAM was evaluated. OHTAM, at concentrations lower than those putatively reached in tissues following the administration of TAM, does not induce significant perturbations on the respiratory control ratio (RCR), ADP/O, transmembrane potential (DeltaPsi), phosphorylative capacity and membrane integrity of mitochondria. However, at high concentrations, OHTAM depresses the DeltaPsi, RCR and ADP/O, affecting the phosphorylation efficiency, as also inferred from the DeltaPsi fluctuations and pH changes associated with ADP phosphorylation. Moreover, OHTAM, at concentrations that stimulate the rate of state 4 respiration in parallel to the decrease in the DeltaPsi and phosphorylation rate, causes mitochondrial swelling and stimulates both ATPase and citrate synthase activities. However, the OHTAM-observed effects, at high concentrations, are not significant relatively to the damaging effects promoted by TAM and suggest alterations to mitochondrial functions due to proton leak across the mitochondrial inner membrane.


Asunto(s)
Mitocondrias Hepáticas/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Tamoxifeno/análogos & derivados , Tamoxifeno/toxicidad , Desacopladores/metabolismo , Nucleótidos de Adenina/metabolismo , Adenosina Trifosfatasas/efectos de los fármacos , Animales , Citrato (si)-Sintasa/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Femenino , Concentración de Iones de Hidrógeno/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Dilatación Mitocondrial/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA