Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34911761

RESUMEN

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Asunto(s)
Hipertensión Pulmonar/patología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular/fisiología , Animales , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión Pulmonar/metabolismo , Isquemia , Quinasa 1 de Quinasa de Quinasa MAP/genética , MAP Quinasa Quinasa Quinasa 3/genética , Ratones , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Transducción de Señal , Tamoxifeno/toxicidad , Factor de Crecimiento Transformador beta/genética
2.
Toxicol Appl Pharmacol ; 475: 116627, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37453479

RESUMEN

SMARCA2 and SMARCA4 are the ATPases of the SWI/SNF chromatin remodeling complex, which play a significant role in regulating transcriptional activity and DNA repair in cells. SMARCA2 has become an appealing synthetic-lethal, therapeutic target in oncology, as mutational loss of SMARCA4 in many cancers leads to a functional dependency on residual SMARCA2 activity. Thus, for therapeutic development, an important step is understanding any potential safety target-associated liabilities of SMARCA2 inhibition. To best mimic a SMARCA2 therapeutic, a tamoxifen-inducible (TAMi) conditional knockout (cKO) rat was developed using CRISPR technology to understand the safety profile of Smarca2 genetic ablation in a model system that avoids potential juvenile and developmental phenotypes. As the rat is the prototypical rodent species utilized in toxicology studies, a comprehensive toxicological and pathological assessment was conducted in both heterozygote and homozygous knockout rats at timepoints up to 28 days, alongside relevant corresponding controls. To our knowledge, this represents the first TAMi cKO rat model utilized for safety assessment evaluations. No significant target-associated phenotypes were observed when Smarca2 was ablated in mature (11- to 15-week-old) rats; however subsequent induction of SMARCA4 was evident that could indicate potential compensatory activity. Similar to mouse models, rat CreERT2-transgene and TAMi toxicities were characterized to avoid confounding study interpretation. In summary, a lack of significant safety findings in Smarca2 cKO rats highlights the potential for therapeutics targeting selective SMARCA2 ATPase activity; such therapies are predicted to be tolerated in patients without eliciting significant on-target toxicities.


Asunto(s)
Neoplasias , Tamoxifeno , Ratones , Ratas , Animales , Tamoxifeno/toxicidad , Adenosina Trifosfatasas , Mutación
3.
Toxicol Appl Pharmacol ; 442: 116002, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35353989

RESUMEN

Tamoxifen is an effective breast cancer therapy in postmenopausal women. However, it can induce hyperglycemia through different mechanisms, such as the impairment of mitochondrial metabolism. Quercetin, a flavonoid with antioxidant potential, has beneficial effects on tamoxifen-induced adverse effects. Therefore, this study aimed to (1) investigate glucose concentration in blood, cerebrospinal fluid, cerebellum, cortex, and hippocampus of tamoxifen-treated ovariectomized female rats, non-treated and treated with quercetin; and (2) establish the metabolic profile of these regions. For that purpose, ovariectomized female rats were divided into four groups: canola oil 1 mL/kg (CONT); tamoxifen 5 mg/kg (TAM); quercetin 22.5 mg/kg (QUER); and tamoxifen 5 mg/kg + quercetin 22.5 mg/kg (TAM + Q); and were treated for 14 days orally. Subsequently, glucose levels were measured in blood, cerebrospinal fluid, cerebellum, cortex, and hippocampus. Pyruvate and lactate concentrations were analyzed in the three brain regions. Tamoxifen-induced hyperglycemia significantly increased glucose concentrations in the cerebrospinal fluid, cortex, and hippocampus, as well as lactate production in the hippocampus. Quercetin significantly prevented the tamoxifen-induced increase in glucose concentrations in all analyzed samples. Besides, quercetin decreased cortical pyruvate production. The copper content decreased only in the hippocampus of group TAM + Q animals. In addition, it is important to highlight that this study also observed that fourteen days of tamoxifen treatment strongly affects brain glucose metabolism, potentially disrupting normal brain functions. Therefore, this drug might represent a risk factor for postmenopausal women undergoing chemoprevention. Meanwhile, quercetin represents a potential intervention to promote metabolic regulation of glucose in tamoxifen-treated women.


Asunto(s)
Hiperglucemia , Tamoxifeno , Animales , Modelos Animales de Enfermedad , Femenino , Glucosa , Hipocampo , Humanos , Hiperglucemia/inducido químicamente , Ácido Láctico , Posmenopausia , Ácido Pirúvico , Quercetina , Ratas , Tamoxifeno/toxicidad
4.
Gut ; 70(10): 1833-1846, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33293280

RESUMEN

OBJECTIVE: Tissue stem cells are central regulators of organ homoeostasis. We looked for a protein that is exclusively expressed and functionally involved in stem cell activity in rapidly proliferating isthmus stem cells in the stomach corpus. DESIGN: We uncovered the specific expression of Iqgap3 in proliferating isthmus stem cells through immunofluorescence and in situ hybridisation. We performed lineage tracing and transcriptomic analysis of Iqgap3 +isthmus stem cells with the Iqgap3-2A-tdTomato mouse model. Depletion of Iqgap3 revealed its functional importance in maintenance and proliferation of stem cells. We further studied Iqgap3 expression and the associated gene expression changes during tissue repair after tamoxifen-induced damage. Immunohistochemistry revealed elevated expression of Iqgap3 in proliferating regions of gastric tumours from patient samples. RESULTS: Iqgap3 is a highly specific marker of proliferating isthmus stem cells during homoeostasis. Iqgap3+isthmus stem cells give rise to major cell types of the corpus unit. Iqgap3 expression is essential for the maintenance of stem potential. The Ras pathway is a critical partner of Iqgap3 in promoting strong proliferation in isthmus stem cells. The robust induction of Iqgap3 expression following tissue damage indicates an active role for Iqgap3 in tissue regeneration. CONCLUSION: IQGAP3 is a major regulator of stomach epithelial tissue homoeostasis and repair. The upregulation of IQGAP3 in gastric cancer suggests that IQGAP3 plays an important role in cancer cell proliferation.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Mucosa Gástrica/citología , Homeostasis/fisiología , Células Madre/citología , Neoplasias Gástricas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neoplasias Gástricas/tratamiento farmacológico , Tamoxifeno/toxicidad
5.
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
6.
Basic Res Cardiol ; 116(1): 8, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33544211

RESUMEN

Conditional, cell-type-specific transgenic mouse lines are of high value in cardiovascular research. A standard tool for cardiomyocyte-restricted DNA editing is the αMHC-MerCreMer/loxP system. However, there is an ongoing debate on the occurrence of cardiac side effects caused by unspecific Cre activity or related to tamoxifen/oil overload. Here, we investigated potential adverse effects of DNA editing by the αMHC-MerCreMer/loxP system in combination with a low-dose treatment protocol with the tamoxifen metabolite 4-hydroxytamoxifen (OH-Txf). αMHC-MerCreMer mice received intraperitoneally OH-Txf (20 mg/kg) for 5 or 10 days. These treatment protocols were highly efficient to induce DNA editing in adult mouse hearts. Multi-parametric magnetic resonance imaging revealed neither transient nor permanent effects on cardiac function during or up to 19 days after 5 day OH-Txf treatment. Furthermore, OH-Txf did not affect cardiac phosphocreatine/ATP ratios assessed by in vivo 31P MR spectroscopy, indicating no Cre-mediated side effects on cardiac energy status. No MRI-based indication for the development of cardiac fibrosis was found as mean T1 relaxation time was unchanged. Histological analysis of myocardial collagen III content after OH-Txf confirmed this result. Last, mean T2 relaxation time was not altered after Txf treatment suggesting no pronounced cardiac lipid accumulation or tissue oedema. In additional experiments, cardiac function was assessed for up to 42 days to investigate potential delayed side effects of OH-Txf treatment. Neither 5- nor 10-day treatment resulted in a depression of cardiac function. Efficient cardiomyocyte-restricted DNA editing that is free of unwanted side effects on cardiac function, energetics or fibrosis can be achieved in adult mice when the αMHC-MerCreMer/loxP system is activated by the tamoxifen metabolite OH-Txf.


Asunto(s)
Edición Génica , Integrasas/genética , Miocitos Cardíacos/efectos de los fármacos , Tamoxifeno/análogos & derivados , Animales , Metabolismo Energético/efectos de los fármacos , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tamoxifeno/farmacología , Tamoxifeno/toxicidad , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Environ Res ; 197: 111121, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823193

RESUMEN

Endoxifen is the main active metabolite of a common cytostatic drug, tamoxifen. Endoxifen has been recently detected in the final effluent of municipal wastewater treatment plants. The antiestrogenic activity of endoxifen could bring negative effects to aquatic life if released to the water environment. This study elucidated the fate and susceptibility of (E)- and (Z)-endoxifen (2 µg mL-1, 1:1 wt ratio between the two easily interchangeable isomers) in wastewater and receiving surface water to sunlight. Phototransformation by-products (PBPs) and their toxicity were determined. Sunlight reduced at least 83% of endoxifen concentration in wastewater samples, whereas in surface water samples, 60% of endoxifen was photodegraded after 180 min of the irradiation. In ultrapure water samples spiked with endoxifen, PBPs were mainly generated via con-rotatory 6π-photocyclization, followed by oxidative aromatization. These PBPs underwent secondary reactions leading to a series of PBPs with different molecular weights. Eight PBPs were identified and the toxicity analysis via the Toxicity Estimation Software Tool revealed that seven of these PBPs are more toxic than endoxifen itself. This is likely due to the formation of poly-aromatic core in the PBPs due to exposure to sunlight. Therefore, highly toxic PBPs may be generated if endoxifen is present in water and wastewater exposed to sunlight. The presence, fates and activities of these PBPs in surface water especially at locations close to treated wastewater discharge points should be investigated.


Asunto(s)
Neoplasias de la Mama , Aguas Residuales , Femenino , Humanos , Luz Solar , Tamoxifeno/análogos & derivados , Tamoxifeno/toxicidad , Agua
8.
FASEB J ; 33(12): 14067-14082, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31657630

RESUMEN

Glucocorticoids (GCs) are important hormones involved in the regulation of multiple physiologic functions. GCs are also widely used in anti-inflammatory/immunosuppressant drugs. GCs are synthesized by the adrenal cortex as part of the hypothalamus-pituitary-adrenal axis and also by intestinal epithelial cells, among other peripheral sites. GCs are one of the main therapy choices for the exacerbations of inflammatory bowel disease, but they are not useful to prolong remission, and development of tolerance with secondary treatment failure is frequent. Thus, GC actions at the intestinal epithelial level are of great importance, both physiologically and pharmacologically. We generated a tamoxifen-inducible nuclear receptor subfamily 3 group C member 1 (NR3C1)ΔIEC mouse model to study the effects of GCs on epithelial cells in vivo. Nr3c1 deletion in epithelial cells of the small intestine and colon was associated with limited colonic inflammation at 1 wk postdeletion, involving augmented epithelial proliferation and mucus production, plus local and systemic immune/inflammatory changes. This phenotype regressed substantially, but not completely, after 2 wk. The mechanism may involve augmented inflammatory signaling by epithelial cells or defective barrier function. We conclude that the epithelial GC receptor plays a significant role in colonic homeostasis in basal conditions, but its deficiency can be compensated in the short term. Future studies are required to assess the impact of Nr3c1 deletion in other conditions such as experimental colitis.-Aranda, C. J., Arredondo-Amador, M., Ocón, B., Lavín, J. L., Aransay, A. M., Martínez-Augustin, O., Sánchez de Medina, F. Intestinal epithelial deletion of the glucocorticoid receptor NR3C1 alters expression of inflammatory mediators and barrier function.


Asunto(s)
Células Epiteliales/metabolismo , Inflamación/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Antagonistas de Estrógenos/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores de Glucocorticoides/genética , Tamoxifeno/toxicidad
9.
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
10.
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
11.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G518-G530, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31369292

RESUMEN

The sodium-dependent multivitamin transporter (SMVT; SLC5A6) is involved in intestinal absorption of vitamin B7 (biotin). We have previously shown that mice with an embryonic intestinal-specific SMVT knockout (KO) develop biotin deficiency and severe spontaneous intestinal inflammation in addition to growth retardation, developmental delays, and death within the first 6-7 wk of life. The profound morbidity and mortality associated with the SMVT-KO has limited our ability to further characterize the intestinal inflammation and other sequelae of this deletion in adult mice with a mature gut microbiota. To overcome this limitation, we generated an intestine-specific, tamoxifen-inducible, conditional SMVT-KO (SMVT-icKO). Our results showed that adult SMVT-icKO mice have reduced body weight, biotin deficiency, shorter colonic length, and bloody diarrhea compared with age- and sex-matched control littermates. All SMVT-icKO mice also developed spontaneous intestinal inflammation associated with induction of calprotectin (S100a8/S100a9), proinflammatory cytokines (IL-1ß, TNF-α, IFN-γ, and IL-6), and an increase in intestinal permeability. Additionally, the intestines of SMVT-icKO showed activation of the NF-κB pathway and the nucleotide-binding domain and leucine-rich repeat pyrin 3 domain (NLRP3) inflammasome. Notably, administration of broad-spectrum antibiotics reduced lethality and led to normalization of intestinal inflammation, proinflammatory cytokines, altered mucosal integrity, and reduced expression of the NLRP3 inflammasome. Overall, these findings support our conclusion that the biotin transport pathway plays an important role in the maintenance of intestinal homeostasis, and that NF-κB and the NLRP3 inflammasome, as well as gut microbiota, drive the development of intestinal inflammation when SMVT is absent.NEW & NOTEWORTHY This study demonstrates that deletion of the intestinal biotin uptake system in adult mice leads to the development of spontaneous gut inflammation and that luminal microbiota plays a role in its development.


Asunto(s)
Enteritis/genética , Antagonistas de Estrógenos/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Simportadores/metabolismo , Tamoxifeno/toxicidad , Envejecimiento , Animales , Biotina/metabolismo , Peso Corporal/efectos de los fármacos , Colon/patología , Citocinas/metabolismo , Diarrea/inducido químicamente , Diarrea/microbiología , Diarrea/patología , Enteritis/inducido químicamente , Enteritis/microbiología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Simportadores/efectos de los fármacos , Simportadores/genética
12.
Toxicol Appl Pharmacol ; 363: 88-97, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30503537

RESUMEN

The endometrium is a particular sensitive target tissue for estradiol that is able to promptly modify its structure. Tamoxifen (TAM), a selective estrogen receptor modulator, was shown to promote a spectrum of uterine abnormalities, though the morphological and stereological effects of this drug in uterus is not clear. In this way, we have used an established model of ovariectomy followed by estradiol benzoate (EB) or TAM treatment and analyzed their effects in uterine histopathology and proliferation. Administration of EB promotes the unfolding and proliferation of the endometrium stroma, increasing uterine volume. No changes were found in uterine histomorphometric analysis upon TAM administration, except in the thickness of the luminal epithelium and endometrium layer. The latter may result from increased complexity and glandular volume density also observed in TAM treatment. In addition, EB induced PAX2 expression, an oncogene commonly found in epithelial tumors of the female genital tract, an effect that was weakened by previous TAM administration. Although treatments did not affect stroma cells proliferating index, in epithelial cells and, contrary to TAM, EB increased PCNA and not Ki67 expression. Collectively, our data suggest that the acute administration of TAM induces ERα-dependent atrophy of the uterine tissue and decreased the expression of proliferating cellular markers. On the contrary, if administered prior to EB, TAM is able to attenuate the action of the hormone in uterine morphology and biochemistry.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Tamoxifeno/toxicidad , Útero/patología , Animales , Atrofia/inducido químicamente , Atrofia/patología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Estradiol/administración & dosificación , Estradiol/análogos & derivados , Receptor alfa de Estrógeno/metabolismo , Ciclo Estral/metabolismo , Femenino , Antígeno Ki-67/metabolismo , Ovariectomía , Factor de Transcripción PAX2/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar , Pruebas de Toxicidad Aguda , Útero/efectos de los fármacos
13.
Exp Eye Res ; 180: 39-42, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30500365

RESUMEN

The RAS gene family members, H-RAS, K-RAS, and N-RAS, are frequently mutated in human cancer. A subset of retinal tumors displays K-RAS mutations; however, the specific role of RAS activation on retinal tumor formation is unclear. To examine the role of RAS in retinal development, we overexpressed the mutant H-RAS gene (G12V) in retinal progenitor cells (RPCs), a multipotent progenitor cell population that gives rise to all six neuron types in the retina and to the Muller glia. The Msi1CreER mouse strain was used to induce mosaic activation of Ras (RasV12) in the RPCs of the postnatal retina. RAS-activated RPCs translocated to the basal part of the retina, differentiated into cells with glial characteristics, and underwent apoptosis. We next induced RAS activation in a large population of RPCs in the embryonic retina using the Pax6Cre mouse strain. In contrast to the phenotype observed in Msi1CreER;RasV12 mice, Ras-activated cells retained their apical attachment. Basal translocation was partially suppressed in the retina of Pax6Cre;RasV12 mice, indicating that basal translocation of Ras-activated cells was not cell autonomous. Notably, RAS-activated retinal cells were highly proliferative and promoted the formation of eye tumors in Pax6Cre;RasV12 mice. Together, our data indicate that the tumorigenicity of RAS activation in RPCs is context dependent, with tumor formation occurring when RAS activity is present in a large cluster of embryonic RPCs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Neoplasias del Ojo/patología , Regulación de la Expresión Génica/fisiología , Genes ras/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Antagonistas de Estrógenos/toxicidad , Neoplasias del Ojo/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción PAX6/genética , Tamoxifeno/toxicidad
14.
Arterioscler Thromb Vasc Biol ; 38(6): 1346-1357, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29650694

RESUMEN

OBJECTIVE: Estrogens exert beneficial effect on the blood vascular system. However, their role on the lymphatic system has been poorly investigated. We studied the protective effect of the 17ß estradiol-the most potent endogenous estrogen-in lymphedema-a lymphatic dysfunction, which results in a massive fluid and fat accumulation in the limb. APPROACH AND RESULTS: Screening of DNA motifs able to mobilize ERs (estrogen receptors) and quantitative real-time polymerase chain reaction analysis revealed that estradiol promotes transcriptional activation of lymphangiogenesis-related gene expression including VEGF (vascular endothelial growth factor)-D, VEGFR (VEGF receptor)-3, lyve-1, and HASs (hyaluronan synthases). Using an original model of secondary lymphedema, we observed a protective effect of estradiol on lymphedema by reducing dermal backflow-a representative feature of the pathology. Blocking ERα by tamoxifen-the selective estrogen modulator-led to a remodeling of the lymphatic network associated with a strong lymphatic leakage. Moreover, the protection of lymphedema by estradiol treatment was abrogated by the endothelial deletion of the receptor ERα in Tie2-Cre; ERαlox/lox mice, which exhibit dilated lymphatic vessels. This remodeling correlated with a decrease in lymphangiogenic gene expression. In vitro, blocking ERα by tamoxifen in lymphatic endothelial cells decreased cell-cell junctions, inhibited migration and sprouting, and resulted in an inhibition of Erk but not of Akt phosphorylation. CONCLUSIONS: Estradiol protection from developing lymphedema is mediated by an activation of its receptor ERα and is antagonized by tamoxifen. These findings reveal a new facet of the estrogen influence in the management of the lymphatic system and provide more evidence that secondary lymphedema is worsened by hormone therapy.


Asunto(s)
Linfedema del Cáncer de Mama/prevención & control , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/agonistas , Terapia de Reemplazo de Hormonas , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Linfedema del Cáncer de Mama/metabolismo , Linfedema del Cáncer de Mama/patología , Linfedema del Cáncer de Mama/fisiopatología , Modelos Animales de Enfermedad , Implantes de Medicamentos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Vasos Linfáticos/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Fosforilación , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Tamoxifeno/toxicidad
15.
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
16.
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
17.
Cell Mol Biol Lett ; 23: 3, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29308069

RESUMEN

Endoxifen, an active metabolite of tamoxifen, has been shown to be an effective anti-estrogenic agent in estrogen receptor-positive breast cancer patients. In melanoma, estrogen receptor expression is shown to be associated with disease progression. However, the therapeutic benefit of endoxifen in melanoma has not yet been evaluated. Here, we present the first demonstration of the anti-melanogenic activity of endoxifen in vitro and in vivo. The in vitro cytotoxic effect of endoxifen was tested using a cell viability assay. The in vivo anti-melanogenic activity was evaluated in B16F10 cell-bearing C57BL/6 mice, a mouse melanoma model. The general toxicity was tested in Swiss albino mice. Endoxifen exhibited greater activity against melanoma cell lines. Treatment of B16F10 mouse and SK-MEL-5 human melanoma cell lines with 10 µM of endoxifen for 48 h respectively resulted in 93.6 and 92.5% cell death. Orally administered endoxifen, at dose levels of 4 and 8 mg/kg body weight/day for 20 consecutive days, respectively reduced metastatic melanoma nodules in the lungs by 26.7 and 82.7%. Endoxifen was found to be a safe and effective anti-melanogenic agent in animal studies.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Tamoxifeno/análogos & derivados , Administración Oral , Animales , Antineoplásicos/toxicidad , Línea Celular Tumoral , Femenino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Tamoxifeno/administración & dosificación , Tamoxifeno/uso terapéutico , Tamoxifeno/toxicidad
18.
Ecotoxicol Environ Saf ; 148: 652-663, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29156432

RESUMEN

The constant release of pharmaceuticals products to aquatic environment even at low concentrations (ng L-1 to µg L-1) could lead to unknown chronic effects to non-target organisms. The aim of this study was to evaluate neurotoxic responses, inflammation, gametogenic activity and energy status on the fresh water clam C. fluminea after exposure to different concentrations of caffeine (CAF), ibuprofen (IBU), carbamazepine (CBZ), novobiocin (NOV) and tamoxifen (TMX) for 21 days under laboratory conditions. During the assay, water was spiked every two days with CAF (0; 0.1; 5; 15; 50µgL-1), IBU (0; 0.1; 5; 10; 50µgL-1), CBZ, NOV, and TMX (0.1, 1, 10, 50µgL-1). After the exposure period, dopamine levels (DOP), monoamine oxidase activity (MAO), arachidonic acid cyclooxygenase activity (COX), vitellogenin-like proteins (VTG), mitochondrial electron transport (MET), total lipids (TLP), and energy expenditure (MET/TLP) were determined in gonad tissues, and acetyl cholinesterase activity (AChE) was determined in digestive gland tissues. Results showed a concentration-dependence response on biomarkers tested, except for MAO. Environmental concentrations of pharmaceuticals induced significant changes (p < 0.05) in the neurotoxic responses analyzed (CAF, CBZ and NOV increased DOP levels and CBZ inhibited AChE activity), inflammation (CAF induced COX), and energy status (MET and TLP increased after exposure to CBZ, NOV and TMX). Responses of clams were related to the mechanism of action (MoA) of pharmaceuticals. Biomarkers applied and the model organism C. fluminea constituted a suitable tool for environmental risk assessment of pharmaceutical in aquatic environment.


Asunto(s)
Cafeína/toxicidad , Carbamazepina/toxicidad , Corbicula/fisiología , Ibuprofeno/toxicidad , Novobiocina/toxicidad , Tamoxifeno/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Biomarcadores/metabolismo , Corbicula/metabolismo , Metabolismo Energético , Células Germinativas , Humanos , Inflamación , Neurotoxinas
19.
Environ Monit Assess ; 190(11): 677, 2018 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-30368600

RESUMEN

Although medicines are less toxic than other toxicants, increased production and usage of pharmaceuticals have led to many concerns regarding their toxic effects on human and non-target organisms. Additionally, reproductive toxicity after long-term exposure is difficult to anticipate. Tamoxifen (TAM), a selective estrogen receptor modulator, has been widely used as an anticancer drug for mammalian breast and endometrial cancers. With increased TAM usage, it has frequently been reported that TAM is a potential endocrine disruptor capable of interfering with reproduction in non-target organisms. However, the mode of action of TAM in the endocrine system is unknown. In this study, we performed a 21-day chronic toxicity test using the crustacean Daphnia magna and investigated the transcriptional modulation of major genes related to the endocrine system, molting, development, and reproduction (i.e., Dm-vtg2, vmo1, cyp314, usp, and ecrb) after TAM exposure for 3, 6, 12, and 24 h. Our results showed a concentration-dependent decrease in the total number of offspring per individual, except for the concentration 25 µg/L; additionally, the expression of oogenesis-related genes was induced early but was later inhibited by TAM exposure. Additionally, molting-related genes were also downregulated in a time-dependent manner. Our findings suggested that TAM regulates reproduction by interfering with the molecular mechanisms involved in oogenesis and molting. This study supports the hypothesis that D. magna are a useful model to rapidly evaluate the reproductive effects of pharmaceuticals.


Asunto(s)
Daphnia/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Reproducción/efectos de los fármacos , Tamoxifeno/toxicidad , Animales , Monitoreo del Ambiente/métodos , Femenino , Pruebas de Toxicidad Crónica , Contaminantes Químicos del Agua/toxicidad
20.
Bioconjug Chem ; 28(12): 2942-2953, 2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29083862

RESUMEN

Weakly basic drugs display poor solubility and tend to precipitate in the stomach's acidic environment causing reduced oral bioavailability. Tracing of these orally delivered therapeutic agents using molecular probes is challenged due to their poor absorption in the gastrointestinal tract (GIT). Therefore, we designed a gastric pH stable bile acid derived amphiphile where Tamoxifen (as a model anticancer drug) is conjugated to lithocholic acid derived phospholipid (LCA-Tam-PC). In vitro studies suggested the selective nature of LCA-Tam-PC for cancer cells over normal cells as compared to the parent drug. Fluorescent labeled version of the conjugate (LCA-Tam-NBD-PC) displayed an increased intracellular uptake compared to Tamoxifen. We then investigated the antitumor potential, toxicity, and median survival in 4T1 tumor bearing BALB/c mice upon LCA-Tam-PC treatment. Our studies confirmed a significant reduction in the tumor volume, tumor weight, and reduced hepatotoxicity with a significant increase in median survival on LCA-Tam-PC treatment as compared to the parent drug. Pharmacokinetic and biodistribution studies using LCA-Tam-NBD-PC witnessed the enhanced gut absorption, blood circulation, and tumor site accumulation of phospholipid-drug conjugate leading to improved antitumor activity. Therefore, our studies revealed that conjugation of chemotherapeutic/imaging agents to bile acid phospholipid can provide a new platform for oral delivery and tracing of chemotherapeutic drugs.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacocinética , Diagnóstico por Imagen/métodos , Ácido Litocólico/química , Hígado/efectos de los fármacos , Fosfolípidos/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Disponibilidad Biológica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Tamoxifeno/química , Tamoxifeno/farmacocinética , Tamoxifeno/farmacología , Tamoxifeno/toxicidad , Distribución Tisular
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda