Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Horm Behav ; 128: 104890, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33221288

RESUMEN

Developmental exposure to endocrine disrupting chemicals (EDCs), e.g., bisphenol A (BPA) or genistein (GEN), causes longstanding epigenome effects. MicroRNAs (miRs) regulate which mRNAs will be translated to proteins and thereby serve as the final checkpoint in epigenetic control. Scant amount is known, however, whether EDCs affect neural miRNA (miR) patterns. We aimed to test the hypothesis that developmental exposure of California mice (Peromyscus californicus) to GEN, BPA, or both chemicals influences hypothalamic miR/small RNA profiles and ascertain the extent such biomolecular alterations correlate with behavioral and metabolic changes. California mice were developmentally exposed to GEN (250 mg/kg feed weight, FW), GEN (250 mg/kg FW)+BPA (5 mg/kg FW), low dose (LD) BPA (5 mg/kg FW), or upper dose (UD) BPA (50 mg/kg FW). Adult offspring were tested in a battery of behavioral and metabolic tests; whereupon, mice were euthanized, brains were collected and frozen, small RNAs were isolated from hypothalamic punches, and subsequently sequenced. California mice exposed to one or both EDCs engaged in one or more repetitive behaviors. GEN, LD BPA, and UD BPA altered aspects of ultrasonic and audible vocalizations. Each EDC exposure led to sex-dependent differences in differentially expressed miR/small RNAs with miR7-2, miR146, and miR148a being increased in all female and male EDC exposed groups. Current findings reveal that developmental exposure to GEN and/or BPA affects hypothalamic miR/small RNA expression patterns, and such changes correlate with EDC-induced behavioral and metabolic alterations. miR146 is likely an important mediator and biomarker of EDC exposure in mammals, including humans.


Asunto(s)
Disruptores Endocrinos , MicroARNs , Animales , Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Femenino , Hipotálamo , Masculino , Ratones , MicroARNs/genética , Peromyscus , Caracteres Sexuales
2.
Proc Natl Acad Sci U S A ; 113(32): E4601-9, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27457945

RESUMEN

Malignant osteolysis associated with inoperable primary bone tumors and multifocal skeletal metastases remains a challenging clinical problem in cancer patients. Nanomedicine that is able to target and deliver therapeutic agents to diseased bone sites could potentially provide an effective treatment option for different types of skeletal cancers. Here, we report the development of polylactide nanoparticles (NPs) loaded with doxorubicin (Doxo) and coated with bone-seeking pamidronate (Pam) for the targeted treatment of malignant skeletal tumors. In vivo biodistribution of radiolabeled targeted Pam-NPs demonstrated enhanced bone tumor accumulation and prolonged retention compared with nontargeted NPs. In a murine model of focal malignant osteolysis, Pam-functionalized, Doxo-loaded NPs (Pam-Doxo-NPs) significantly attenuated localized osteosarcoma (OS) progression compared with nontargeted Doxo-NPs. Importantly, we report on the first evaluation to our knowlege of Pam-Doxo-NPs in dogs with OS, which possess tumors of anatomic size and physiology comparable to those in humans. The repeat dosing of Pam-Doxo-NPs in dogs with naturally occurring OS indicated the therapeutic was well tolerated without hematologic, nonhematologic, and cardiac toxicities. By nuclear scintigraphy, the biodistribution of Pam-Doxo-NPs demonstrated malignant bone-targeting capability and exerted measurable anticancer activities as confirmed with percent tumor necrosis histopathology assessment.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/administración & dosificación , Doxorrubicina/administración & dosificación , Nanoconjugados/administración & dosificación , Osteólisis/tratamiento farmacológico , Animales , Difosfonatos/farmacocinética , Doxorrubicina/toxicidad , Femenino , Ratones , Ratones Endogámicos BALB C , Pamidronato
3.
Horm Behav ; 103: 97-106, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29920269

RESUMEN

This study investigated the efficacy of components of licorice root to alter performance on two different recognition tasks, a hippocampus-sensitive metric change in object location (MCOL) task and a striatum-sensitive double object recognition (DOR) task. Isoliquiritigenin (ISL), licorice root extract (LRE), and whole licorice root powder (LRP) were assessed. Young adult female rats were ovariectomized (OVX) and exposed to ISL, LRE or LRP at 0.075%, 0.5% or 5% respectively in the diet. An estradiol group was included as a positive control based on our prior findings. Rats were allowed to explore two objects for three 5-min study trials (separated by 3-min intervals) before a fourth 5-min test trial where the objects were moved closer together (MCOL task) or replaced with two new objects (DOR task). Rats typically habituate to the objects across the three study trials. An increase in object exploration time in the test trial suggests the rat detected the change. Estradiol improved MCOL performance and impaired DOR performance, similar to previously shown effects of estradiol and other estrogens, which tend to improve learning and memory on hippocampus-sensitive tasks and impair striatum-sensitive cognition. LRP had no effect on recognition while exposure to ISL and LRE improved MCOL performance. Exposure to ISL, LRE and LRP failed to attenuate DOR, contrary to effects of estradiol shown here and to previous reports in young-adult OVX rats. These findings suggest components of licorice root may prove to be effective therapies targeting memory enhancement without unintended deleterious cognitive effects.


Asunto(s)
Estrógenos/farmacología , Glycyrrhiza/química , Extractos Vegetales/farmacología , Raíces de Plantas/química , Reconocimiento en Psicología/efectos de los fármacos , Percepción Visual/efectos de los fármacos , Animales , Estradiol/farmacología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Memoria/efectos de los fármacos , Ovariectomía , Ratas , Ratas Long-Evans , Navegación Espacial/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 112(15): 4737-42, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825714

RESUMEN

Recurrent estrogen receptor α (ERα)-positive breast and ovarian cancers are often therapy resistant. Using screening and functional validation, we identified BHPI, a potent noncompetitive small molecule ERα biomodulator that selectively blocks proliferation of drug-resistant ERα-positive breast and ovarian cancer cells. In a mouse xenograft model of breast cancer, BHPI induced rapid and substantial tumor regression. Whereas BHPI potently inhibits nuclear estrogen-ERα-regulated gene expression, BHPI is effective because it elicits sustained ERα-dependent activation of the endoplasmic reticulum (EnR) stress sensor, the unfolded protein response (UPR), and persistent inhibition of protein synthesis. BHPI distorts a newly described action of estrogen-ERα: mild and transient UPR activation. In contrast, BHPI elicits massive and sustained UPR activation, converting the UPR from protective to toxic. In ERα(+) cancer cells, BHPI rapidly hyperactivates plasma membrane PLCγ, generating inositol 1,4,5-triphosphate (IP3), which opens EnR IP3R calcium channels, rapidly depleting EnR Ca(2+) stores. This leads to activation of all three arms of the UPR. Activation of the PERK arm stimulates phosphorylation of eukaryotic initiation factor 2α (eIF2α), resulting in rapid inhibition of protein synthesis. The cell attempts to restore EnR Ca(2+) levels, but the open EnR IP3R calcium channel leads to an ATP-depleting futile cycle, resulting in activation of the energy sensor AMP-activated protein kinase and phosphorylation of eukaryotic elongation factor 2 (eEF2). eEF2 phosphorylation inhibits protein synthesis at a second site. BHPI's novel mode of action, high potency, and effectiveness in therapy-resistant tumor cells make it an exceptional candidate for further mechanistic and therapeutic exploration.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/antagonistas & inhibidores , Biosíntesis de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Indoles/química , Indoles/farmacología , Células MCF-7 , Ratones Desnudos , Estructura Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bibliotecas de Moléculas Pequeñas/química , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Proc Natl Acad Sci U S A ; 111(43): 15344-9, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25316794

RESUMEN

Nanomedicines (NMs) offer new solutions for cancer diagnosis and therapy. However, extension of progression-free interval and overall survival time achieved by Food and Drug Administration-approved NMs remain modest. To develop next generation NMs to achieve superior anticancer activities, it is crucial to investigate and understand the correlation between the physicochemical properties of NMs (particle size in particular) and their interactions with biological systems to establish criteria for NM optimization. Here, we systematically evaluated the size-dependent biological profiles of three monodisperse drug-silica nanoconjugates (NCs; 20, 50, and 200 nm) through both experiments and mathematical modeling and aimed to identify the optimal size for the most effective anticancer drug delivery. Among the three NCs investigated, the 50-nm NC shows the highest tumor tissue retention integrated over time, which is the collective outcome of deep tumor tissue penetration and efficient cancer cell internalization as well as slow tumor clearance, and thus, the highest efficacy against both primary and metastatic tumors in vivo.


Asunto(s)
Antineoplásicos/química , Nanomedicina , Tamaño de la Partícula , Animales , Antineoplásicos/uso terapéutico , Humanos , Células MCF-7 , Ratones Desnudos , Nanoconjugados , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Dióxido de Silicio/química , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
6.
BMC Complement Altern Med ; 17(1): 516, 2017 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-29197355

RESUMEN

BACKGROUND: Metastasis refers to the spread of a primary tumor cell from the primary site to other locations in the body and it is generally associated with the severity of a tumor. Extract of Ginkgo biloba (EGb) contains various bioactive compounds and it exerts beneficial effects including improvements in brain function and reduced risk of cardiovascular diseases. On the other hand, increased risk of thyroid and liver cancers by EGb have been reported in animals. METHODS: A colon cancer metastasis model was established using intrasplenic injection of a human colon cancer cell line, SW620-luc in athymic mice to investigate the potential impact of EGb on colon cancer progression. After tumor establishment, EGb was intraperitonically injected daily for 5 wks. RESULTS: EGb significantly increased the rate of metastasis in mouse liver and decreased the number of necrotic and apoptotic cells in the metastatic liver when compared to the control. Meanwhile, EGb significantly induced proliferation of tumor cells in the metastatic liver, indicated by increased staining of Ki67 and H3S10p. mRNA expression of genes involved in cell cycle, metastasis, apoptosis, and oxidative stress were altered by EGb treatment in livers with tumors. Moreover, EGb activated the stress-responsive MAPK pathways in the liver with metastatic tumors. CONCLUSIONS: EGb exacerbated liver metastasis in a mouse colon cancer metastasis model. This is potentially due to the increased tumor cell proliferation involving stimulated MAPK pathways.


Asunto(s)
Neoplasias del Colon , Ginkgo biloba , Neoplasias Hepáticas , Hígado/efectos de los fármacos , Extractos Vegetales , Animales , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Extractos Vegetales/efectos adversos , Extractos Vegetales/química , Extractos Vegetales/farmacología
7.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25865775

RESUMEN

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Microambiente Tumoral/genética , Antineoplásicos/uso terapéutico , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/prevención & control , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos
8.
Semin Cancer Biol ; 35 Suppl: S151-S184, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25951989

RESUMEN

Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.


Asunto(s)
Antineoplásicos/uso terapéutico , Inflamación/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Transformación Celular Neoplásica/efectos de los fármacos , Heterogeneidad Genética/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de los fármacos
9.
Semin Cancer Biol ; 35 Suppl: S185-S198, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25818339

RESUMEN

Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.


Asunto(s)
Carcinogénesis/inmunología , Evasión Inmune , Neoplasias/inmunología , Neoplasias/terapia , Presentación de Antígeno/inmunología , Carcinogénesis/efectos de los fármacos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Neoplasias/patología , Fitoquímicos/uso terapéutico , Linfocitos T Reguladores/inmunología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología
10.
Semin Cancer Biol ; 35 Suppl: S5-S24, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25869442

RESUMEN

Genomic instability can initiate cancer, augment progression, and influence the overall prognosis of the affected patient. Genomic instability arises from many different pathways, such as telomere damage, centrosome amplification, epigenetic modifications, and DNA damage from endogenous and exogenous sources, and can be perpetuating, or limiting, through the induction of mutations or aneuploidy, both enabling and catastrophic. Many cancer treatments induce DNA damage to impair cell division on a global scale but it is accepted that personalized treatments, those that are tailored to the particular patient and type of cancer, must also be developed. In this review, we detail the mechanisms from which genomic instability arises and can lead to cancer, as well as treatments and measures that prevent genomic instability or take advantage of the cellular defects caused by genomic instability. In particular, we identify and discuss five priority targets against genomic instability: (1) prevention of DNA damage; (2) enhancement of DNA repair; (3) targeting deficient DNA repair; (4) impairing centrosome clustering; and, (5) inhibition of telomerase activity. Moreover, we highlight vitamin D and B, selenium, carotenoids, PARP inhibitors, resveratrol, and isothiocyanates as priority approaches against genomic instability. The prioritized target sites and approaches were cross validated to identify potential synergistic effects on a number of important areas of cancer biology.


Asunto(s)
Inestabilidad Genómica/efectos de los fármacos , Neoplasias/dietoterapia , Neoplasias/genética , Centrosoma/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Dieta , Inestabilidad Genómica/genética , Humanos , Neoplasias/patología , Pronóstico , Telomerasa/antagonistas & inhibidores , Telomerasa/genética
11.
Semin Cancer Biol ; 35 Suppl: S78-S103, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25936818

RESUMEN

Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.


Asunto(s)
Apoptosis/genética , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
12.
Toxicol Appl Pharmacol ; 293: 53-62, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26792615

RESUMEN

Genistein is a naturally occurring isoflavone phytoestrogen commonly found in plant products such as soybeans, lentils, and chickpeas. Genistein, like other phytoestrogens, has the potential to mimic, enhance, or impair the estradiol biosynthesis pathway, thereby potentially altering ovarian follicle growth. Previous studies have inconsistently indicated that genistein exposure may alter granulosa cell proliferation and hormone production, but no studies have examined the effects of genistein on intact antral follicles. Thus, this study was designed to test the hypothesis that genistein exposure inhibits follicle growth and steroidogenesis in intact antral follicles. To test this hypothesis, antral follicles isolated from CD-1 mice were cultured with vehicle (dimethyl sulfoxide; DMSO) or genistein (6.0 and 36µM) for 18-96h. Every 24h, follicle diameters were measured to assess growth. At the end of each culture period, the media were pooled to measure hormone levels, and the cultured follicles were collected to measure expression of cell cycle regulators and steroidogenic enzymes. The results indicate that genistein (36µM) inhibits growth of mouse antral follicles. Additionally, genistein (6.0 and 36µM) increases progesterone, testosterone, and dehydroepiandrosterone (DHEA) levels, but decreases estrone and estradiol levels. The results also indicate that genistein alters the expression of steroidogenic enzymes at 24, 72 and 96h, and the expression of cell cycle regulators at 18h. These data indicate that genistein exposure inhibits antral follicle growth by inhibiting the cell cycle, alters sex steroid hormone levels, and dysregulates steroidogenic enzymes in cultured mouse antral follicles.


Asunto(s)
Genisteína/toxicidad , Folículo Ovárico/efectos de los fármacos , Fitoestrógenos/toxicidad , 17-Hidroxiesteroide Deshidrogenasas/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Ciclo Celular/genética , Sistema Enzimático del Citocromo P-450/genética , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Hormonas Esteroides Gonadales/metabolismo , Ratones , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Fosfoproteínas/genética
13.
Arch Biochem Biophys ; 591: 98-110, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26682631

RESUMEN

Previously, we observed that wild yam (Dioscorea villosa) root extract (WYRE) was able to activate GATA3 in human breast cancer cells targeting epigenome. This study aimed to find out if dioscin (DS), a bioactive compound of WYRE, can modulate GATA3 functions and cellular invasion in human breast cancer cells. MCF-7 and MDA-MB-231 cells were treated in the absence/presence of various concentrations of DS and subjected to gene analysis by RT-qPCR, immunoblotting, and immunocytochemistry. We determined the ability of MDA-MB-231 cells to migrate into wound area and examined the effects of DS on cellular invasion using invasion assay. DS reduced cell viability of both cell lines in a concentration and time-dependent manner. GATA3 expression was enhanced by DS (5.76 µM) in MDA-MB-231 cells. DS (5.76 µM)-treated MDA-MB-231 cells exhibited the morphological characteristic of epithelial-like cells; mRNA expression of DNMT3A, TET2, TET3, ZFPM2 and E-cad were increased while TET1, VIM and MMP9 were decreased. Cellular invasion of MDA-MB-231 was reduced by 65 ± 5% in the presence of 5.76 µM DS. Our data suggested that DS-mediated pathway could promote GATA3 expression at transcription and translation levels. We propose that DS has potential to be used as an anti-invasive agent in breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Dioscorea/química , Diosgenina/análogos & derivados , Extractos Vegetales/administración & dosificación , Raíces de Plantas/química , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diosgenina/administración & dosificación , Relación Dosis-Respuesta a Droga , Humanos , Células MCF-7 , Invasividad Neoplásica , Fitosteroles/administración & dosificación , Saponinas/administración & dosificación , Resultado del Tratamiento
14.
Toxicol Appl Pharmacol ; 284(2): 101-12, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25748669

RESUMEN

Developmental bisphenol A (BPA) exposure increases adulthood hepatic steatosis with reduced mitochondrial function. To investigate the potential epigenetic mechanisms behind developmental BPA-induced hepatic steatosis, pregnant Sprague-Dawley rats were dosed with vehicle (oil) or BPA (100µg/kg/day) from gestational day 6 until postnatal day (PND) 21. After weaning, offspring were either challenged with a high-fat (HF; 45% fat) or remained on a control (C) diet until PND110. From PND60 to 90, both BPA and HF diet increased the fat/lean ratio in males only, and the combination of BPA and HF diet appeared to cause the highest ratio. On PND110, Oil-HF, BPA-C, and BPA-HF males had higher hepatic lipid accumulation than Oil-C, with microvesicular steatosis being marked in the BPA-HF group. Furthermore, on PND1, BPA increased and modified hepatic triglyceride (TG) and free fatty acid (FFA) compositions in males only. In PND1 males, BPA increased hepatic expression of FFA uptake gene Fat/Cd36, and decreased the expression of TG synthesis- and ß-oxidation-related genes (Dgat, Agpat6, Cebpα, Cebpß, Pck1, Acox1, Cpt1a, Cybb). BPA altered DNA methylation and histone marks (H3Ac, H4Ac, H3Me2K4, H3Me3K36), and decreased the binding of several transcription factors (Pol II, C/EBPß, SREBP1) within the male Cpt1a gene, the key ß-oxidation enzyme. In PND1 females, BPA only increased the expression of genes involved in FFA uptake and TG synthesis (Lpl, Fasn, and Dgat). These data suggest that developmental BPA exposure alters and reprograms hepatic ß-oxidation capacity in males, potentially through the epigenetic regulation of genes, and further alters the response to a HF diet.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Dieta Alta en Grasa/efectos adversos , Hígado Graso/genética , Hígado/efectos de los fármacos , Hígado/fisiología , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/genética , Animales , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Ácidos Grasos no Esterificados/genética , Ácidos Grasos no Esterificados/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Masculino , Oxidación-Reducción/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triglicéridos/genética , Triglicéridos/metabolismo
15.
J Sci Food Agric ; 95(2): 393-400, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24817038

RESUMEN

BACKGROUND: Micronutrient deficiencies continue to afflict children rural populations around the world. A micronutrient delivery vehicle (MDV) was developed as a point-of-use technology for fortification of meals for school-age children beneficiaries of the Healthy Schools Program (HSP) in Honduras. RESULTS: MDV combines micronutrient powder through a traditional dough-making process, using staple flours (wheat and nixtamalized corn), oil and water as ingredients. After mixing the ingredients and kneading, dough is extruded through a specially designed hand press into noodles. After drying (overnight, 23°C), noodles are broken into small pieces, mixed (1:100 w/w) with rice and cooked as customary. Dispersion studies with NaFeEDTA showed adequate distribution (<10% RSD) and recovery (>90%) in white rice. Color changes in MDV due to addition of vitamin A and iron (NaFeEDTA) carried forward into cooked rice. In Honduras, children from two rural schools (N = 47, 6-12 years) were not able to differentiate (triangle test) between control and unfortified MDV mixed (1:100 w/w) with white rice. Children from four schools (N = 83, 7-12 years) accepted control and iron fortified rice (3 mg Fe per serving) based on color and flavor similarly. CONCLUSION: This is a feasible point-of-use fortification technology for improvement of meals provided by the HSP in Honduras.


Asunto(s)
Enfermedades Carenciales/prevención & control , Dieta , Alimentos Fortificados , Hierro/administración & dosificación , Micronutrientes/administración & dosificación , Oryza , Vitamina A/administración & dosificación , Niño , Color , Comportamiento del Consumidor , Femenino , Preferencias Alimentarias , Honduras , Humanos , Hierro/uso terapéutico , Deficiencias de Hierro , Masculino , Comidas , Micronutrientes/deficiencia , Micronutrientes/uso terapéutico , Polvos , Triticum , Vitamina A/uso terapéutico , Zea mays
16.
FASEB J ; 27(11): 4406-18, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23882126

RESUMEN

Because little is known about the actions of botanical estrogens (BEs), widely consumed by menopausal women, we investigated the mechanistic and cellular activities of some major BEs. We examined the interactions of genistein, daidzein, equol, and liquiritigenin with estrogen receptors ERα and ERß, with key coregulators (SRC3 and RIP140) and chromatin binding sites, and the regulation of gene expression and proliferation in MCF-7 breast cancer cells containing ERα and/or ERß. Unlike the endogenous estrogen, estradiol (E2), BEs preferentially bind to ERß, but their ERß-potency selectivity in gene stimulation (340- to 830-fold vs. E2) is enhanced at several levels (coregulator recruitment, chromatin binding); nevertheless, at high (0.1 or 1 µM) concentrations, BEs also fully activate ERα. Because ERα drives breast cancer cell proliferation and ERß dampens this, the relative levels of these two ERs in target cells and the BE dose greatly affect gene expression and proliferative response and will be crucial determinants of the potential benefits vs. risks of BEs. Our findings reveal key and novel mechanistic differences in the estrogenic activities of BEs vs. E2, with BEs displaying patterns of activity distinctly different from those seen with E2 and provide valuable information to inform future studies.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Fitoestrógenos/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sitios de Unión , Proliferación Celular , Cromatina/metabolismo , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Humanos , Células MCF-7 , Proteínas Nucleares/metabolismo , Coactivador 3 de Receptor Nuclear/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Transcripción Genética
17.
Carcinogenesis ; 33(4): 895-901, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22266527

RESUMEN

The present study examined the effect of dietary genistein, a soy isoflavone, on breast cancer patients who take tamoxifen, an antiestrogen treatment, using a preclinical model. The interaction of various doses of genistein with tamoxifen on the growth of estrogen receptor-positive breast cancer MCF-7 cells was investigated by subcutaneously injecting MCF-7 cells into the flank of ovariectomized athymic mice. Animals were randomized into eight experimental groups with 10-13 mice per group: control (C), estrogen (E) (0.08 mg E implant), tamoxifen (T) (3 mg T implant), estrogen + tamoxifen (E + T), tamoxifen + 500 p.p.m. genistein (T + G500), estrogen + tamoxifen + 250 p.p.m. genistein (E + T + G250), estrogen + tamoxifen + 500 p.p.m. genistein (E + T + G500) and estrogen + tamoxifen + 1000 p.p.m. genistein (E + T + G1000). Treatment of tamoxifen significantly reduced the estrogen-induced MCF-7 tumor prevalence and tumor size. This inhibitory effect of tamoxifen was significantly negated by the low doses of dietary genistein (250 and 500 p.p.m.), whereas the 1000 p.p.m. genistein did not have the same effect. Cells harvested from tamoxifen-treated tumors retained estrogen responsiveness of their progenitor MCF-7 cells, indicating that the abrogating effect of genistein on tamoxifen-treated tumor growth was not caused by a diminished tamoxifen response but directly by genistein. The low doses of dietary genistein abrogated the inhibitory effect of tamoxifen potentially by acting on the tumor cell proliferation/apoptosis ratio and the messenger RNA (mRNA) expression of cyclin D1 in addition to regulating the mRNA expression of progesterone receptor. Therefore, data from the current study suggest that caution is warranted regarding the consumption of dietary genistein by breast cancer patients while on tamoxifen therapy.


Asunto(s)
Antineoplásicos Hormonales/antagonistas & inhibidores , Dieta , Genisteína/farmacología , Tamoxifeno/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cartilla de ADN , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estrógenos/sangre , Genisteína/administración & dosificación , Ratones , Ratones Desnudos
18.
Horm Behav ; 62(4): 491-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22944517

RESUMEN

Endogenous estrogens have bidirectional effects on learning and memory, enhancing or impairing cognition depending on many variables, including the task and the memory systems that are engaged. Moderate increases in estradiol enhance hippocampus-sensitive place learning, yet impair response learning that taps dorsal striatal function. This memory modulation likely occurs via activation of estrogen receptors, resulting in altered neural function. Supplements containing estrogenic compounds from plants are widely consumed despite limited information about their effects on brain function, including learning and memory. Phytoestrogens can enter the brain and signal through estrogen receptors to affect cognition. Enhancements in spatial memory and impairments in executive function have been found following treatment with soy phytoestrogens, but no tests of actions on striatum-sensitive tasks have been made to date. The present study compared the effects of acute exposure to the isoflavone genistein with the effects of estradiol on performance in place and response learning tasks. Long-Evans rats were ovariectomized, treated with 17ß-estradiol benzoate, genistein-containing sucrose pellets, or vehicle (oil or plain sucrose pellets) for 2 days prior to behavioral training. Compared to vehicle controls, estradiol treatment enhanced place learning at a low (4.5 µg/kg) but not high dose (45 µg/kg), indicating an inverted pattern of spatial memory facilitation. Treatment with 4.4 mg of genistein over 2 days also significantly enhanced place learning over vehicle controls. For the response task, treatment with estradiol impaired learning at both low and high doses; likewise, genistein treatment impaired response learning compared to rats receiving vehicle. Overall, genistein was found to mimic estradiol-induced shifts in place and response learning, facilitating hippocampus-sensitive learning and slowing striatum-sensitive learning. These results suggest signaling through estrogen receptor ß and membrane-associated estrogen receptors in learning enhancements and impairments given the preferential binding of genistein to the ERß subtype and affinity for GPER.


Asunto(s)
Estradiol/farmacología , Genisteína/farmacología , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Factores de Edad , Animales , Biomimética , Esquema de Medicación , Femenino , Genisteína/administración & dosificación , Aprendizaje/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Condicionamiento Físico Animal , Postura/fisiología , Ratas , Ratas Long-Evans , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
19.
Mol Nutr Food Res ; 66(11): e2100974, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35319818

RESUMEN

SCOPE: A dose-ranging study is performed using young estrogen-depleted rats to determine whether dietary isoliquiritigenin (ILQ) alters bone metabolism and if the effects are associated with estrogen receptor signaling. METHODS AND RESULTS: Six-week-old rats (ovariectomized at 4 weeks of age) are fed diets containing 0, 100, 250, or 750 ppm ILQ (n = 5/treatment) for 7 days. Gene expression in femur and uterus, blood markers of bone turnover, body composition, and uterine weight and epithelial cell height are determined. Because ILQ lowers bone resorption, the effect of ILQ on in vitro differentiation of osteoclasts from bone marrow of mice is assessed. Treatment resulted in a dose-dependent increases in serum ILQ but no changes in serum osteocalcin, a marker of global bone formation. Contrastingly, ILQ administration results in reduced serum CTX-1, a marker of global bone resorption, and reduces tartrate resistant acid phosphatase expression in osteoclast culture. ILQ treatment and endogenous estrogen production had limited overlap on gene expression in femur and uterus. However, uterine epithelial cell hyperplasia is observed in two of five animals treated with 750 ppm. CONCLUSIONS: In conclusion, dietary ILQ reduces bone resorption in vivo and osteoclast differentiation in vitro, by mechanisms likely differing from actions of ovarian hormones.


Asunto(s)
Resorción Ósea , Osteoclastos , Animales , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Diferenciación Celular , Chalconas , Estrógenos/metabolismo , Femenino , Humanos , Ratones , Ovariectomía , Ratas , Fosfatasa Ácida Tartratorresistente/metabolismo , Fosfatasa Ácida Tartratorresistente/farmacología
20.
Biol Reprod ; 83(1): 114-21, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20357267

RESUMEN

Soy-based infant formulas are widely used in the United States and some other countries. These formulas contain high levels of the estrogenic isoflavone genistein, leading to concern that neonatal genistein exposure could cause acute and/or long-term adverse effects on reproductive and other organs. However, previous work to assess genistein effects in rodent models has not typically replicated the route of delivery and/or serum genistein concentrations reported for soy formula-fed human infants. Our objective was to develop a mouse model that more closely mimics the oral genistein exposure and total serum genistein concentrations observed in soy formula-fed infants. Mouse pups were dosed orally with genistein in a soy formula-corn oil emulsion from Postnatal Day (PND) 1 to PND 5, then effects on reproductive and non-reproductive organs were assessed after dosing and during subsequent development. Neonatal treatment resulted in changes both at the completion of dosing (PND 5) and in adult animals. At PND 5, neonatal genistein treatment caused increased relative uterine weight and down-regulation of progesterone receptor in uterine epithelia. Estrogenic effects of genistein were also seen in the neonatal ovary and thymus, which had an increase in the incidence of multioocyte follicles (MOFs) and a decrease in thymic weight relative to body weight, respectively. The increased incidence of MOFs persisted into adulthood for neonatally treated genistein females, and estrous cycle abnormalities were seen at 6 mo of age despite normal fertility in these mice. The immediate and long-term effects in this neonatal animal model raise concerns that high serum concentrations of genistein are estrogenic and could potentially impact the development of human infants fed soy formula.


Asunto(s)
Genisteína/administración & dosificación , Genitales Femeninos/efectos de los fármacos , Fitoestrógenos/administración & dosificación , Administración Oral , Animales , Animales Recién Nacidos , Aceite de Maíz , Femenino , Genisteína/sangre , Genisteína/farmacocinética , Humanos , Lactante , Fórmulas Infantiles , Ratones , Ratones Endogámicos C57BL , Fitoestrógenos/sangre , Fitoestrógenos/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA