Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Redox Biol ; 70: 103070, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38359745

RESUMEN

Although widely known as a tumor suppressor, the breast cancer 1 susceptibility protein (BRCA1) is also important in development, where it regulates fetal DNA repair pathways that protect against DNA damage caused by physiological and drug-enhanced levels of reactive oxygen species (ROS). We previously showed that conditional heterozygous (+/-) knockout (cKO) mouse embryos with a minor 28% BRCA1 deficiency developed normally in culture, but when exposed to the ROS-initiating drug, alcohol (ethanol, EtOH), exhibited embryopathies not evident in wild-type (+/+) littermates. Herein, we characterized a directBrca1 +/- knockout (KO) model with a 2-fold greater (58%) reduction in BRCA1 protein vs. the cKO model. We also characterized and compared learning & memory deficits in both the cKO and KO models. Even saline-exposed Brca1 +/- vs. +/+ KO progeny exhibited enhanced oxidative DNA damage and embryopathies in embryo culture and learning & memory deficits in females in vivo, which were not observed in the cKO model, revealing the potential pathogenicity of physiological ROS levels. The embryopathic EtOH concentration for cultured direct KO embryos was half that for cKO embryos, and EtOH affected Brca1 +/+ embryos only in the direct KO model. The spectrum and severity of EtOH embryopathies in culture were greater in both Brca1 +/- vs. +/+ embryos, and direct KO vs. cKO +/- embryos. Motor coordination deficits were evident in both male and female Brca1 +/- KO progeny exposed in utero to EtOH. The results in our direct KO model with a greater BRCA1 deficiency vs. cKO mice provide the first evidence for BRCA1 protein dose-dependent susceptibility to developmental disorders caused by physiological and drug-enhanced oxidative stress.


Asunto(s)
Enfermedades Fetales , Trastornos del Neurodesarrollo , Humanos , Masculino , Femenino , Ratones , Animales , Etanol/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Ratones Noqueados , Estrés Oxidativo , Daño del ADN , Enfermedades Fetales/metabolismo , Enfermedades Fetales/patología , Trastornos del Neurodesarrollo/inducido químicamente , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo
2.
Free Radic Biol Med ; 208: 272-284, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37541454

RESUMEN

The breast cancer 1 (Brca1) susceptibility gene regulates the repair of reactive oxygen species (ROS)-mediated DNA damage, which is implicated in neurodevelopmental disorders. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), including abnormal brain function, associated with enhanced ROS-initiated DNA damage. Herein, oxidative DNA damage in fetal brains and neurodevelopmental disorders were enhanced in saline-exposed +/- vs. +/+ Brca1 littermates. A single EtOH exposure during gestation further enhanced oxidative DNA damage, altered the expression of developmental/DNA damage response genes in fetal brains, and resulted in neurodevelopmental disorders, all of which were BRCA1-dependent. Pretreatment with the ROS inhibitor phenylbutylnitrone (PBN) blocked DNA damage and some neurodevelopmental disorders in both saline- and EtOH-exposed progeny, corroborating a ROS-dependent mechanism. Fetal BRCA1 protects against altered gene expression and neurodevelopmental disorders caused by both physiological and EtOH-enhanced levels of ROS formation. BRCA1 deficiencies may enhance the risk for FASD.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Neoplasias , Trastornos del Neurodesarrollo , Embarazo , Humanos , Femenino , Especies Reactivas de Oxígeno/metabolismo , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/genética , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Neurodesarrollo/inducido químicamente , Trastornos del Neurodesarrollo/genética , Expresión Génica , Proteína BRCA1/genética
3.
Redox Biol ; 48: 102148, 2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34736119

RESUMEN

The breast cancer 1 protein (BRCA1) facilitates DNA repair, preventing embryolethality and protecting the fetus from reactive oxygen species (ROS)-induced developmental disorders mediated by oxidatively damaged DNA. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), characterized by aberrant behaviour and enhanced ROS formation and proteasomal protein degradation. Herein, ROS-producing NADPH oxidase (NOX) activity was higher in Brca1 +/- vs. +/+ fetal and adult brains, and further enhanced by a single EtOH exposure. EtOH also enhanced catalase and proteasomal activities, while conversely reducing BRCA1 protein levels without affecting Brca1 gene expression. EtOH-initiated adaptive postnatal freezing behaviour was lost in Brca1 +/- progeny. Pretreatment with the free radical spin trap and ROS inhibitor phenylbutylnitrone blocked all EtOH effects, suggesting ROS-dependent mechanisms. This is the first in vivo evidence of NOX regulation by BRCA1, and of EtOH-induced, ROS-mediated depletion of BRCA1, revealing novel mechanisms of BRCA1 protection in FASD.

4.
Methods Mol Biol ; 1965: 313-328, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31069684

RESUMEN

Reactive oxygen species (ROS) can oxidize cellular macromolecules like DNA, causing DNA damage. The most common form of DNA damage is the 8-oxoguanine (8-oxoG) lesion, typically repaired by the base excision repair (BER) pathway, which is initiated by the enzyme oxoguanine glycosylase 1 (OGG1). ROS are produced endogenously and can be enhanced by environmental factors, such as xenobiotics, radiation, and microbial pathogens. As a commonly used biomarker of oxidative damage, 8-oxoG can be measured in two different ways described herein. Commercially available ELISA kits allow for easy detection of the 8-oxoG lesion, while more difficult HPLC assays with UV and electrochemical detection allow for a more definitive identification and quantification of 8-oxoG.


Asunto(s)
Daño del ADN , Guanina/análogos & derivados , Cromatografía Líquida de Alta Presión , ADN Glicosilasas/metabolismo , Técnicas Electroquímicas , Ensayo de Inmunoadsorción Enzimática , Guanina/análisis , Humanos , Especies Reactivas de Oxígeno/metabolismo
5.
Methods Mol Biol ; 1965: 351-374, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31069686

RESUMEN

Known for its tumor suppressor activity in breast and ovarian cancers, the breast cancer 1 susceptibility gene (Brca1) is involved in a variety of cellular pathways including DNA repair, antioxidant signaling, apoptosis, and cell cycle regulation. BRCA1 can translocate between the cytoplasm and nucleus to perform its various roles. Herein is a procedure for measuring BRCA1 protein levels in the whole cell lysate (WCL), as well as in the nuclear (N) and cytoplasmic (C) fractions of mouse tissues at different gestational ages. The method employs multiple loading controls to ensure proper separation of fractions and a total protein stain for more consistent comparisons of dissimilar samples. This method is useful for identifying BRCA1 deficiencies and localization in a variety of research fields, including development, neurodegeneration, and cancer.


Asunto(s)
Proteína BRCA1/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Animales , Western Blotting , Fraccionamiento Celular , Femenino , Edad Gestacional , Ratones , Embarazo , Transporte de Proteínas
6.
Birth Defects Res ; 111(12): 714-748, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31033255

RESUMEN

This review covers molecular mechanisms involving oxidative stress and DNA damage that may contribute to morphological and functional developmental disorders in animal models resulting from exposure to alcohol (ethanol, EtOH) in utero or in embryo culture. Components covered include: (a) a brief overview of EtOH metabolism and embryopathic mechanisms other than oxidative stress; (b) mechanisms within the embryo and fetal brain by which EtOH increases the formation of reactive oxygen species (ROS); (c) critical embryonic/fetal antioxidative enzymes and substrates that detoxify ROS; (d) mechanisms by which ROS can alter development, including ROS-mediated signal transduction and oxidative DNA damage, the latter of which leads to pathogenic genetic (mutations) and epigenetic changes; (e) pathways of DNA repair that mitigate the pathogenic effects of DNA damage; (f) related indirect mechanisms by which EtOH enhances risk, for example by enhancing the degradation of some DNA repair proteins; and, (g) embryonic/fetal pathways like NRF2 that regulate the levels of many of the above components. Particular attention is paid to studies in which chemical and/or genetic manipulation of the above mechanisms has been shown to alter the ability of EtOH to adversely affect development. Alterations in the above components are also discussed in terms of: (a) individual embryonic and fetal determinants of risk and (b) potential risk biomarkers and mitigating strategies. FASD risk is likely increased in progeny which/who are biochemically predisposed via genetic and/or environmental mechanisms, including enhanced pathways for ROS formation and/or deficient pathways for ROS detoxification or DNA repair.


Asunto(s)
Daño del ADN , Embrión de Mamíferos , Trastornos del Espectro Alcohólico Fetal , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Animales , Embrión de Mamíferos/embriología , Embrión de Mamíferos/patología , Femenino , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Espectro Alcohólico Fetal/patología , Humanos , Embarazo
7.
J Control Release ; 282: 35-45, 2018 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-29673642

RESUMEN

Triple negative breast cancer is an aggressive disease that accounts for at least 15% of breast cancer diagnoses, and a disproportionately high percentage of breast cancer related morbidity. Intensive research efforts are focused on the development of more efficacious treatments for this disease, for which therapeutic options remain limited. The high incidence of mutations in key DNA repair pathways in triple negative breast cancer results in increased sensitivity to DNA damaging agents, such as platinum-based chemotherapies. Hyperthermia has been successfully used in breast cancer treatment to sensitize tumors to radiation therapy and chemotherapy. It has also been used as a mechanism to trigger drug release from thermosensitive liposomes. In this study, mild hyperthermia is used to trigger release of cisplatin from thermosensitive liposomes in the vasculature of human triple negative breast cancer tumors implanted orthotopically in mice. This heat-triggered liposomal formulation of cisplatin resulted in significantly delayed tumor growth and improved overall survival compared to treatment with either non-thermosensitive liposomes containing cisplatin or free cisplatin, as was observed in two independent tumor models (i.e. MDA-MB-231 and MDA-MB-436). The in vitro sensitivity of the cell lines to cisplatin and hyperthermia alone and in combination was characterized extensively using enzymatic assays, clonogenic assays, and spheroid growth assays. Evaluation of correlations between the in vitro and in vivo results served to identify the in vitro approach that is most predictive of the effects of hyperthermia in vivo. Relative expression of several heat shock proteins and the DNA damage repair protein BRCA1 were assayed at baseline and in response to hyperthermia both in vitro and in vivo. Interestingly, delivery of cisplatin in thermosensitive liposomes in combination with hyperthermia resulted in the most significant tumor growth delay, relative to free cisplatin, in the less cisplatin-sensitive cell line (i.e. MDA-MB-231). This work demonstrates that thermosensitive cisplatin liposomes used in combination with hyperthermia offer a novel method for effective treatment of triple negative breast cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos/métodos , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Mama/irrigación sanguínea , Mama/efectos de los fármacos , Mama/patología , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Humanos , Hipertermia Inducida/métodos , Liposomas/química , Ratones SCID , Neoplasias de la Mama Triple Negativas/patología
8.
Birth Defects Res C Embryo Today ; 108(2): 108-30, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27345013

RESUMEN

In utero exposure of mouse progeny to alcohol (ethanol, EtOH) and methamphetamine (METH) causes substantial postnatal neurodevelopmental deficits. One emerging pathogenic mechanism underlying these deficits involves fetal brain production of reactive oxygen species (ROS) that alter signal transduction, and/or oxidatively damage cellular macromolecules like lipids, proteins, and DNA, the latter leading to altered gene expression, likely via non-mutagenic mechanisms. Even physiological levels of fetal ROS production can be pathogenic in biochemically predisposed progeny, and ROS formation can be enhanced by drugs like EtOH and METH, via activation/induction of ROS-producing NADPH oxidases (NOX), drug bioactivation to free radical intermediates by prostaglandin H synthases (PHS), and other mechanisms. Antioxidative enzymes, like catalase in the fetal brain, while low, provide critical protection. Oxidatively damaged DNA is normally rapidly repaired, and fetal deficiencies in several DNA repair proteins, including oxoguanine glycosylase 1 (OGG1) and breast cancer protein 1 (BRCA1), enhance the risk of drug-initiated postnatal neurodevelopmental deficits, and in some cases deficits in untreated progeny, the latter of which may be relevant to conditions like autism spectrum disorders (ASD). Risk is further regulated by fetal nuclear factor erythroid 2-related factor 2 (Nrf2), a ROS-sensing protein that upregulates an array of proteins, including antioxidative enzymes and DNA repair proteins. Imbalances between conceptal pathways for ROS formation, versus those for ROS detoxification and DNA repair, are important determinants of risk. Birth Defects Research (Part C) 108:108-130, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Encéfalo/metabolismo , Daño del ADN , Embrión de Mamíferos/metabolismo , Etanol/efectos adversos , Femenino , Desarrollo Fetal/fisiología , Enfermedades Fetales , Feto/metabolismo , Humanos , Metanfetamina/efectos adversos , Ratones , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/fisiopatología , Estrés Oxidativo/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...