Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Biol Int ; 46(11): 1759-1774, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35930599

RESUMO

Glioblastoma (GBM) is a deadly and common primary brain tumor. Poor prognosis is linked to high proliferation and cell heterogeneity. Sex differences may play a role in patient outcome. Previous studies showed that ER-α36, a variant of the estrogen receptor (ER), mediated non-genomic estrogen signaling and is highly expressed in many ER-negative malignant tumors. ER-α36 also associates with epidermal growth factor receptor (EGFR). The primary purpose of this study is to investigate the cross talk between ER-α36 and EGFR in estrogen-mediated GBM cell proliferation. Here, we showed that ER-α36 was highly expressed and confirmed that ER-α36 co-labels with EGFR in human GBM samples using immunohistochemical techniques. We also investigated the mechanisms of estrogen-induced proliferation in ER-α-negative cell lines. We found that GBM cells showed varying responsive to mitogenic estrogen signaling which correlated with ER-α36 expression, and knockdown of ER-α36 diminished the response. Exposure to estrogen also caused upregulation of cyclin protein expression in vitro. We also found that low concentration of estrogen promoted SRC-Y-416 and inhibited SRC-Y-527 phosphorylation, corresponding with activated SRC signaling. Inhibiting SRC or EGFR abolished estrogen-induced mitogenic signaling, including cyclin expression and MAPK phosphorylation. Cumulatively, our results demonstrate that ER-α36 promotes non-genomic estrogen signaling via the EGFR/SRC/MAPK pathway in GBM. This may be important for the treatment of ER-α-negative GBMs that retain high level of ER-α36, since estrogen may be a viable therapeutic target for these patients.


Assuntos
Neoplasias da Mama , Glioblastoma , Linhagem Celular Tumoral , Ciclinas , Receptores ErbB/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Masculino , Receptores de Estrogênio
2.
Exp Dermatol ; 29(9): 876-884, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32654294

RESUMO

The genomic actions of the vitamin D are mediated via its biologically most potent metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ) and the transcription factor vitamin D receptor (VDR). Activation of VDR by 1,25(OH)2 D3 leads to change in the expression of more 1000 genes in various human tissues. Based on (epi)genome, transcriptome and crystal structure data the molecular details of this nuclear vitamin D signalling pathway are well understood. Vitamin D is known for its role on calcium homeostasis and bone formation, but it also modulates energy metabolism, innate and adaptive immunity as well as cellular growth, differentiation and apoptosis. The observation of rapid, non-genomic effects of 1,25(OH)2 D3 at cellular membranes and in the cytosol initiated the question, whether there are alternative vitamin D-binding proteins in these cellular compartments. So far, the best candidate is the enzyme PDIA3 (protein disulphide isomerase family A member 3), which is found at various subcellular locations. Furthermore, also VDR seems to play a role in membrane-based responses to vitamin D. In this viewpoint, we will dispute whether these rapid, non-genomic pathways are a meaningful addition to the genome-wide effects of vitamin D.


Assuntos
Receptores de Calcitriol/metabolismo , Vitamina D/fisiologia , Animais , Humanos , Transdução de Sinais
3.
Front Neuroendocrinol ; 47: 86-108, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28739508

RESUMO

For decades, there has been speculation regarding the interaction of cannabinoids with glucocorticoid systems. Given the functional redundancy between many of the physiological effects of glucocorticoids and cannabinoids, it was originally speculated that the biological mechanisms of cannabinoids were mediated by direct interactions with glucocorticoid systems. With the discovery of the endocannabinoid system, additional research demonstrated that it was actually the opposite; glucocorticoids recruit endocannabinoid signaling, and that the engagement of endocannabinoid signaling mediated many of the neurobiological and physiological effects of glucocorticoids. With the development of advances in pharmacology and genetics, significant advances in this area have been made, and it is now clear that functional interactions between these systems are critical for a wide array of physiological processes. The current review acts a comprehensive summary of the contemporary state of knowledge regarding the biological interactions between glucocorticoids and endocannabinoids, and their potential role in health and disease.


Assuntos
Endocanabinoides/metabolismo , Glucocorticoides/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo
4.
Horm Behav ; 104: 165-172, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29964007

RESUMO

Contribution to Special Issue on Fast effects of steroids. Estrogen signals both slowly to regulate transcription and rapidly to activate kinases and regulate calcium levels. Both rapid, non-genomic signaling as well as genomic transcriptional signaling via intracellular estrogen receptors (ER)s can change behavior. Rapid non-genomic signaling is initiated from the plasma membrane by a G-protein coupled receptor called GPER1 that binds 17ß-estradiol. GPER1 or GPR30 is one of the candidates for a membrane ER (mER) that is not only highly expressed in pathology i.e. cancers but also in several behaviorally-relevant brain regions. In the brain, GPER1 signaling, in response to estrogen, facilitates neuroprotection, social behaviors and cognition. In this review, we describe several notable characteristics of GPER1 such as the ability of several endogenous steroids as well as artificially synthesized molecules to bind the GPER1. In addition, GPER1 is localized to the plasma membrane in breast cancer cell lines but may be present in the endoplasmic reticulum or the Golgi apparatus in the hippocampus. Unusually, GPER1 can also translocate to the perinuclear space from the plasma membrane. We explore the idea that subcellular localization and ligand promiscuity may determine the varied downstream signaling cascades of the activated GPER1. Lastly, we suggest that GPER1 can act as a modulator of ERα-mediated action on a convergent target, spinogenesis, in neurons that in turn drives female social behaviors such as lordosis and social learning.


Assuntos
Encéfalo/metabolismo , Receptor alfa de Estrogênio/fisiologia , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Estrogênios/farmacologia , Feminino , Humanos , Proteínas de Membrana/fisiologia , Neurônios/metabolismo , Neuroproteção/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Front Biosci (Landmark Ed) ; 29(6): 238, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38940045

RESUMO

BACKGROUND: Hormone receptors exert their function through binding with their ligands, which results in cellular signaling activation mediated by genomic or non-genomic mechanisms. The intrinsic molecular communication of tick Rhipicephalus microplus and its host Bos taurus comprises an endocrine regulation involving hormones. In the present study, we performed a molecular and in silico analysis of a Membrane Associated Progesterone Receptor in R. microplus (RmMAPRC). METHODS: The RmMAPRC protein sequence was analyzed with bioinformatics tools, and its structure was characterized by three-dimensional (3D) modeling and molecular docking. A semi-quantitative reverse transcription and polymerase chain reaction (sqRT-PCR) assessed the RmMAPRC gene presence and relative expression in tick organs and embryonic cells. RESULTS: RmMAPRC relative expression in salivary glands, ovaries, and embryonic cells showed overexpression of 3%, 13%, and 24%, respectively. Bioinformatic analysis revealed that RmMAPRC corresponded to a Progesterone Receptor Membrane Component 1 (RmPGRMC1) of ~23.7 kDa, with an N-terminal transmembrane domain and a C-terminal Cytochrome b5-like heme/steroid binding domain. The docking results suggest that RmPGRMC1 could bind to progesterone (P4), some progestins, and P4 antagonists. The phylogenetic reconstruction showed that Rhipicephalus spp. MAPRC receptors were clustered in a clade that includes R. appendiculatus, R. sanguineus, and R. microplus (RmMAPRC), and mammals and helminths MAPRC receptors clustered in two separated clades away from ticks. CONCLUSIONS: The presence of RmPGRMC1 highlights the importance of transregulation as a conserved adaptive mechanism that has succeeded for arthropod parasites, making it a target for tick control.


Assuntos
Progesterona , Receptores de Progesterona , Rhipicephalus , Animais , Rhipicephalus/metabolismo , Rhipicephalus/genética , Receptores de Progesterona/metabolismo , Receptores de Progesterona/genética , Progesterona/metabolismo , Bovinos , Simulação de Acoplamento Molecular , Interações Hospedeiro-Parasita , Feminino , Sequência de Aminoácidos , Ligação Proteica , Filogenia
6.
J Steroid Biochem Mol Biol ; 241: 106523, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38636681

RESUMO

G protein-coupled estrogen receptor (GPER) plays a prominent role in facilitating the rapid, non-genomic signaling of estrogens in breast cancer cells. Herein, a comprehensive overview of the role of GPER in ER-ɑ-negative breast cancer is provided. Activation of GPER affected proliferation, metastasis and epithelial mesenchymal transition in ER-ɑ negative breast cancer cells. Clinical studies have demonstrated that GPER positivity was strongly correlated with larger tumor size and advanced clinical stage, suggesting that GPER/ERK signaling may play a role in promoting tumor progression. Strong evidence existed that environmental contaminants like bisphenol A have a carcinogenic potential mediated by GPER activation. The complexity of the cross talk between GPER and other receptors including ER-ß, ER-α36, Estrogen-related receptor α (ERRα) and androgen receptor has been discussed. The potential utility of small molecules and phytoestrogens targeting GPER, adds valuable insights into its therapeutic potential. This review holds promises in advancing our understanding of GPER role in ER-ɑ-negative breast cancer. Overall, the consequences of GPER activation are still an area of active research and the implication are not entirely clear.


Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Receptores de Estrogênio , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptores de Estrogênio/metabolismo , Feminino , Receptor alfa de Estrogênio/metabolismo , Transdução de Sinais , Animais , Transição Epitelial-Mesenquimal , Fenóis/metabolismo , Fenóis/farmacologia , Proliferação de Células , Compostos Benzidrílicos
7.
Cells ; 12(6)2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36980205

RESUMO

One of the most fundamental discoveries in human biology was that of the existence of essential micronutrients that the body cannot synthesize but nonetheless requires for proper functioning [...].


Assuntos
Receptores do Ácido Retinoico , Tretinoína , Humanos , Tretinoína/farmacologia , Receptores X de Retinoides
8.
Mol Cell Endocrinol ; 539: 111467, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34626731

RESUMO

Estrogen receptor alpha (ERα) and beta (ERß) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Animais , Modelos Animais de Doenças , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Humanos , Mutação , Domínios Proteicos , Transdução de Sinais
9.
Endocrinology ; 162(8)2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34038515

RESUMO

Steroid hormones bind receptors in the cell nucleus and in the cell membrane. The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the cell nucleus. Nuclear receptors, such as estrogen receptor alpha, can also be anchored to the plasma membrane, where they respond to steroids by activating signaling pathways independent of their function as transcription factors. Steroids can also bind integral membrane proteins, such as the G protein-coupled estrogen receptor. Membrane estrogen and progestin receptors have been cloned and characterized in vitro and influence the development and function of many organ systems. Membrane androgen receptors were cloned and characterized in vitro, but their function as androgen receptors in vivo is unresolved. We review the identity and function of membrane proteins that bind estrogens, progestins, and androgens. We discuss evidence that membrane glucocorticoid and mineralocorticoid receptors exist, and whether glucocorticoid and mineralocorticoid nuclear receptors act at the cell membrane. In many cases, integral membrane steroid receptors act independently of nuclear steroid receptors, even though they may share a ligand.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Receptores de Esteroides/metabolismo , Animais , Membrana Celular/metabolismo , Humanos
10.
Mol Cell Endocrinol ; 538: 111453, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34520815

RESUMO

Steroid receptors (SRs) are members of the nuclear receptor family, which are ligand-activated transcription factors. SRs regulate many physiological functions including development and reproduction, though they can also be involved in several pathologies, especially cancer. Highly controlled cellular responses to steroids involve transcriptional regulation (genomic activity) combined with direct activation of signaling cascades (non-genomic activity). Non-genomic signaling has been extensively studied in cancer, mainly in breast cancer for ER and PR, and prostate cancer for AR. Even though most of the studies have been conducted in cells, some of them have been confirmed in vivo, highlighting the relevance of this pathway in cancer. This review provides an overview of the current and emerging knowledge on non-genomic signaling with a focus on breast and prostate cancers and its clinical relevance. A thorough understanding of ER, PR, AR and GR non-genomic pathways may open new perspectives for the development of therapeutic strategies.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Próstata/metabolismo , Receptores de Esteroides/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Transdução de Sinais
11.
Mol Cell Endocrinol ; 502: 110680, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31838026

RESUMO

Agrochemicals represent a significant class of endocrine disrupting chemicals that humans and animals around the world are exposed to constantly. Agrochemicals can act as endocrine disrupting chemicals through a variety of mechanisms. Recent studies have shown that several mechanisms of action involve the ability of agrochemicals to mimic the interaction of endogenous hormones with nuclear receptors such as estrogen receptors, androgen receptors, peroxisome proliferator activated receptors, the aryl hydrocarbon receptor, and thyroid hormone receptors. Further, studies indicate that agrochemicals can exert toxicity through non-nuclear receptor-mediated mechanisms of action. Such non-genomic mechanisms of action include interference with peptide, steroid, or amino acid hormone response, synthesis and degradation as well as epigenetic changes (DNA methylation and histone modifications). This review summarizes the major mechanisms of action by which agrochemicals target the endocrine system.


Assuntos
Agroquímicos/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Epigênese Genética/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
12.
Artigo em Inglês | MEDLINE | ID: mdl-31055067

RESUMO

Molluscs have been widely utilized to evaluate the effects of estrogenic compounds, one of the most widespread classes of Endocrine Disrupting Chemicals-EDCs. However, knowledge on steroid signaling and metabolism in molluscs has considerably increased in the last decade: from these studies, a considerable debate emerged on the role of 'natural' steroids in physiology, in particular in reproduction, of this invertebrate group. In this work, available information on the effects and mechanisms of action of estrogens in molluscs will be reviewed, with particular emphasis on bivalves that, widespread in aquatic ecosystems, are most likely affected by exposure to estrogenic EDCs. Recent advances in steroid uptake and metabolism, and estrogen receptors-ERs in molluscs, as well as in estrogen signaling in vertebrates, will be considered. The results so far obtained with 17ß-estradiol and different estrogenic compounds in the model bivalve Mytilus spp., demonstrate specific effects on immune function, development and metabolism. Transcriptomic data reveal non genomic estrogen signaling pathways in mussel tissues that are supported by new observations at the cellular level. In vitro and in vivo data show, through independent lines of evidence, that estrogens act through non-genomic signaling pathways in bivalves. In this light, regardless of whether molluscs synthesize estrogens de novo or not, and despite their ERs are not directly activated by ligand binding, estrogens can interact with multiple signaling components, leading to modulation of different physiological functions. Increasing knowledge in endocrine physiology of molluscs will provide a framework for a better evaluation and interpretation of data on the impact of estrogenic EDCs in this invertebrate group.


Assuntos
Disruptores Endócrinos/toxicidade , Estrogênios/toxicidade , Moluscos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais
13.
Artigo em Inglês | MEDLINE | ID: mdl-30416486

RESUMO

Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.

14.
Steroids ; 133: 34-37, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29122548

RESUMO

Non-genomic effects of estrogen receptor α (ERα) signaling have been described for decades. However, the mechanisms and physiological processes resulting solely from non-genomic signaling are poorly understood. Challenges in studying these effects arise from the strongly nucleophilic tendencies of estrogen receptor, and many approaches to excluding ERα from the nucleus have been explored over the years. In this review, we discuss past strategies for studying ERα's non-genomic action and current models, specifically H2NES ERα, first described by Burns et al. (2011). In vitro and preliminary in vivo data from H2NES ERα and H2NES mice suggest a promising avenue for pinpointing specific non-genomic ERα action.


Assuntos
Estrogênios/metabolismo , Modelos Animais , Transdução de Sinais , Animais , Humanos
15.
Artigo em Inglês | MEDLINE | ID: mdl-28144231

RESUMO

Androgen receptor (AR) is a validated drug target for prostate cancer based on its role in proliferation, survival, and metastases of prostate cancer cells. Unfortunately, despite recent improvements to androgen deprivation therapy and the advent of better antiandrogens with a superior affinity for the AR ligand-binding domain (LBD), most patients with recurrent disease will eventually develop lethal metastatic castration-resistant prostate cancer (CRPC). Expression of constitutively active AR splice variants that lack the LBD contribute toward therapeutic resistance by bypassing androgen blockade and antiandrogens. In the canonical pathway, binding of androgen to AR LBD triggers the release of AR from molecular chaperones which enable conformational changes and protein-protein interactions to facilitate its nuclear translocation where it regulates the expression of target genes. However, preceding AR function in the nucleus, initial binding of androgen to AR LBD in the cytoplasm may already initiate signal transduction pathways to modulate cellular proliferation and migration. In this article, we review the significance of signal transduction pathways activated by rapid, non-genomic signaling of the AR during the progression to metastatic CRPC and put into perspective the implications for current and novel therapies that target different domains of AR.

16.
Cell Signal ; 28(5): 348-356, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26829214

RESUMO

The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.


Assuntos
Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Antagonistas de Receptores de Andrógenos/uso terapêutico , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Próstata/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Transcrição Gênica
17.
Methods Mol Biol ; 1366: 471-488, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26585158

RESUMO

Estrogens are important modulators of a broad spectrum of physiological functions in humans. However, despite their beneficial actions, a number of lines of evidence correlate the sustained exposure to exogenous estrogen with increased risk of the onset of various cancers. Mainly these steroid hormones induce their effects by binding and activating estrogen receptors (ERα and ERß). These receptors belong to the family of ligand-regulated transcription factors, and upon activation they regulate the expression of different target genes by binding directly to specific DNA sequences. On the other hand, in recent years it has become clear that the G protein-coupled estrogen receptor 30 (GPR30/GPER) is able to mediate non-genomic action of estrogens in different cell contexts. In particular, GPER has been shown to specifically bind estrogens, and in turn to functionally cross-react with diverse cell signaling systems such as the epidermal growth factor receptor (EGFR) pathway, the Notch signaling pathway and the mitogen-activated protein kinases (MAPK) pathway. In this chapter we will present some of the different experimental techniques currently used to demonstrate the functional role of GPER in mediating non-genomic actions of estrogens, such as the dual luciferase assay, assessment of the involvement of GPER in the stimulation of cell migration in breast cancer cell lines and in cancer-associated fibroblasts, and chromatin immunoprecipitation assay. Overall, the experimental procedures described herein represent key instruments for assessing the biological role of GPER in mediating non-genomic signals of estrogen.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/farmacologia , Fibroblastos/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Estrogênios/farmacologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fluxo de Trabalho
18.
Neurosci Lett ; 612: 32-37, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26673886

RESUMO

Filial imprinting is a behavior characterized by the sensitive or critical period restricted to the first few days after hatching. Once the sensitive period is closed, it is widely believed that chicks can never be imprinted under natural conditions. Previously, we showed that the exogenous injection of T3 reopened the sensitive period which was already closed. That study suggested that T3 functioned by way of a rapid non-genomic action; however, the molecular mechanism of how T3 reopens the sensitive period remains unknown. Here, we show that the phosphorylation level of nucleotide diphosphate kinase 2 (NDPK2) was upregulated following T3 injection. Pharmacological deprivation of the kinase activity of NDPK hampered the molecular process prerequisite for the reopening of the sensitive period of filial imprinting. Moreover, it is shown that the kinase activity of NDPK2 participates in the priming process by T3 signaling which endows the potential for learning. Our data indicate that NDPK2 plays a crucial role downstream of T3 action and that its phosphorylation is involved in the non-genomic signaling during imprinting.


Assuntos
Galinhas/fisiologia , Fixação Psicológica Instintiva , Núcleosídeo-Difosfato Quinase/metabolismo , Animais , Encéfalo/metabolismo , Galinhas/crescimento & desenvolvimento , Núcleosídeo-Difosfato Quinase/antagonistas & inibidores , Fosfoproteínas/metabolismo , Fosforilação , Proteoma/metabolismo , Tri-Iodotironina/farmacologia , Regulação para Cima
19.
Acta Histochem ; 118(4): 377-86, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27094116

RESUMO

The present study investigated the influence of the androgen receptor (AR) agonists testosterone (T) and dihydrotestosterone (DHT), and vinclozolin (Vnz), a fungicide with antiandrogenic activity, on non-genomic signal transduction within ovarian follicles. Porcine granulosa cells (GCs) isolated from mature follicles were cultured for 48h. For the last 24h of culture, they were exposed to T (10(-7)M), DHT (10(-7)M), Vnz (1.4×10(-5)M), T and Vnz (T+Vnz), or DHT and Vnz (DHT+Vnz) at the same concentrations. To better imitate in vivo conditions, whole follicles (4-6mm in diameter) were incubated (24h) in an organ culture system with the same factors. Expression of AR mRNA and protein was determined by real-time PCR and western blot analyses. To demonstrate AR localization in cultured GCs and whole follicles, immunocytochemistry and immunohistochemistry were performed, respectively. To elucidate the possible non-genomic action of Vnz in GCs, protein expression and the activity of ERK1/2 and Akt kinases were determined by western blot and ELISA analyses. The immunocytochemistry and immunohistochemistry results showed that exposure of GCs and follicles to Vnz resulted in cytoplasmic and perinuclear AR localization. Real-time PCR and western blot analysis showed that AR mRNA and protein expression increased (P≤0.001) in GC cultures after combined treatment with an androgen and Vnz. In whole follicles, such treatment also increased AR mRNA with a decrease in the respective protein expression (P≤0.001). Moreover, addition of T or DHT with Vnz increased the activity of ERK1/2 and Akt kinases in cultured GCs (P≤0.001). The results suggest a novel mechanism for Vnz action in porcine ovarian follicles on both AR mRNA and protein levels. Thus, this environmental antiandrogen activates non-genomic signaling pathways, as indicated by the increased activity of both investigated kinases observed within minutes of Vnz addition. Given the widespread presence of Vnz in the environment, elucidation of its non-genomic action should be the subject of studies on female fertility.


Assuntos
Antagonistas de Androgênios/farmacologia , Células da Granulosa/metabolismo , Oxazóis/farmacologia , Receptores Androgênicos/metabolismo , Animais , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Avaliação Pré-Clínica de Medicamentos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Androgênicos/genética , Transdução de Sinais , Sus scrofa
20.
Toxicol Sci ; 153(1): 174-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27413106

RESUMO

Methoxychlor (MXC) and vinclozolin (VIN) are well-recognized endocrine disrupting chemicals known to alter epigenetic regulations and transgenerational inheritance; however, non-endocrine disruption endpoints are also important. Thus, we determined the effects of MXC and VIN on the dysregulation of gap junctional intercellular communication (GJIC) and activation of mitogen-activated protein kinases (MAPKs) in WB-F344 rat liver epithelial cells. Both chemicals induced a rapid dysregulation of GJIC at non-cytotoxic doses, with 30 min EC50 values for GJIC inhibition being 10 µM for MXC and 126 µM for VIN. MXC inhibited GJIC for at least 24 h, while VIN effects were transient and GJIC recovered after 4 h. VIN induced rapid hyperphosphorylation and internalization of gap junction protein connexin43, and both chemicals also activated MAPK ERK1/2 and p38. Effects on GJIC were not prevented by MEK1/2 inhibitor, but by an inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC), resveratrol, and in the case of VIN, also, by a p38 inhibitor. Estrogen (ER) and androgen receptor (AR) modulators (estradiol, ICI 182,780, HPTE, testosterone, flutamide, VIN M2) did not attenuate MXC or VIN effects on GJIC. Our data also indicate that the effects were elicited by the parental compounds of MXC and VIN. Our study provides new evidence that MXC and VIN dysregulate GJIC via mechanisms involving rapid activation of PC-PLC occurring independently of ER- or AR-dependent genomic signaling. Such alterations of rapid intercellular and intracellular signaling events involved in regulations of gene expression, tissue development, function and homeostasis, could also contribute to transgenerational epigenetic effects of endocrine disruptors.


Assuntos
Inseticidas/toxicidade , Fígado/efeitos dos fármacos , Metoxicloro/toxicidade , Oxazóis/toxicidade , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Conexina 43/metabolismo , Junções Comunicantes/efeitos dos fármacos , Fígado/citologia , Fígado/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Células-Tronco/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA