Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Pharmaceuticals (Basel) ; 16(11)2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-38004379

RESUMO

Ovarian cancer (OC) is one of the most lethal gynecological malignancies. The use of biological compounds such as non-coding RNAs (ncRNAs) is being considered as a therapeutic option to improve or complement current treatments since the deregulation of ncRNAs has been implicated in the pathogenesis and progression of OC. Old drugs with antitumoral properties have also been studied in the context of cancer, although their antitumor mechanisms are not fully clear. For instance, the antidiabetic drug metformin has shown pleiotropic effects in several in vitro models of cancer, including OC. Interestingly, metformin has been reported to regulate ncRNAs, which could explain its diverse effects on tumor cells. In this review, we discuss the mechanism of epigenetic regulation described for metformin, with a focus on the evidence of metformin-dependent microRNA (miRNAs) and long non-coding RNA (lncRNAs) regulation in OC.

2.
Front Endocrinol (Lausanne) ; 14: 1143261, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37056674

RESUMO

For many years, luteinizing hormone-releasing hormone or gonadotropin-releasing hormone (GnRH) analogs have been used to treat androgen or estrogen-dependent tumors. However, emerging evidence shows that the GnRH receptor (GnRH-R) is overexpressed in several cancer cells, including ovarian, endometrial, and prostate cancer cells, suggesting that GnRH analogs could exert direct antitumoral actions in tumoral tissues that express GnRH-R. Another recent approach based on this knowledge was the use of GnRH peptides for developing specific targeted therapies, improving the delivery and accumulation of drugs in tumoral cells, and decreasing most side effects of current treatments. In this review, we discuss the conventional uses of GnRH analogs, together with the recent advances in GnRH-based drug delivery for ovarian, breast, and prostatic cancer cells.


Assuntos
Hormônio Liberador de Gonadotropina , Neoplasias da Próstata , Masculino , Feminino , Humanos , Neoplasias da Próstata/tratamento farmacológico , Próstata , Ovário , Sistemas de Liberação de Medicamentos
3.
Int J Mol Sci ; 23(7)2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35409282

RESUMO

Polycystic ovary syndrome (PCOS) is an endocrine/metabolic disorder associated with insulin resistance (IR) and obesity. Endometria from women with PCOS present failures in insulin action, glucose uptake and signaling of insulin-sensitizing molecules, such as adiponectin, with consequences for reproduction. Metformin (MTF) treatment improves insulin signaling in endometrial tissues, but its mechanism is not fully understood. This study addresses the MTF effect, as well as adiponectin agonist action, on levels of molecules associated with insulin and adiponectin signaling pathways in endometrial tissue and cells, as assessed by immunohistochemistry and immunocytochemistry, respectively. Endometrial tissues were obtained from women and divided into five groups: Normal Weight (control); Obesity + IR; Obesity + IR + PCOS; Obesity + IR + MTF; Obesity + IR + PCOS + MTF. Endometrial cells stimulated with TNFα (as obesity-marker) were also used to partially emulate an obesity environment. The results showed low levels of insulin/adiponectin signaling in the endometria from women with obesity, IR and PCOS compared with the control group. MTF re-established these levels, independently of PCOS. TNFα-associated molecules were elevated in pathologic endometria, whereas MTF diminished these levels. The low levels of insulin/adiponectin molecules in endometrial cells treated with TNFα were reverted by MTF, similar to what was observed in the case of the adiponectin agonist. Therefore, independently of PCOS, MTF can re-establish levels of molecules involved in insulin/adiponectin signaling in endometrial cells, suggesting an improvement in insulin action and reproductive failures observed in endometria from women with obesity/PCOS.


Assuntos
Resistência à Insulina , Metformina , Síndrome do Ovário Policístico , Adiponectina/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Insulina/metabolismo , Metformina/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
4.
Int J Mol Sci ; 23(4)2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35216240

RESUMO

Nerve growth factor (NGF) and its high-affinity receptor TRKA are overexpressed in epithelial ovarian cancer (EOC) displaying a crucial role in the disease progression. Otherwise, NGF interacts with its low-affinity receptor P75, activating pro-apoptotic pathways. In neurons, P75 could be cleaved by metalloproteinases (α and γ-secretases), leading to a decrease in P75 signaling. Therefore, this study aimed to evaluate whether the shedding of P75 occurs in EOC cells and whether NGF/TRKA could promote the cleavage of the P75 receptor. The immunodetection of the α-secretase, ADAM17, TRKA, P75, and P75 fragments was assessed by immunohisto/cytochemistry and Western blot in biopsies and ovarian cell lines. The TRKA and secretases' inhibition was performed using specific inhibitors. The results show that P75 immunodetection decreased during EOC progression and was negatively correlated with the presence of TRKA in EOC biopsies. NGF/TRKA increases ADAM17 levels and the fragments of P75 in ovarian cells. This effect is abolished when cells are previously treated with ADAM17, γ-secretase, and TRKA inhibitors. These results indicate that NGF/TRKA promotes the shedding of P75, involving the activation of secretases such as ADAM17. Since ADAM17 has been proposed as a screening marker for early detection of EOC, our results contribute to understanding better the role of ADAM17 and NGF/TRKA in EOC pathogenesis, which includes the NGF/TRKA-mediated cleavage of P75.


Assuntos
Proteína ADAM17/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator de Crescimento Neural/metabolismo , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Receptor trkA/metabolismo , Fatores de Transcrição/metabolismo , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Pessoa de Meia-Idade , Neurônios/metabolismo , Neoplasias Ovarianas/patologia , Ovário/patologia , Transdução de Sinais/fisiologia
5.
Artigo em Espanhol | LILACS-Express | LILACS | ID: biblio-1506434

RESUMO

Introducción: Desde el registro de los primeros casos de COVID-19 en México, se han derivado una serie de respuestas emocionales caracterizadas por miedo y estrés. Dicho impacto emocional se debe en gran medida a la inundación de información paralela a las fases de la pandemia y la transición entre ellas y la percepción que los individuos tienen de la enfermedad. El objetivo del presente trabajo fue comparar la percepción del COVID-19 entre la fase 1 y 2 de la pandemia y entre los medios de información usados para informarse en población mexicana. Métodos: Considerando un muestreo en cadena, se realizó un estudio comparativo en el que se diseminó por medio de correo electrónico y redes sociales una batería de evaluación que respondieron 1560 participantes. Resultados: La preocupación por las consecuencias del COVID-19 y su impacto emocional incrementaron al pasar de la fase 1 a la fase 2 de la pandemia. Además, se identificó que el impacto emocional fue mayor en quienes se informaron a través de Facebook® y televisión. Conclusiones: La pandemia tendrá un impacto emocional progresivo en medida en que avancen sus fases y en la importancia de informarse en medios adecuados para prevenir consecuencias emocionales.


Background: Since the first COVID-19 cases in Mexico there have been a variety of emotional responses which have in common fear and stress. The emotional impact of COVID-19 is builded in some way because the information flooding parallel to the pandemic phases, the transition between them and illness perception. The aim of the present work was to compare the perception of COVID-19 between phase 1 and 2 of the pandemic and between the information media used to inform themselves in the Mexican population. Methods: Considering a chain sampling, a comparative study was carried out in which an evaluation battery was disseminated through email and social networks, which was answered by 1560 participants. Results The concern about the consequences of COVID-19 and its emotional impact increased when going from phase 1 to phase 2 of the pandemic. In addition, it was identified that the emotional impact was greater in those who reported through Facebook® and television. Conclusions: The pandemic will have a progressive emotional impact as its phases progress and the importance of informing oneself in adequate means to prevent emotional consequences.

6.
Salud ment ; 43(6): 285-292, Nov.-Dec. 2020. tab, graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1156875

RESUMO

Abstract Introduction The prevention of COVID-19 infections involves the implementation of behaviors to reduce risk and protect health. However, engaging in these behaviors depends on the perception of the threat posed by the illness. Previous research shows the importance of illness perception in the case of communicable and non-communicable diseases, showing that they can change depending on the severity and risk attributed to them. Objective Compare the illness perception and the practice of preventive and exposure behavior based on the severity and the risk attributed to COVID-19 at the end of phase 1 and the beginning of phase 2 of the pandemic in Mexico. Method By means of a chain sampling, a comparative study was conducted in which an evaluation battery was disseminated through e-mail and social networks. Results It was found that evaluating COVID-19 as a serious disease and perceiving oneself as being at risk of contracting it had small and moderate effects on the perception of the consequences of the illness (r = .34; r = .26), emotional impact (r = .32; r = .25), personal control (r = .24) and engagement in preventive (r = .05), and exposure behaviors (r = .07; r = .07). Discussion and conclusion This article shows the relevance of the perceptual variables that impact concern due to the social and emotional consequences of COVID-19, as well as those that encourage preventive behaviors and the minimization of exposure behavior.


Resumen Introducción Si bien la prevención de contagios de COVID-19 supone la implementación de comportamientos para reducir el riesgo y proteger la salud, la práctica de estas conductas está en función de la percepción que se tenga sobre la amenaza de la enfermedad. Investigaciones previas evidencian la importancia de la percepción de enfermedad cuando se trata de padecimientos transmisibles y no transmisibles, señalando que ésta puede modificarse en función de la gravedad y el riesgo atribuido. Objetivo Comparar la percepción de enfermedad y la práctica de conductas de prevención y exposición con base en la gravedad y el riesgo atribuidos al COVID-19 al término de la fase 1 e inicio de la fase 2 de la pandemia en México. Método A partir de un muestreo en cadena, se realizó un estudio comparativo en el que se diseminó una batería de evaluación por medio de correo electrónico y redes sociales. Resultados Se encontró que evaluar el COVID-19 como una enfermedad grave y percibirse en riesgo de contraerla tiene efectos moderados y leves, respectivamente, sobre la percepción de las consecuencias de la enfermedad (r = .34; r = .26), el impacto emocional (r = .32; r = .25), el control personal (r = .24) y la práctica de conductas de prevención (r = .05) y exposición (r = .07; r = .07). Discusión y conclusión El presente estudio muestra la relevancia de las variables perceptuales que impactan en la preocupación por consecuencias sociales y emocionales del COVID-19, así como de aquellas que favorecen las conductas de prevención y la minimización de conducta de exposición.

7.
Pharmaceuticals (Basel) ; 13(10)2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-33081077

RESUMO

Epithelial ovarian cancer (EOC) is a lethal gynaecological neoplasm characterized by rapid growth and angiogenesis. Nerve growth factor (NGF) and its high affinity receptor tropomyosin receptor kinase A (TRKA) contribute to EOC progression by increasing the expression of c-MYC, survivin and vascular endothelial growth factor (VEGF) along with a decrease in microRNAs (miR) 23b and 145. We previously reported that metformin prevents NGF-induced proliferation and angiogenic potential of EOC cells. In this study, we sought to obtain a better understanding of the mechanism(s) by which metformin blocks these NGF-induced effects in EOC cells. Human ovarian surface epithelial (HOSE) and EOC (A2780/SKOV3) cells were stimulated with NGF and/or metformin to assess the expression of c-MYC, ß-catenin, survivin and VEGF and the abundance of the tumor suppressor miRs 23b and 145. Metformin decreased the NGF-induced transcriptional activity of MYC and ß-catenin/T-cell factor/lymphoid enhancer-binding factor (TCF-Lef), as well as the expression of c-MYC, survivin and VEGF in EOC cells, while it increased miR-23b and miR-145 levels. The preliminary analysis of ovarian biopsies from women users or non-users of metformin was consistent with these in vitro results. Our observations shed light on the mechanisms by which metformin may suppress tumour growth in EOC and suggest that metformin should be considered as a possible complementary therapy in EOC treatment.

8.
Histol Histopathol ; 35(9): 961-971, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32369181

RESUMO

Ovarian cancer is the first cause of death for gynecological malignances in developed countries and around 80% correspond to Epithelial Ovarian Cancer (EOC). Overexpression of Nerve Growth Factor (NGF) and its high affinity receptor TRKA are involved in EOC progression, modulating several oncogenic processes such as angiogenesis by the increase of Vascular Endothelial Growth Factor (VEGF). FSH receptors (FSH-R) are present in EOC, but their changes and contribution during EOC progression are still not thoroughly known. The aims of this study were to evaluate the abundance of FSH receptors during EOC differentiation and to determine whether FSH modulates oncoproteins such as NGF and VEGF in ovarian cells. FSH-R expression in EOC tissues and cell lines (A2780, poorly differentiated EOC cells and HOSE, non-tumoral ovarian surface epithelial cells) were measured by RT-PCR and laser capture of epithelial cells from EOC samples by qPCR. FSH-R protein levels were evaluated by immunohisto/cytochemistry. Additionally, ovarian explants and ovarian cell lines were stimulated with FSH and/or FSH-R inhibitor to assess NGF and VEGF mRNA and protein levels. The results showed that FSH-R levels decreased during EOC progression, nevertheless these receptors are still present in poorly differentiated EOC. FSH increased NGF expression in ovarian cells, which was prevented using a FSH-R inhibitor. Similarly, in ovarian cancer explants, FSH increased NGF and VEGF mRNA, as well as NGF protein levels. These results suggest that FSH would display a key role not only in initial stages of EOC, but also in late stages of this disease, by modulation of NGF and VEGF levels in EOC cells.


Assuntos
Cistadenocarcinoma Seroso/metabolismo , Células Epiteliais/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Fator de Crescimento Neural/metabolismo , Neoplasias Ovarianas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular , Cistadenocarcinoma Seroso/patologia , Células Epiteliais/metabolismo , Feminino , Humanos , Neoplasias Ovarianas/patologia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Receptores do FSH/metabolismo
9.
Reprod Sci ; 27(1): 290-300, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-32046436

RESUMO

A pro-inflammatory environment is characteristic of obesity and polycystic ovary syndrome (PCOS). This environment through cytokines secretion negatively affects insulin action. Endometria from women with both conditions (obesity and PCOS) present high TNF-α level and altered insulin signaling. In addition, these patients present reproductive failures that could be associated to an abnormal endometrial function. Here, TNF-α and IL-6 effects on insulin signaling pathway were evaluated. Serum and endometrial IL-6, phospho-IRS1-S270 (inactive form) and phospho-IRS1-Y612 (active form) levels were evaluated in women with: Normal-Weight, Obesity and Obesity-PCOS. In endometrial cells under hyperandrogenic/hyperinsulinic conditions resembling PCOS, it was evaluated IL-6/TNF-α effects on phospho-IRS1-S270, phospho-IRS1-Y612, phospho-AKT-S473 levels, and S6K and JNK activation (IRS1-inactivating molecules). In obesity groups, diminution of IRS1-active form was observed, being more significantly in Obesity-PCOS; whereas, IRS1-inactive form increased in Obesity-PCOS. Serum and endometrial IL-6 were higher in Obesity-groups compared to Normal-Weight. In endometrial cells, TNF-α increases phospho-IRS1-S270, while IL-6 decreases phospho-IRS1-Y612. Importantly, TNF-α and IL-6 promote S6K and JNK activation; TNF-α increases and IL-6 decreases phospho-AKT-S473 levels. Thus, pro-inflammatory cytokines in endometrium could negatively influence insulin signaling by different mechanisms: TNF-α promotes activation of IRS1-inactivating kinases, whereas, IL-6 decreases IRS1 and AKT activation. Moreover, when obesity and PCOS are present the disruption of insulin signaling is aggravated. These effects could explain endometrial abnormal function and reproductive failures observed in women with obesity and PCOS.


Assuntos
Endométrio/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Interleucina-6/farmacologia , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Adulto , Linhagem Celular , Endométrio/efeitos dos fármacos , Endométrio/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Obesidade/patologia , Fosforilação/efeitos dos fármacos , Síndrome do Ovário Policístico/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Adulto Jovem
10.
Am J Physiol Endocrinol Metab ; 318(2): E237-E248, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31874063

RESUMO

Polycystic ovary syndrome (PCOS) is an endocrine-metabolic disorder characterized by hyperandrogenism and ovulatory dysfunction but also obesity and hyperinsulinemia. These characteristics induce an insulin-resistant state in tissues such as the endometrium, affecting its reproductive functions. Myo-inositol (MYO) is an insulin-sensitizing compound used in PCOS patients; however, its insulin-sensitizing mechanism is unclear. To understand the relationship of MYO with insulin action in endometrial cells, sodium/myo-inositol transporter 1 (SMIT-1) (MYO-transporter), and MYO effects on protein levels related to the insulin pathway were evaluated. SMIT-1 was assessed in endometrial tissue from women with normal weight, obesity, insulin resistance, and PCOS; additionally, using an in vitro model of human endometrial cells exposed to an environment resembling hyperinsulinemic-obese-PCOS, MYO effect was evaluated on p-AMPK and GLUT-4 levels and glucose uptake by Western blot, immunocytochemistry, and confocal microscopy, respectively. SMIT-1 was detected in endometrial tissue from all groups and decreased in PCOS and obesity (P < 0.05 vs. normal weight). In the in vitro model, PCOS conditions decreased p-AMPK levels, while they were restored with MYO (P < 0.05). The diminished GLUT-4 protein levels promoted by PCOS environment were restored by MYO through SMIT-1 and p-AMPK-dependent mechanism (P < 0.05). Also, MYO restored glucose uptake in cells under PCOS condition through a p-AMPK-dependent mechanism. Finally, these results were similar to those obtained with metformin treatment in the same in vitro conditions. Consequently, MYO could be a potential insulin sensitizer through its positive effects on insulin-resistant tissues as PCOS-endometrium, acting through SMIT-1, provoking AMPK activation and elevated GLUT-4 levels and, consequently, increase glucose uptake by human endometrial cells. Therefore, MYO may be used as an effective treatment option in insulin-resistant PCOS women.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Endométrio/metabolismo , Transportador de Glucose Tipo 4/biossíntese , Inositol/metabolismo , Resistência à Insulina , Síndrome do Ovário Policístico/metabolismo , Adulto , Células Cultivadas , Endométrio/citologia , Ativação Enzimática , Feminino , Glucose/metabolismo , Transportador de Glucose Tipo 4/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Obesidade/metabolismo , Síndrome do Ovário Policístico/genética , Simportadores/metabolismo
11.
Cancers (Basel) ; 11(12)2019 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-31817839

RESUMO

Epithelial ovarian cancer (EOC) is a lethal gynecological neoplasia characterized by extensive angiogenesis and overexpression of nerve growth factor (NGF). Here, we investigated the mechanism by which NGF increases vascular endothelial growth factor (VEGF) expression and the vasculogenic potential of EOC cells, as well as the contribution of the cyclooxygenase 2/prostaglandin E2 (COX-2/PGE2) signaling axis to these events. EOC biopsies and ovarian cell lines were used to determine COX-2 and PGE2 levels, as well as those of the potentially pro-angiogenic proteins c-MYC (a member of the Myc transcription factors family), survivin, and ß-catenin. We observed that COX-2 and survivin protein levels increased during EOC progression. In the EOC cell lines, NGF increased the COX-2 and PGE2 levels. In addition, NGF increased survivin, c-MYC, and VEGF protein levels, as well as the transcriptional activity of c-MYC and ß-catenin/T-cell factor/lymphoid enhancer-binding factor (TCF-Lef) in a Tropomyosin receptor kinase A (TRKA)-dependent manner. Also, COX-2 inhibition prevented the NGF-induced increases in these proteins and reduced the angiogenic score of endothelial cells stimulated with conditioned media from EOC cells. In summary, we show here that the pro-angiogenic effect of NGF in EOC depends on the COX-2/PGE2 signaling axis. Thus, inhibition COX-2/PGE2 signaling will likely be beneficial in the treatment of EOC.

12.
Front Oncol ; 9: 913, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31608227

RESUMO

Angiogenesis, or generation of new blood vessels from other pre-existing, is a key process to maintain the supply of nutrients and oxygen in tissues. Unfortunately, this process is exacerbated in pathologies such as retinopathies and cancers with high angiogenesis as ovarian cancer. Angiogenesis is regulated by multiple systems including growth factors and neurotrophins. One of the most studied angiogenic growth factors is the vascular endothelial growth factor (VEGF), which is overexpressed in several cancers. It has been recently described that neurotrophins could regulate angiogenesis through direct and indirect mechanisms. Neurotrophins are a family of proteins that include nerve growth factor (NGF), brain-derived growth factor (BDNF), and neurotrophins 3 and 4/5 (NT 3, NT 4/5). These molecules and their high affinity receptors (TRKs) regulate the development, maintenance, and plasticity of the nervous system. Furthermore, it was recently described that they display essential functions in non-neuronal tissues, such as reproductive organs among others. Studies have shown that several types of cancer overexpress neurotrophins such as NGF and BDNF, which might contribute to tumor progression and angiogenesis. Besides, in recent years the FDA has approved the use of pharmacologic inhibitors of pan-TRK receptors in patients with TRKs fusion-positive cancers. In this review, we discuss the mechanisms by which neurotrophins stimulate tumor progression and angiogenesis, with emphasis on gynecological cancers.

13.
Ther Adv Med Oncol ; 10: 1758835918770984, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29774060

RESUMO

BACKGROUND: Epithelial ovarian cancer (EOC) is characterized by exacerbated angiogenesis regulated by proangiogenic and growth factors. Nerve growth factor (NGF) is overexpressed in EOC where it promotes proliferation as well as survival and is considered a proangiogenic factor. Metformin, a drug commonly used in the treatment of diabetes, is attributed to antineoplastic effects, but the underlying mechanisms remain unknown. Given that current therapies yield modest results in EOC patients, the aim of this study was to determine the effects of metformin on NGF-enhanced proliferation of EOC cells and the angiogenic potential of endothelial cells. METHODS: A2780 (EOC), HOSE (human ovarian surface epithelial) and EA.hy926 (endothelial) cells were treated with NGF and metformin. Cell viability, cell proliferation and cell cycle were evaluated in all three cell lines, and the angiogenic potential in endothelial EA.hy926 cells. RESULTS: NGF enhanced cell proliferation in A2780, HOSE and EA.hy926 cells (p < 0.05), while metformin treatment decreased cell proliferation in A2780 and EA.hy926 cells (p < 0.05). Moreover, the NGF-enhanced angiogenic score in EA.hy926 cells was prevented by metformin (p < 0.05). CONCLUSIONS: Given that NGF plays a significant role in EOC progression, our current findings suggest that metformin holds considerable promise as an adjuvant treatment in ovarian cancer.

14.
Reprod Sci ; 25(7): 1000-1009, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28946816

RESUMO

Polycystic ovary syndrome (PCOS) affects not only ovarian functions but is also able to affect endometrium metabolism. Around 80% of women with PCOS are obese. High tumor necrosis factor (TNF)-α production and low adiponectin levels are characteristics of obesity. Interestingly, endometrium from obese women with PCOS presents an insulin-resistance condition, high TNF-α levels, and low adiponectin levels. However, TNF-α effect on molecules associated with insulin action in endometrial cells remains unclear. Therefore, the objective of this work was to evaluate TNF-α effect on expression of molecules associated with adiponectin (insulin sensitizing) and TNF-α signaling pathways and on Glucose Transporter type 4 (GLUT-4) levels in human endometrial cells under the characteristic conditions of hyperandrogenic/hyperinsulinic (HA/HI) PCOS. Two human endometrial stromal cell lines (T-HESC/St-T1b) under HA/HI conditions were used to assay the effect of high TNF-α concentration (100 ng/mL) on adiponectin, AdipoR1-AdipoR2 receptors, Adaptor protein phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1), Phospho-AMP-activated protein kinase T172 (p-AMPKT172), GLUT-4, Tumor necrosis factor receptor 1 (TNFR1)-Tumor necrosis factor receptor 2 (TNFR2) receptors protein levels, and nuclear factor κB (NFκB) nuclear content, by Western blot or immunocytochemistry. The NFκB participation in TNF-α effect on adiponectin expression was assayed using an NFκB inhibitor (pyrrolidine dithiocarbamate). The TNF-α increases the expression of molecules associated with its own signaling pathway ( P < .05) and decreases the protein levels of adiponectin and its associated molecules ( P < .05). Moreover, TNF-α increases NFκB nuclear content ( P < .001), whereas with NFκB inhibition the decrease in adiponectin content induced by TNF-α was not observed. GLUT-4 levels were lower with TNF-α treatment ( P < .01). Thus, in human endometrial stromal cells, high TNF-α levels negatively affect the insulin action through decreased adiponectin signaling and GLUT-4 protein. This could explain the failures observed in endometrial function of obese women with PCOS.


Assuntos
Endométrio/metabolismo , Insulina/metabolismo , Síndrome do Ovário Policístico/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adiponectina/metabolismo , Células Cultivadas , Endométrio/citologia , Feminino , Transportador de Glucose Tipo 4 , Humanos , Insulina/administração & dosagem , NF-kappa B/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem
15.
Steroids ; 126: 85-91, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28827068

RESUMO

Normal endometrial function requires of cell proliferation and differentiation; therefore, disturbances in these processes could lead to pathological entities such as hyperplasia and endometrial adenocarcinoma, where cell proliferation is increased. The development of these pathologies is highly related to alterations in the levels and/or action of sexual steroids. In the present review, it has been analyzed how steroids, particularly estrogens, androgens and progestagens are involved in the etiopathogenesis of hyperplasia and endometrial endometrioid adenocarcinoma. The emphasis is given on pathological and pharmacological conditions that are presented as risk factors for endometrial pathologies, such as obesity, polycystic ovarian syndrome and hormone replacement postmenopausal women therapy, among others. Steroids alterations may promote changes at molecular level that enhance the development of hyperplasia and endometrioid cancer. In fact, there are solid data that indicate that estrogens stimulate cell-proliferation in this tissue; meanwhile, progestagens are able to stop cell proliferation and to increase differentiation. Nevertheless, the role of androgens is less clear, since there is contradictory information. It is most likely that the major contribution of steroids to the development of cell proliferation pathologies in endometria would be in early stages, where there is a high sensitivity to these molecules. This phenomenon is present even in stages previous to the occurrence of hyperplasia, like in the condition of polycystic ovarian syndrome, where the endometria have a greater sensitivity to steroids and high expression of cell cycle molecules. These abnormalities would contribute to the pathogenesis of hyperplasia and then in the progression to endometrioid adenocarcinoma.


Assuntos
Endométrio , Esteroides/metabolismo , Animais , Proliferação de Células , Endométrio/patologia , Feminino , Humanos , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Receptores de Esteroides/metabolismo
16.
Int J Oncol ; 50(4): 1261-1270, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28260038

RESUMO

Ovarian cancer is the seventh most common cancer among women worldwide, causing approximately 120,000 deaths every year. Immunotherapy, designed to boost the body's natural defenses against cancer, appears to be a promising option against ovarian cancer. Calreticulin (CRT) is an endoplasmic reticulum (ER) resident chaperone that, translocated to the cell membrane after ER stress, allows cancer cells to be recognized by the immune system. The nerve growth factor (NGF) is a pro-angiogenic molecule overexpressed in this cancer. In the present study, we aimed to determine weather NGF has an effect in CRT translocation induced by cytotoxic and ER stress. We treated A2780 ovarian cancer cells with NGF, thapsigargin (Tg), an ER stress inducer and mitoxantrone (Mtx), a chemotherapeutic drug; CRT subcellular localization was analyzed by immunofluorescence followed by confocal microscopy. In order to determine NGF effect on Mtx and Tg-induced CRT translocation from the ER to the cell membrane, cells were preincubated with NGF prior to Mtx or Tg treatment and CRT translocation to the cell surface was determined by flow cytometry. In addition, by western blot analyses, we evaluated proteins associated with the CRT translocation pathway, both in A2780 cells and human ovarian samples. We also measured NGF effect on cell apoptosis induced by Mtx. Our results indicate that Mtx and Tg, but not NGF, induce CRT translocation to the cell membrane. NGF, however, inhibited CRT translocation induced by Mtx, while it had no effect on Tg-induced CRT exposure. NGF also diminished cell death induced by Mtx. NGF effect on CRT translocation could have consequences in immunotherapy, potentially lessening the effectiveness of this type of treatment.


Assuntos
Calreticulina/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Neoplasias Epiteliais e Glandulares/imunologia , Fator de Crescimento Neural/metabolismo , Neoplasias Ovarianas/imunologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Calreticulina/imunologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Citometria de Fluxo , Humanos , Imunoterapia , Mitoxantrona/farmacologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais , Tapsigargina/farmacologia
17.
Int J Mol Sci ; 18(3)2017 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-28245631

RESUMO

Ovarian cancer is the eighth most common cancer in women worldwide, and epithelial ovarian cancer (EOC) represents 90% of cases. Nerve growth factor (NGF) and its high affinity receptor tyrosine kinase A receptor (TRKA) have been associated with the development of several types of cancer, including EOC; both NGF and TRKA levels are elevated in this pathology. EOC presents high angiogenesis and several molecules have been reported to induce this process. NGF increases angiogenesis through its TRKA receptor on endothelial cells, and by indirectly inducing vascular endothelial growth factor expression. Other molecules controlled by NGF include ciclooxigenase-2, disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) and calreticulin (CRT), proteins involved in crucial processes needed for EOC progression. These molecules could be modified through microRNA regulation, which could be regulated by NGF. MicroRNAs are the widest family of non-coding RNAs; they bind to 3'-UTR of mRNAs to inhibit their translation, to deadenilate or to degraded them. In EOC, a deregulation in microRNA expression has been described, including alterations of miR-200 family, cluster-17-92, and miR-23b, among others. Since the NGF-microRNA relationship in pathologies has not been studied, this review proposes that some microRNAs could be associated with NGF/TRKA activation, modifying protein levels needed for EOC progression.


Assuntos
MicroRNAs/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Fator de Crescimento Neural/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Animais , Biomarcadores , Carcinoma Epitelial do Ovário , Proteínas de Transporte , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Neoplasias Epiteliais e Glandulares/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/patologia , Ligação Proteica , Interferência de RNA , Transdução de Sinais
18.
Histol Histopathol ; 32(11): 1187-1196, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28116735

RESUMO

INTRODUCTION: Ovarian cancer presents a high angiogenesis (formation of new blood vessels) regulated by pro-angiogenic factors, mainly vascular endothelial growth factor (VEGF) and nerve growth factor (NGF). An association between endogenous levels of estrogen and increased risk of developing ovarian cancer has been reported. Estrogen action is mediated by the binding to its specific receptors (ERα and ERß), altered ERα/ERß ratio may constitute a marker of ovarian carcinogenesis progression. OBJECTIVE: To determine the effect of estradiol through ERα on the expression of NGF and VEGF in epithelial ovarian cancer (EOC). METHODOLOGY: Levels of phosphorylated estrogen receptor alpha (pERα) were evaluated in well, moderate and poorly differentiated EOC samples (EOC-I, EOC-II, EOC-III). Additionally, ovarian cancer explants were stimulated with NGF (0, 10 and 100 ng/ml) and ERα, ERß and pERα levels were detected. Finally, human ovarian surface epithelial (HOSE) and epithelial ovarian cancer (A2780) cell lines were stimulated with estradiol, where NGF and VEGF protein levels were evaluated. RESULTS: In tissues, ERs were detected being pERα levels significantly increased in EOC-III samples compared with EOC-I (p<0.05). Additionally, ovarian explants treated with NGF increased pERα levels meanwhile total ERα and ERß levels did not change. Cell lines stimulated with estradiol revealed an increase of NGF and VEGF protein levels (p<0.05). CONCLUSIONS: Estradiol has a positive effect on pro-angiogenic factors such as NGF and VEGF expression in EOC, probably through the activation of ERα; generating a positive loop induced by NGF increasing pERα levels in epithelial ovarian cells.


Assuntos
Estradiol/farmacologia , Neoplasias Epiteliais e Glandulares/patologia , Fator de Crescimento Neural/biossíntese , Neoplasias Ovarianas/patologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Carcinoma Epitelial do Ovário , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/metabolismo , Fator de Crescimento Neural/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo
19.
Fetal Diagn Ther ; 41(3): 202-208, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27513943

RESUMO

OBJECTIVE: The aim of this study was to determine the role of nerve growth factor (NGF) in the first-trimester screening for preeclampsia (PE). METHODS: Uterine artery Doppler (UtAD) was determined transvaginally. Maternal concentrations of NGF were assessed in 42 patients who subsequently developed PE and in 95 controls. Quantile and multivariate regression analyses were performed for the NGF and UtAD adjustment and expressed as the multiple of the median (MoM) of the unaffected group. Logistic regression analysis was conducted to identify the best model for the prediction of PE. RESULTS: The maternal plasma concentration of NGF exhibited a trend towards lower values in patients who subsequently developed early-onset PE (e-PE) compared to controls (10.7 vs. 38.2 pg/ml, respectively; p = not significant). The median MoM NGF in the all-PE, e-PE and control groups was 0.97 (95% CI 0.13-3.36), 0.62 (95% CI 0.16-2.19) and 1.00 (95% CI 0.20-2.94), respectively (p = not significant). The best predictors of PE were previous PE, chronic hypertension and UtAD. With a false-positive rate of 10%, the detection rates (DRs) of all-PE and e-PE were 38 and 50%, respectively. The addition of MoM NGF did not improve the DR of PE. CONCLUSION: First-trimester NGF tends to be lower in patients who subsequently develop e-PE.


Assuntos
Fator de Crescimento Neural/sangue , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/diagnóstico por imagem , Primeiro Trimestre da Gravidez/sangue , Ultrassonografia Doppler em Cores , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Neovascularização Patológica/sangue , Neovascularização Patológica/diagnóstico por imagem , Projetos Piloto , Fator de Crescimento Placentário/sangue , Gravidez , Estudos Retrospectivos , Ultrassonografia Doppler em Cores/métodos
20.
Reprod Sci ; 24(7): 1079-1087, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27879454

RESUMO

Proliferation in endometria of women with polycystic ovarian syndrome (PCOS) is increased, similar to the biosynthesis of androstenediol (estrogenic metabolite). As previously shown, in human endometrial cells, androstenediol increases CYCLIN D1 levels and KI67 and decreases P27 content. The objective of the present investigation was to determine the mechanisms by which androstenediol promotes endometrial cell-cycle progression. Estrogen receptor α (ERα) activation and changes in CYCLIN D1 and P27 levels were evaluated by Western blot in T-HESC and St-T1b endometrial cell lines, using receptor antagonists; activation of PI3K-protein kinase B (AKT) and mitogen-activated protein kinases-extracellular signal-regulated kinases (MAPK-ERK)1/2 pathways was evaluated using PI3K, MAPK/ERK kinase (MEK)1/2, and RNA-polymerase II inhibitors. The data showed that androstenediol treatment significantly increases CYCLIN D1 and decreases P27 levels through ERα activation ( P < .05). In addition, an increase in AKT/ERK1/2 phosphorylations was determined ( P < .05). In the presence of RNA-polymerase II inhibitor, phosphorylation of AKT/ERK1/2 decreased ( P < .05), meaning that endometrial cells need transcriptional activity to activate the kinases involved. It was also observed that PI3K action is required for P27 and CYCLIN D1 changes. Therefore, the action of androstenediol in endometria depends on PI3K-AKT and MAPK-ERK1/2 pathways activation, together with cell transcriptional machinery. This could be of clinical significance, as in pathologies such as PCOS, increased endometrial levels of androstenediol together with a high prevalence of endometrial hyperplasia and adenocarcinoma have been reported.


Assuntos
Androstenodiol/farmacologia , Proliferação de Células/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Butadienos/farmacologia , Cromonas/farmacologia , Endométrio/citologia , Endométrio/metabolismo , Inibidores Enzimáticos/farmacologia , Estradiol/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Morfolinas/farmacologia , Nitrilas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/metabolismo , Testosterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA