Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Free Radic Biol Med ; 164: 369-380, 2021 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-33450374

RESUMEN

Metastasis is the major cause of death in women with advanced ovarian cancer. Epithelial ovarian cancer cells can dissociate directly from extracellular matrix (ECM) and form spheroids to spread through the peritoneal cavity. Loss of ECM hinders the survival of ECM-detached epithelial cells. It is still largely unknown how ovarian cancer spheroids maintain their viability after loss of ECM. We find that spheroids derived either from ovarian cancer ascites or cell lines are iron-replete. In accordance with iron-replete condition, proteins involved in iron uptake, transport and storage including divalent metal ion transporter 1 (DMT1), transferrin receptor 1 (TFR1), ferritin, poly(rC)-binding proteins 1 and 2 (PCBP1 and 2) and nuclear factor E2-related factor 2 (NRF2) all increase in ovarian cancer spheroids. Genes linking iron homeostasis and lipid metabolism including stearoyl coenzyme A desaturase 1 (SCD1) are up-regulated in ovarian cancer spheroids. The product of SCD1 oleic acid can restore the viability of ovarian cancer spheroids inhibited by deprivation of iron. Extracellular signal-regulated kinase (ERK) activation contributes to autophagy activation in ovarian cancer spheroids. Impairment of autophagy by U0126 or Olaparib results in lysosomal iron accumulation and decrease of the cytosolic labile iron pool, leading to reduction of SCD1, lipid level and cell viability. Combination of U0126 and Olaparib has synergistic cytotoxicity toward ovarian cancer spheroids. Our findings reveal that ovarian cancer spheroids develop efficient iron utilization system to survive. Targeting iron utilization in ovarian cancer spheroids may have the potential to become new treatment strategies for ovarian cancer metastasis.


Asunto(s)
Hierro , Neoplasias Ováricas , Carcinoma Epitelial de Ovario/genética , Línea Celular Tumoral , Matriz Extracelular , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Esferoides Celulares
2.
Cell Death Dis ; 11(9): 726, 2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901009

RESUMEN

Author Xiao Li was incorrectly associated with Department of Obstetrics and Gynaecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai 200233, China. The correct affiliation is Shanghai Municipal Key Clinical Speciality, Shanghai 20030, China.

3.
Cell Death Dis ; 11(8): 644, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811805

RESUMEN

Chronic stress has been shown to facilitate progression of epithelial ovarian cancer (EOC), however, the neuro-endocranial mechanism participating in this process still remains unclear. Here, we reported that chronic restraint stress (CRS) promoted the abdominal implantation metastasis of EOC cells and the expression of epithelial-mesenchymal transition-related markers in tumor-bearing mouse model, including TWIST, SLUG, SNAIL, and ß-catenin. We observed that ß-catenin co-expressed with SLUG and norepinephrine (NE) in tumor tissues obtained from nude mice. Further ex vivo experiments revealed that NE promoted migration and invasion of ovarian cancer cells and SLUG expression through upregulating expression and improving transcriptional function of ß-catenin in vitro. A human phosphor-kinase array suggested that NE activated various kinases in ovarian cancer cells, and we further confirmed that AKT inhibitor reduced NE-mediated pro-metastatic impacts and activation of the ß-catenin/SLUG axis. Furthermore, the expression levels of NE and ß-catenin were examined in ovarian tumor tissues by using tumor tissue arrays. Results showed that the expression levels of both NE and ß-catenin were associated with poor clinical stage of serous EOC. Moreover, we found that melatonin (MLT) effectively reduced the abdominal tumor burden of ovarian cancer induced by CRS, which was partially related to the inhibition of the NE/AKT/ß-catenin/SLUG axis. Collectively, these findings suggest a novel mechanism for CRS-mediated ovarian cancer metastasis and MLT has a potential therapeutic efficacy against ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Melatonina/farmacología , Animales , Carcinoma Epitelial de Ovario/fisiopatología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melatonina/metabolismo , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/fisiopatología , Norepinefrina/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Estrés Fisiológico/fisiología , beta Catenina/metabolismo
4.
Cell Cycle ; 19(13): 1677-1695, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32453975

RESUMEN

Chronic stress which is common in the current society can be harmful to female reproduction and is associated with oocyte defects. However, the underlying mechanisms remain largely unknown. Herein, by using a mouse model of chronic restraint stress, we demonstrated that chronic stress could induce meiotic spindle abnormalities, chromatin misalignment, mitochondrial dysfunction and elevated ROS levels in oocytes in vivo, all of which were normalized by the administration of melatonin. Consistently, melatonin treatment during in vitro maturation also attenuated the meiotic defects induced by H2O2 by regulating autophagy and SIRT1, which could be abolished by SIRT1 inhibitor, Ex527 and autophagy inhibitor Bafilomycin A1 (Baf A1). These data indicate that melatonin can mitigate chronic stress-induced oxidative meiotic defects in mice MII oocytes by regulating SIRT1 and autophagy, providing new understanding for stress-related meiotic errors in MII oocytes and suggesting melatonin and SIRT1 could be new targets for optimizing culture system of oocytes as well as fertility management.


Asunto(s)
Meiosis , Melatonina/farmacología , Oocitos/patología , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Sirtuina 1/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Cromosomas de los Mamíferos/metabolismo , ADN Mitocondrial/genética , Femenino , Dosificación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Macrólidos/farmacología , Meiosis/efectos de los fármacos , Meiosis/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Restricción Física , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
5.
Int J Oncol ; 55(1): 179-190, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31059002

RESUMEN

Epithelial ovarian cancer (EOC) is the most lethal of all gynecologic tumors. Cancer spheroid culture is a widely used model to study cancer stem cells. Previous studies have demonstrated the effectiveness of cytokine­induced killer (CIK) cell­based therapies against cancer and cancer stem cells. However, it is not clear how EOC spheroid cells respond to CIK­mediated cellular lysis, and the mechanisms involved have never been reported before. A flow cytometry­based method was used to evaluate the anti­cancer effects of CIK cells against adherent A2780 cells and A2780 spheroids. To demonstrate the association between hypoxia inducible factor­1α (HIF1A) and intercellular adhesion molecule­1 (ICAM­1), two HIF1A short hairpin RNA (shRNA) stable transfected cell lines were established. Furthermore, the protein expression levels of hypoxia/HIF1A­associated signaling pathways were evaluated, including transforming growth factor­ß1 (TGF­ß1)/mothers against decapentaplegic homologs (SMADs) and nuclear factor­κB (NF­κB) signaling pathways, comparing A2780 adherent cells and cancer spheroids. Flow cytometry revealed that A2780 spheroid cells were more resistant to CIK­mediated cellular lysis, which was partially reversed by an anti­ICAM­1 antibody. HIF1A was significantly upregulated in A2780 spheroids compared with adherent cells. Using HIF1A shRNA stable transfected cell lines and cobalt chloride, it was revealed that hypoxia/HIF1A contributed to downregulation of ICAM­1 in A2780 spheroid cells and adherent cells. Furthermore, hypoxia/HIF1A­associated signaling pathways, TGF­ß1/SMADs and NF­κB, were activated in A2780 spheroid cells by using western blotting. The findings indicate that EOC stem­like cells resist the CIK­mediated cellular lysis via HIF1A­mediated downregulation of ICAM­1, which may be instructive for optimizing and enhancing CIK­based therapies.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Células Asesinas Inducidas por Citocinas/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Células Madre Neoplásicas/citología , Neoplasias Ováricas/metabolismo , Adulto , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/terapia , Línea Celular Tumoral , Proliferación Celular , Células Asesinas Inducidas por Citocinas/trasplante , Regulación hacia Abajo , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Molécula 1 de Adhesión Intercelular/genética , Masculino , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia
6.
Mol Ther Nucleic Acids ; 16: 407-418, 2019 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-31022607

RESUMEN

Premature ovarian failure (POF) is one of the most common complications among female patients with tumors treated with chemotherapy and requires advanced treatment strategies. Human amniotic epithelial cell (hAEC)-based therapy mediates tissue regeneration in a variety of diseases, and increasing evidence suggests that the therapeutic efficacy of hAECs mainly depends on paracrine action. This study aimed to identify exosomes derived from hAECs and explored the therapeutic potential in ovaries damaged by chemotherapy and the underlying molecular mechanism. hAEC-derived exosomes exhibited a cup- or sphere-shaped morphology with a mean diameter of 100 nm and were positive for Alix, CD63, and CD9. hAEC exosomes increased the number of follicles and improved ovarian function in POF mice. During the early stage of transplantation, hAEC exosomes significantly inhibited granulosa cell apoptosis, protected the ovarian vasculature from damage, and were involved in maintaining the number of primordial follicles in the injured ovaries. Enriched microRNAs (miRNAs) existed in hAEC exosomes, and target genes were enriched in phosphatidylinositol signaling and apoptosis pathways. Studies in vitro demonstrated that hAEC exosomes inhibited chemotherapy-induced granulosa cell apoptosis via transferring functional miRNAs, such as miR-1246. Our findings demonstrate that hAEC-derived exosomes have the potential to restore ovarian function in chemotherapy-induced POF mice by transferring miRNAs.

7.
Acta Biochim Biophys Sin (Shanghai) ; 50(12): 1227-1235, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30395153

RESUMEN

Epithelial ovarian cancer (EOC) is the most fatal gynecologic malignancy due to its late diagnosis and lack of curative therapy. The antimalaria compound artemisinin and its derivatives, such as artesunate (ART) and dihydroartemisinin (DHA), have proven to be potent anticancer drugs and act through various anticancer mechanisms. To identify novel targets of artemisinin derivatives in EOC cells, we investigated the effects of ART and DHA on SKOV3 and primary EOC cell growth via CCK-8 assay. Both ART and DHA inhibited EOC cell growth. A cell cycle distribution analysis showed that ART and DHA caused G2/M cell cycle arrest. Moreover, ART and DHA induced autophagy in EOC cells, whereas autophagy inhibitors reversed the cell growth inhibition and cell cycle arrest induced by ART and DHA. Western blot analysis showed that ART and DHA also suppressed the cell cycle-related NF-κB-signaling pathway in EOC cells. These data suggest that artemisinin derivatives induce autophagy, block the cell cycle, and inhibit cell growth in EOC cells. Our research provides new targets for artemisinin derivatives for EOC treatment.


Asunto(s)
Artemisininas/farmacología , Artesunato/farmacología , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Antimaláricos/farmacología , Autofagia/genética , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , FN-kappa B/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos
8.
Oxid Med Cell Longev ; 2018: 7010472, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319732

RESUMEN

Epithelial ovarian cancer has the highest mortality rate of all gynecologic cancers. Cancer stem cells are considered to be the initiating cells of tumors. It is known that spheroid culture promotes ovarian cancer cells to acquire stem cell characteristics and to become stem cell-like. But the mechanisms remain largely unclear. Our data show that autophagy is sustainably activated in ovarian cancer spheroid cells. Inhibition of autophagy by knockdown of ATG5 abolishes the self-renewal ability of ovarian cancer spheroid cells. Knockdown of ATG5 prevents ovarian cancer spheroid cells to enter quiescent state. Autophagy is critical for quiescent ovarian cancer spheroid cells to reenter the cell cycle because rapamycin can promote quiescent ovarian cancer spheroid cells to form colonies on soft agar and knockdown of ATG5 can arrest ovarian cancer cells in G0/G1. Autophagy and NRF2 form a positive feedback regulation loop to regulate reactive oxygen species (ROS) levels in ovarian cancer spheroid cells. The optimal ROS level, neither too high nor too low, facilitates the self-renewal marker, NOTCH1, to reach to the highest level. Bafilomycin A1 can impair the self-renewal of ovarian cancer spheroid cells by disturbing ROS levels.


Asunto(s)
Autofagia/fisiología , Carcinoma Epitelial de Ovario/patología , Autorrenovación de las Células/fisiología , Células Madre Neoplásicas/patología , Línea Celular Tumoral , Femenino , Humanos , Esferoides Celulares
9.
Cell Cycle ; 17(13): 1591-1601, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29911914

RESUMEN

Psychological stress, which exerts detrimental effects on human reproduction, may compromise the meiotic competence of oocytes. Meiotic resumption, germinal vesicle breakdown (GVBD), is the first milestone to confer meiotic competence to oocytes. In the practice of assisted reproductive technology (ART), the timing for GVBD is associated with the rates of cleavage and blastocyst formation. However, whether chronic stress compromises oocyte competence by influencing GVBD and the underlying mechanisms are unclear. In the present study, a chronic restraint stress (CRS) mouse model was used to investigate the effects of stress on oocyte meiotic resumption, as well as the mechanisms. Following a 4-week chronic restraint stress in female mice, the percentage of abnormal bipolar spindles increased and indicated compromised oocyte competence in the CRS group. Furthermore, we identified a decreased percentage of GVBD and prolonged time of GVBD in the CRS mouse oocytes compared with the control group. CRS simultaneously reduced the expression of cyclin B1 (CCNB1), which represents a regulatory subunit of M-phase/mature promoting factor (MPF). However, MG132, an inhibitor of anaphase-promoting complex/cyclosome (APC/C), could rescue the prolonged time of GVBD and increase the expression level of CCNB1 of oocytes from the CRS mice. Collectively, our results demonstrated that stress disturbed meiotic resumption through APC/C-mediated CCNB1 degradation, thus providing a novel understanding for stress-related oocyte quality decline; moreover, it may provide a non-invasive approach to select high-quality gametes and novel targets for molecular therapy to treat stress-related female infertility.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclina B1/metabolismo , Meiosis , Oocitos/citología , Oocitos/metabolismo , Proteolisis , Restricción Física , Estrés Psicológico/patología , Animales , Corticosterona/sangre , Modelos Animales de Enfermedad , Femenino , Leupeptinas/farmacología , Mesotelina , Ratones Endogámicos BALB C , Oocitos/efectos de los fármacos , Proteolisis/efectos de los fármacos , Securina/metabolismo , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Estrés Psicológico/sangre , Aumento de Peso
10.
Stem Cell Res Ther ; 8(1): 270, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29179771

RESUMEN

BACKGROUND: Human amniotic epithelial cells (hAECs) are attractive candidates for regenerative medical therapy, with the potential to replace deficient cells and improve functional recovery after injury. Previous studies have demonstrated that transplantation of hAECs effectively alleviate chemotherapy-induced ovarian damage via inhibiting granulose cells apoptosis in animal models of premature ovarian failure/insufficiency (POF/POI). However, the underlying molecular mechanism accounting for hAECs-mediated ovarian function recovery is not fully understood. METHODS: To investigate whether hAECs-secreting cytokines act as molecular basis to attenuate chemotherapy-induced ovarian injury, hAECs or hAEC-conditioned medium (hAEC-CM) was injected into the unilateral ovary of POF/POI mouse. Follicle development was evaluated by H&E staining at 1, 2 months after hAECs or hAEC-CM treatment. In addition, we performed a cytokine array containing 507 human cytokines on hAECs-derived serum-free conditioned medium. Finally, we further investigated whether hAECs could affect chemotherapy-induced apoptosis in primary human granulosa-lutein (hGL) cells and the tube formation of human umbilical vein endothelial cells (hUVECs) via a co-culture system in vitro. RESULTS: We observed the existence of healthy and mature follicles in ovaries treated with hAECs or hAEC-CM, whereas seriously fibrosis and many atretic follicles were found in the contralateral untreated ovaries of the same mouse. To distinguish cytokines involved in the process of hAECs-restored ovarian function, hAEC-CM was analyzed with a human cytokines array. Results revealed that 109 cytokines in hAEC-CM might participate in a variety of biological processes including apoptosis, angiogenesis, cell cycle and immune response. In vitro experiments, hAECs significantly inhibited chemotherapy-induced apoptosis and activated TGF-ß/Smad signaling pathway within primary granulosa-lutein cells in paracrine manner. Furthermore, hAEC-CM was shown to promote angiogenesis in the injured ovaries and enhance the tube formation of human umbilical vein endothelial cells (hUVECs) in co-culture system. CONCLUSIONS: These findings demonstrated that paracrine might be a key pathway in the process of hAECs-mediating ovarian function recovery in animal models of premature ovarian failure/insufficiency (POF/POI).


Asunto(s)
Busulfano/antagonistas & inhibidores , Medios de Cultivo Condicionados/farmacología , Ciclofosfamida/antagonistas & inhibidores , Células Epiteliales/metabolismo , Comunicación Paracrina , Insuficiencia Ovárica Primaria/prevención & control , Amnios/citología , Amnios/metabolismo , Animales , Busulfano/efectos adversos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Ciclofosfamida/efectos adversos , Células Epiteliales/citología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/patología , Análisis por Matrices de Proteínas , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Int J Oncol ; 51(5): 1405-1414, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29048644

RESUMEN

It is reported that human amniotic epithelial cells (hAECs) endow intrinsic antitumor effects on certain kinds of cancer. This research was designed to evaluate whether hAECs endowed potential anticancer properties on epithelial ovarian cancer (EOC) cells in vivo and in vitro, which has not been reported before. In this study, we established a xenografted BALB/c nude mouse model by subcutaneously co-injecting ovarian cancer cell line, SK-OV-3, and hAECs for 28 days. In ex vivo experiments, CCK­8 cell viability assay, real-time PCR, cell counting assay, cell cycle analysis and immunohistochemistry (IHC) assay were used to detect the effects of hAEC­secreted factors on the proliferation and cell cycle progression of EOC cells. A cytokine array was conducted to detect anticancer-related cytokines released from hAECs. Human recombinant TGF­ß1 and TGF­ß1 antibody were used to treat EOC cells and analyzed whether TGF­ß1 contributed to the cell cycle arrest. Results from in vivo and ex vivo experiments showed that hAEC-secreted factors and rhTGF­ß1 decreased proliferation of EOC cells and induced G0/G1 cell cycle arrest in cancer cells, which could be partially reversed by excess TGF­ß1 antibody. These data indicate that hAECs endow potential anticancer properties on epithelial ovarian cancer in vivo and in vitro which is partially mediated by hAEC­secreted TGF­ß1-induced cell cycle arrest. This study suggests a potential application of hAEC­based therapy against epithelial ovarian cancer.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/terapia , Factor de Crecimiento Transformador beta1/genética , Líquido Amniótico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Epitelial de Ovario , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Rep ; 7(1): 9599, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851995

RESUMEN

Decline in semen quality has become a global public health concern. Psychological stress is common in the current modern society and is associated with semen decline. Increasing evidence demonstrated that melatonin has anti-apoptotic and antioxidant functions. Whether melatonin can ameliorate the damage in testes induced by psychological stress has never been investigated. Here, a mouse model of restraint stress demonstrated that melatonin normalized the sperm density decline, testicular cells apoptosis, and testicular oxidative stress in stressed male mice. Melatonin decreased reactive oxygen species (ROS) level, increased superoxide dismutase (SOD) and glutathione (GSH) activities, and downregulated inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α) activities in stressed mice testes. Furthermore, melatonin reduced the stress-induced activation of the NF-κB signaling pathway by decreasing the phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) and p65 nuclear translocation. In addition, melatonin upregulated the expression of anti-oxidant proteins including nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Meanwhile, in vitro studies also demonstrated melatonin could reduce oxidative apoptosis of testicular cells. Collectively, melatonin mitigated psychological stress-induced spermatogenic damage, which provides evidence for melatonin as a therapy against sperm impairment associated with psychological stress.


Asunto(s)
Apoptosis , Hemo-Oxigenasa 1/metabolismo , Melatonina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo , Transducción de Señal , Testículo/metabolismo , Animales , Peso Corporal , Corticosterona/sangre , Masculino , Melatonina/sangre , Melatonina/farmacología , Ratones , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos
13.
Sci Rep ; 6: 37019, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845405

RESUMEN

Epithelial ovarian cancer (EOC) is the most lethal tumor of all gynecologic tumors. There is no curative therapy for EOC thus far. The tumor-homing ability of adult mesenchymal stem cells (MSCs) provide the promising potential to use them as vehicles to transport therapeutic agents to the site of tumor. Meanwhile, studies have showed the intrinsic anti-tumor properties of MSCs against various kinds of cancer, including epithelial ovarian cancer. Human endometrial mesenchymal stem cells (EnSCs) derived from menstrual blood are a novel source for adult MSCs and exert restorative function in some diseases. Whether EnSCs endow innate anti-tumor properties on EOC cells has never been reported. By using tumor-bearing animal model and ex vivo experiments, we found that EnSCs attenuated tumor growth by inducing cell cycle arrest, promoting apoptosis, disturbing mitochondria membrane potential and decreasing pro-angiogenic ability in EOC cells in vitro and/or in vivo. Furthermore, EnSCs decreased AKT phosphorylation and promoted nuclear translocation of Forkhead box O-3a (FoxO3a) in EOC cells. Collectively, our findings elucidated the potential intrinsic anti-tumor properties of EnSCs on EOC cells in vivo and in vitro. This research provides a potential strategy for EnSC-based anti-cancer therapy against epithelial ovarian cancer.


Asunto(s)
Apoptosis/inmunología , Endometrio/inmunología , Células Madre Mesenquimatosas/inmunología , Neoplasias Ováricas/inmunología , Adulto , Animales , Línea Celular Tumoral , Endometrio/patología , Femenino , Humanos , Potencial de la Membrana Mitocondrial/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/inmunología , Mitocondrias/patología , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/patología
14.
Cell Cycle ; 15(10): 1376-85, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27074587

RESUMEN

RNA-Seq and gene set enrichment anylysis revealed that ovarian cancer associated fibroblasts (CAFs) are mitotically active compared with normal fibroblasts (NFs). Cellular senescence is observed in CAFs treated with H2O2 as shown by elevated SA-ß-gal activity and p21 (WAF1/Cip1) protein levels. Reactive oxygen species (ROS) production and p21 (WAF1/Cip1) elevation may account for H2O2-induced CAFs cell cycle arrest in S phase. Blockage of autophagy can increase ROS production in CAFs, leading to cell cycle arrest in S phase, cell proliferation inhibition and enhanced sensitivity to H2O2-induced cell death. ROS scavenger NAC can reduce ROS production and thus restore cell viability. Lactate dehydrogenase A (LDHA), monocarboxylic acid transporter 4 (MCT4) and superoxide dismutase 2 (SOD2) were up-regulated in CAFs compared with NFs. There was relatively high lactate content in CAFs than in NFs. Blockage of autophagy decreased LDHA, MCT4 and SOD2 protein levels in CAFs that might enhance ROS production. Blockage of autophagy can sensitize CAFs to chemotherapeutic drug cisplatin, implicating that autophagy might possess clinical utility as an attractive target for ovarian cancer treatment in the future.


Asunto(s)
Autofagia/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo/efectos de los fármacos , Acetilcisteína/farmacología , Actinas/metabolismo , Proteína 5 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Especies Reactivas de Oxígeno/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Superóxido Dismutasa/metabolismo
15.
Acta Biochim Biophys Sin (Shanghai) ; 48(3): 282-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26819076

RESUMEN

Chemoresistance blocks the efficient treatment of epithelial ovarian cancer, which is the most lethal of all gynecological cancers. Cancer stem cells are believed to be at least partially responsible for the development of chemoresistance. In this study, the effect of cisplatin (CDP) on the enrichment and proliferation of cancer stem-like cells (CSLCs) was investigated, and the underlying mechanisms of action were elucidated. An in vitro anchor-free system was employed to enrich CSLCs from the SKOV3 human epithelial ovarian cancer cell line. Our results showed that treatment with low concentrations of CDP resulted in better-enriched CSLCs, with higher proliferative activities. Low dose of CDP was found to induce the expression of chemokine (C-X-C motif) receptor 4 (CXCR4), which is an important stemness marker in cancer stem cells as well as a promising therapeutic target for ovarian cancer treatment. Results also showed that overexpressed CXCR4 generated chemoresistance. Based on these results, it may be concluded that, at low concentrations, CDP itself contributes to the development of drug resistance. This finding provides novel insight into the mechanisms underlying chemoresistance and has significant therapeutic implications for epithelial ovarian cancer treatment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Células Madre Neoplásicas/patología , Neoplasias Ováricas/patología , Receptores CXCR4/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos
16.
Cell Reprogram ; 18(1): 48-53, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26757253

RESUMEN

Culture conditions have been established to maintain the pluripotency of mouse naïve and primed embryonic stem cells (ESCs) using human amnion epithelial cells (hAECs) as the feeder layer. In this study, the ultrastructures of mouse primed ESCs grown on hAECs were analyzed by transmission electron microscopy. Consistent with mouse naïve ESCs, the undifferentiated mouse primed ESC line ESD-EpiSC [ESC-derived epiblast stem cells (EpiSCs)] revealed typical characteristics, including large nuclei, reticulated nucleoli, scanty cytoplasm, and low cytoplasm-to-nuclear ratios. Cells had prominent Golgi apparatus and well-developed endoplasmic reticulum. Adjacent cells were tightly in contact with dense junction desmosomes. However, in EpiSC colonies, cell contact was no longer close like naïve ESCs, and differentiated cells existed. The differentiated cells had small nucleoli with large cytoplasm, which represented primitive mesenchyme. Phagosomes or apoptotic cells were also common in the cytoplasm of differentiated cells, which suggests a differentiation potential. When exposed to leukemia inhibitory factor (LIF), ESD-EpiSCs could convert to naïve-like cells. We further analyzed the ultrastructure of converted EpiSCs (cESCs). As compared to ESD-EpiSCs, cESCs showed similar ultrastructural characteristics as naïve ESCs. These findings suggest that ultrastructure could be used to evaluate the pluripotency of ESCs.


Asunto(s)
Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/ultraestructura , Amnios/citología , Animales , Diferenciación Celular , Células Cultivadas , Células Nutrientes , Estratos Germinativos/citología , Estratos Germinativos/ultraestructura , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...