Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Ann Diagn Pathol ; 56: 151861, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34953233

RESUMEN

OBJECTIVE: The study aimed to develop a prediction model combining clinical and histological features to predict recurrence in patients with stage I-II endometrial cancer (EC) after surgical treatment. METHODS: A total of 746 stage I-II EC patients who had received primary surgical treatment at Taizhou People's Hospital between 2014 and 2018 were included and randomly divided as a Training cohort (n = 520) and a Validation cohort (n = 226) at a 7:3 ratio. Clinical features including age, body mass index, comorbidities, lymphadenectomy, and adjuvant treatment, and histological features including histologic type, myometrial invasion, cervical stromal invasion, and expression levels of Ki67, estrogen receptor (ER), progesterone receptor (PR), whey acidic protein 4-disulphide core domain 2 (WFDC2), and p53 were used to develop a prediction model for EC recurrence in the Training cohort using a multivariable Cox regression model. Model discrimination and calibration were further evaluated in the Validation cohort. RESULTS: EC recurrence was observed in 60 (11.54%) patients in the Training cohort with a median length of follow-up of 39 months. Age, adjuvant treatment, histologic type, cervical stromal invasion, and expression levels of Ki67, ER, PR, and WFDC2 were factors significantly associated with EC recurrence based on univariable Cox regression analysis. After a model selection by AIC in a stepwise algorithm, the final model incorporated the above predictors showed a C-index of 0.85 and fair calibration in the Training cohort. In the Validation cohort, the model still showed good discrimination power (C-index 0.80) but moderate calibration. CONCLUSIONS: The developed prediction model combining clinical and histological features can help to predict the EC recurrence in patients with stage I-II EC after surgical treatment.


Asunto(s)
Carcinoma Endometrioide/patología , Neoplasias Endometriales/patología , Recurrencia Local de Neoplasia/diagnóstico , Adulto , Biomarcadores de Tumor/metabolismo , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/cirugía , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/cirugía , Femenino , Humanos , Persona de Mediana Edad , Modelos Teóricos , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
2.
Drug Dev Res ; 83(6): 1383-1393, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35808943

RESUMEN

Ovarian cancer is one of the three major gynecological malignancies. It has been reported that Icariside II was able to block the occurrence and development of ovarian cancer. However, the detailed mechanism by which Icariside II regulates the development of ovarian cancer is widely unknown. EdU staining and transwell assays were applied to detect the proliferation, migration, and invasion of ovarian cancer cells. Next, the relationship between miR-144-3p and IGF2R was verified by the dual-luciferase reporter assay. Moreover, in vivo animal model was constructed to verify the effect of Icariside II on the development of ovarian cancer. Icariside II notably inhibited the proliferation, migration, and invasion and induced the apoptosis of ovarian cancer cells. Additionally, Icariside II markedly increased the level of miR-144-3p in ovarian cancer cells. Moreover, IGF2R was targeted by miR-144-3p directly. Icariside II significantly decreased the expression of IGF2R and the phosphorylation level of AKT and mTOR in ovarian cancer cells, which were partially reversed by miR-144-3p inhibitor. Meanwhile, Icariside II remarkably promoted the autophagy of ovarian cancer cells, as confirmed by the increased expression of Beclin-1 and ATG-5 and decreased expression of p62; however, co-treatment with miR-144-3p inhibitor notably decreased autophagy. Furthermore, the result of animal study suggested Icariside II notably inhibited ovarian tumor growth as well. Collectively, Icariside II could suppress the tumorigenesis and development of ovarian cancer by promoting autophagy via miR-144-3p/IGF2R axis. These results may be beneficial for future studies on the use of Icariside II to treat ovarian cancer.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Animales , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Flavonoides , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética
3.
J Obstet Gynaecol Res ; 47(1): 452-455, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33197962

RESUMEN

Teratomas are one of the most common germ cell tumors, and they usually occur in ovaries. Extragonadal teratomas are rare, especially immature ones. Only several cases of primary teratomas of the uterus have been reported since 1929. Here, the case of an 11-year-old patient who had a 6-month history of sustained abnormal vaginal discharge is presented. Transabdominal ultrasonography revealed a solid mass in her uterus, resulting in the patient undergoing surgery. Examination of PET-CT scans revealed a mass in the right ovary of the patient 20 days after surgery. The patient underwent a second surgery followed by chemotherapy. This is the youngest case among reported patients of primary immature uterine teratoma, and this patient showed no evidence of recurrence during 2 years of follow-up.


Asunto(s)
Neoplasias Ováricas , Teratoma , Niño , Femenino , Humanos , Recurrencia Local de Neoplasia , Neoplasias Ováricas/cirugía , Ovario , Tomografía Computarizada por Tomografía de Emisión de Positrones , Teratoma/diagnóstico por imagen , Teratoma/cirugía , Útero/diagnóstico por imagen , Útero/cirugía
4.
Placenta ; 131: 75-81, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36521318

RESUMEN

INTRODUCTION: Human umbilical cord mesenchymal stem cells (UCMSCs) play an important role in repairing the damaged endometrium of intrauterine adhesion (IUA). Meanwhile, exosomes released by UCMSCs can mediate intercellular communication by delivering miRNAs. It has been shown that miR-543 level was reduced in IUA tissues. However, the role of miR-543 in the progression of IUA remains largely unknown. Therefore, we investigated the role of UCMSCs-derived exosomal miR-543 in IUA. METHODS: In this study, human endometrial epithelial cells (hEECs) were treated with TGF-ß1 for mimicking endometrial fibrosis in vitro. In addition, the IUA-like mouse model in vivo was established by a dual damage method of curettage and LPS infection. RESULTS: The level of miR-543 was markedly reduced in hEECs exposed to TGF-ß1 and in endometrium tissues of IUA mice. Additionally, miR-543 could be transferred from UCMSCs to hEECs via exosomes. Meanwhile, exosomal miR-543-derived from UCMSCs significantly reduced the expressions of N-cadherin, α-SMA, fibronectin 1 and elevated the expression of E-cadherin in TGF-ß1-treated hEECs. Furthermore, UCMSCs-derived exosomal miR-543 attenuated IUA-induced endometrial fibrosis in vivo, as shown by the decreased N-cadherin, α-SMA and fibronectin 1 protein expressions. DISCUSSION: Collectively, UCMSCs-derived exosomal miR-543 was able to prevent endometrial fibrosis both in vitro and in vivo via downregulating N-cadherin. These results may provide an insight into the clinical treatment for IUA.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , MicroARNs , Enfermedades Uterinas , Femenino , Humanos , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Fibronectinas/metabolismo , Enfermedades Uterinas/terapia , Endometrio/metabolismo , MicroARNs/metabolismo , Adherencias Tisulares/metabolismo , Adherencias Tisulares/patología , Adherencias Tisulares/terapia , Exosomas/metabolismo , Cordón Umbilical , Cadherinas/genética , Cadherinas/metabolismo
5.
Genes Genomics ; 44(5): 527-537, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35094286

RESUMEN

BACKGROUND: Endometriosis (EM) is a gynecological disease that poses severe health risks to women, although its pathogenesis has yet to be fully elucidated. It has been shown that long non-coding RNAs (lncRNAs) are closely associated with EM initiation and have a role in the development of this disease. Previous studies exploring the expression of the lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) have shown that this lncRNA functions as a tumor promoter in endometrial cancer. However, its exact mechanism of action in EM remains unclear. OBJECTIVE: This report was designed to illustrate the potential molecular mechanisms of lncRNA NEAT1 on EM. METHODS: Endometrial tissues were extracted from EM model rats and patients with EM. Hematoxylin and eosin staining was applied to detect the morphological changes that occurred in rats after construction of the model. Endometrial stromal cells (ESCs) were extracted from either ectopic endometrium (EC) or eutopic endometrium (EU) tissues from patients with EM. LncRNA NEAT1 and miR-124-3p expression in EM tissues and cells were subsequently evaluated by reverse transcription-quantitative (RT-q)PCR analysis. MTT assay, flow cytometric analysis, western blot assay and Transwell assay were then employed to examine the effect of NEAT1 and miR-124-3p on EC-ESC proliferation, apoptosis, migration and invasion, respectively. The targeted relationship between lncRNA NEAT1 and miR-124-3p was subsequently confirmed by dual-luciferase and co-transfection assays. RESULTS: MiR-124-3p was identified as a target of NEAT1, and could be negatively regulated by NEAT1 in EC-ESCs. The expression level of NEAT1 was evidently increased, whereas that of miR-124-3p was decreased, in the EM in vivo model, EM tissues and EC-ESCs from patients with EM. The loss-of-function assays further established that silencing of NEAT1 could inhibit EC-ESC proliferation, migration, and invasion, but it led to the promotion of apoptosis via targeting miR-124-3p. CONCLUSIONS: NEAT1 is significantly upregulated in EM, promoting malignant behavior in EM through targeting miR-124-3p expression.


Asunto(s)
Endometriosis , MicroARNs , ARN Largo no Codificante , Animales , Apoptosis/genética , Endometriosis/genética , Endometrio/metabolismo , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratas
6.
Cancer Manag Res ; 14: 327-337, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35115831

RESUMEN

BACKGROUND: Ovarian cancer is a life-threatening disease with a high mortality rate in women. Our previous work presented that long non-coding RNA (lncRNA) activated by transforming growth factor beta (TGF-ß) (lncRNA ATB) played a role of oncogene in ovarian cancer. However, whether exosomal lncRNA ATB from ovarian cancer cells could regulate the tumorigenesis of ovarian cancer remains unclear. METHODS: RT-qPCR assay was performed to evaluate the level of lncRNA ATB in cancer cells (SKOV3 and A2780). In addition, ovarian cancer cells-secreted exosomes were collected with ultracentrifugation. CCK8 assay was performed to detect the viability of ovarian cells and HUVECs. Meanwhile, Western blot was performed to detect the expression of mechanism related protein and tube formation assay was used to observe the angiogenesis of HUVECs. Finally, xenograft mice model was used to verify the role of ovarian cancer cell-derived exosomes in vivo. RESULTS: Ovarian cancer cells-derived exosomes promoted the viability, angiogenesis and migration of HUVECs; however, knockdown of lncRNA ATB in HUVECs reversed these phenomena. In addition, exosomal lncRNA ATB promoted the tumorigenesis of ovarian cancer via regulating miR-204-3p/TGFßR2 axis. Furthermore, ovarian cancer cells-secreted exosomal lncRNA ATB increased tumor growth in vivo. CONCLUSION: Exosomal lncRNA ATB derived from ovarian cancer cells could improve tumor microenvironment via regulating miR-204-3p/TGFßR2 axis. Thus, this study might provide new knowledge for the treatment of ovarian cancer.

7.
J Ovarian Res ; 14(1): 109, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34454554

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) were reported to be aberrantly expressed and related to the pathogenesis of ovarian cancer. However, the role and regulatory mechanism of MSC-AS1 in ovarian cancer has yet to be fully elucidated. METHODS: Expression of lncRNA MSC-AS1 (MSC-AS1) and microRNA-425-5p (miR-425-5p) in the ovarian cancer tissue samples and cell lines was examined by quantitative real-time polymerase chain reaction (qRT-PCR). The functions of MSC-AS1 on ovarian cancer cell proliferation, cell cycle and apoptosis were determined using MTT, colony formation and flow cytometry analyses. The protein expression levels were evaluated using western blot assay. The targeting relationship MSC-AS1 and miR-425-5p was verified via dual-luciferase reporter assay. RESULTS: MSC-AS1 expression level was lowly expressed, while miR-425-5p level was highly in ovarian cancer tissues and cells. Elevation of MSC-AS1 has the ability to significantly inhibit cell proliferation and facilitate cell apoptosis in SKOV3 and A2780 cells. Moreover, MSC-AS1 targeted and negatively modulated miR-425-5p. MiR-425-5p up-regulation has been proved to partially reverse the tumor suppressive function of MSC-AS1 overexpression CONCLUSION: MSC-AS1 sponged miR-425-5p to inhibit the ovarian cancer progression. These findings may provide a promising therapeutic target for the treatment of ovarian cancer.


Asunto(s)
MicroARNs/metabolismo , Neoplasias Ováricas/metabolismo , ARN Largo no Codificante/metabolismo , Apoptosis/fisiología , Proliferación Celular/fisiología , Progresión de la Enfermedad , Femenino , Humanos , MicroARNs/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Largo no Codificante/genética
8.
Int J Biol Macromol ; 158: 1135-1140, 2020 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-32360469

RESUMEN

Self-bonding natural fiber materials (SNFMs) were prepared at different initial moisture contents (IMCs) through a molding pressing process. The self-bonding mechanism of the SNFMs was deduced from the chemical and structural changes of lignin and their mechanical strengths. The structural transformations of milled wood lignin (MWL) in the SNFMs were investigated by two-dimensional heteronuclear single quantum coherence, quantitative 31P-nuclear magnetic resonance spectra, gelpermeation chromatography, and thermogravimetric analysis. As IMC increased from 0% to 80%, the tensile strength increased from 23.0 to 70.0 MPa and the density increased from 0.99 to 1.05 g/cm3. IMC affected the distribution and abundance of the typical lignin linkages (ß-O-4', ß-ß, and ß-5') and the S-OH/G-OH ratios of lignin. Moreover, as IMC increased, the aliphatic hydroxyl groups proportionally decreased, while the condensed phenolic and non-condensed phenolic hydroxyl groups increased, the molecular weight of MWL became larger, and the thermal stability of lignin improved. These findings indicate the simultaneous occurrence of depolymerization and condensation reactions of lignin. The condensation reaction dominated, improving the mechanical strength of the material. Our results explain (at least partly) the self-bonding mechanism of SNFMs.

9.
Exp Ther Med ; 19(2): 965-971, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32010258

RESUMEN

This study aimed to elucidate the role of long non-coding RNA activated by transforming growth factor-ß (lncRNA-ATB) in ovarian cancer and its underlying mechanisms of action. Expression levels of lncRNA-ATB in ovarian cancer cell line SKOV3 and in a healthy human ovarian cell line were compared using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The results indicated that lncRNA-ATB was expressed at significantly higher levels in SKOV3 cells compared with the healthy cell line. After downregulation of lncRNA-ATB expression in SKOV3 cells using lncRNA-ATB-short hairpin RNA, cell proliferation, apoptosis, invasion and migration were assessed using Cell counting kit-8, Live Dead staining, Transwell assay and wound healing assay, respectively. RT-qPCR and western blotting were used to quantify the expression of signal transducer and activator of transcription 3 (STAT3), phosphorylated (p)-STAT3, and the additional epithelial to mesenchymal transition (EMT)-related proteins E-cadherin and vimentin in SKOV3 cells. LncRNA-ATB downregulation significantly reduced SKOV3 cell proliferation, invasion and migration, promoted apoptosis, decreased the expression of p-STAT3 and vimentin, and increased E-cadherin expression. Taken together, these results suggest that lncRNA-ATB downregulation can inhibit ovarian cancer cell proliferation, invasion and migration, and promote cell apoptosis. Lnc-RNA-ATB may therefore be a new target for ovarian cancer treatment.

10.
Oncol Lett ; 18(1): 402-410, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31289511

RESUMEN

Decreased expression of microRNA (miR)-148a is associated with poor prognosis in ovarian cancer. The aim of the present study was to investigate the impact of miR-148a on tumor cell viability and invasion via targeting forkhead box protein O3 (FOXO3). Expression of miR-148a was detected in paired tumor and adjacent normal tissues. OVCAR3 cells were transfected with miR-148a mimic and inhibitor. Cell viability, apoptosis and invasion were determined. A luciferase reporter assay was used to study the association between miR-148a and FOXO3. In addition, the influence of miR-148a on tumor cell growth was investigated by performing xenograft assays in nude mice. RT-qPCR showed that miR-148a was downregulated in ovarian cancer tissues. Overexpression of miR-148a in OVCAR3 cells inhibited cell viability, suppressed invasion and promoted cellular apoptosis. The dual-luciferase assay indicated that miR-148a directly regulated the expression of FOXO3, a transcription factor of caspase-3. Western blotting confirmed that the expression of caspase-3 was regulated by the modulation of miR-148a expression. In vivo assays revealed that miR-148a overexpression inhibited the growth of OVCAR3 ×enograft tumors in nude mice. miR-148a is a tumor suppressor in ovarian cancer OVCAR3 cells and in nude mice. The suppressive effect is due to inhibiting cell viability and invasion as well as promoting apoptosis. These results may provide theoretical basis for targeting miR-148a in the treatment of ovarian cancer.

11.
Clin Chim Acta ; 489: 225-232, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30107158

RESUMEN

BACKGROUND: This study aimed to determine the in vitro and in vivo properties of sixteen frequently used endometrial cancer (EC) cell lines, including the cell proliferation rate, morphology, hormone receptor expression patterns, PTEN, hMLH1 expression, p53 mutation, karyotype, and tumorigenicity in mouse xenograt model. METHODS: Twelve type I (AN3, ECC-1, EN, EN-1, EN-11, HEC-1A, HEC1B, Ishikawa, KLE, MFE-280, MFE-296, MFE-319) and four type II (ARK1, ARK2, HEC-155/180, SPEC-2) endometrial cancer cell lines were studied. Cell proliferation and morphology were determined using cell growth curves and light microscopy, respectively. Real-time PCR was performed to measure the mRNA levels of target genes. Denaturing High Performance Chromatography (DHPLC) screening and PCR/sequencing were performed to identify p53 mutations. G-banding was applied for karyotyping. Tumorigenicity was evaluated using mouse xenograft. RESULTS: The population doubling time of the cell lines ranged between 19 and 41 h. Ishikawa, ECC-1, and MFE-280 have high while AN3 and EN1 have low expression of ER-α and ER-ß. Expression of total PR and PR-B uniformly decreased in all type II cell lines and several type I cell lines (AN3, HEC-1A, HEC1B, KLE, EN-1). Regression analyses revealed significant correlations between PR-B and total PR (p < .001), between isoforms ER-α and ER-ß (p < .001), and between total PR and ER (p < .001), mRNA levels in type I cell lines. p53 mutations were detected in exons 5-8 of seven out of twelve type I and one out of four type II cell lines. PTEN expression was more uniformly suppressed in type II than type I cells, while hMLH1 did not show this pattern. All the five cell lines tested contained severe karyotype abnormalities. Mouse xenograft results indicated that HEC-1A, HEC-1B and EN-1 type I as well as ARK1 and ARK2 type II cell lines had potent tumorigenic activities. Low PR-B and ER-α expression in type I cell lines were associated with high tumorigenic activity. CONCLUSIONS: This study provides resource information on EC cell lines commonly used in laboratories, which could be used for choosing cell lines suitable for specific research purposes. The results of karyotype analysis and p53 mutation together with hormone receptor expression pattern and morphology comparison strongly suggested an independent nature of these cell lines, excluding the possibility of cross-contamination between cell lines. Additionally, this information suggests potential directions for future studies on the pathogenic mechanisms of endometrial cancer.


Asunto(s)
Carcinogénesis , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Cariotipo , Ratones , Homólogo 1 de la Proteína MutL/genética , Homólogo 1 de la Proteína MutL/metabolismo , Mutación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Proteína p53 Supresora de Tumor/genética
12.
J Zhejiang Univ Sci B ; 16(6): 549-59, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26055917

RESUMEN

Proliferation suppression and apoptosis are the prominent characteristics induced by heat stress (HS) in cells, whereas the effects of HS on cell growth (mass accumulation) are unknown. In this study, Lantang swine (an indigenous breed of China) skeletal muscle satellite cells (SCs) were pre-cultured at 37 °C for 24 h. The HS group was subjected to HS at 41 °C, while the control group was maintained at 37 °C. Heat shock protein 70 (HSP70) expression and SC size are significantly increased (P<0.05) by HS, but cell proliferation is suppressed (P<0.05) and apoptosis is induced (P<0.05). HS led to a lower percentage of SCs in the G0/G1 phase (P<0.05) together with a higher percentage of SCs in the S phase (P<0.05). However, the percentage of SCs in the G2/M phase was decreased (P<0.05) at 48 h but then increased (P<0.05) at 72 h with HS. In addition, the phosphorylation ratios of protein kinase b (Akt), ribosomal protein S6 kinase (S6K), and ribosomal protein S6 were increased (P<0.05) by HS. Nevertheless, the phosphorylation ratios of the 4E binding protein 1 and the eukaryotic initiation factor-4E were indistinguishable (P>0.05) from those of the control group. The phosphorylation ratio of the mammalian target of rapamycin (mTOR) (Ser(2448)) increased (P<0.05) within 48 h, and apparent differences were abrogated at 72 h (P>0.05). Moreover, cleaved caspase-3 expression was increased at 72 h (P<0.05). These findings indicate that HS induces apoptosis and disrupts cell cycle distribution to decrease the number of cells. Additionally, HS can promote SC growth via an activated Akt/mTOR/S6K signaling pathway.


Asunto(s)
Apoptosis/fisiología , Puntos de Control del Ciclo Celular/fisiología , Proliferación Celular/fisiología , Respuesta al Choque Térmico/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Aclimatación/fisiología , Animales , Aumento de la Célula , Células Cultivadas , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA