Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
5.
Bioengineered ; 13(1): 1346-1358, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34983308

RESUMEN

Long noncoding RNA muskelin 1 antisense RNA (MKLN1-AS) acted as an oncogenic regulator in hepatocellular carcinoma (HCC). This study was performed to investigate the functional mechanism of MKLN1-AS. MKLN1-AS, microRNA-22-3p (miR-22-3p) and ETS Proto-Oncogene 1 (ETS1) levels were examined using reverse transcription-quantitative polymerase-chain reaction. Protein expression was detected by Western blot. The target relation was analyzed by dual-luciferase reporter assay, RNA immunoprecipitation assay and RNA pull-down assay. Cell proliferation ability was determined through cell counting kit-8 assay, colony formation assay and ethylenediurea assay. Angiogenesis was examined by tube formation assay. Cell migration and invasion were assessed via transwell assay. In vivo research was conducted by xenograft tumor model in nude mice. MKLN1-AS was upregulated in HCC tissues and cells. ETS1 promoted the ETS1 expression by binding to the 582-596 sites. Silence of MKLN1-AS suppressed cell growth, angiogenesis, migration, and invasion. MKLN1-AS interacted with miR-22-3p in HCC cells. The function of MKLN1-AS downregulation was relieved by miR-22-3p inhibition in HCC cells. ETS1 was validated as a target of miR-22-3p, and MKLN1-AS upregulated the ETS1 expression by sponging miR-22-3p. Overexpression of miR-22-3p retarded HCC progression by downregulating the level of ETS1. Tumor growth in vivo was also enhanced by MKLN1-AS through the regulation of miR-22-3p/ETS1 axis. These data demonstrated that ETS1-mediated MKLN1-AS contributed to the malignant phenotypes of HCC cells via depending on the miR-22-3p/ETS1 regulatory axis.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , MicroARNs/genética , Proteína Proto-Oncogénica c-ets-1/genética , ARN Largo no Codificante/genética , Animales , Carcinoma Hepatocelular/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones , Trasplante de Neoplasias , Regulación hacia Arriba
6.
Cancer Manag Res ; 12: 233-246, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32021439

RESUMEN

OBJECTIVE: To explore the effect of miR106a on the growth of breast cancer xenografts and the sensitivity of chemotherapeutic agents. METHODS: Breast cancer cell lines (MDA-MB231 and MCF7) were transfected with an miR106 mimic and miR106a inhibitor. BALB/c female nude mice were selected to construct a transplanted-tumor model. Cisplatin treatment was performed 2 weeks after inoculation. After 5 weeks, tumor tissue was weighed. Apoptosis of tumor cells was detected by TUNEL staining. The expression of these proteins (Ki67, ß-catenin, cyclin D1 and cMyc) was detected by immunohistochemistry. The contents of P53, RUNX3, ABCG2, ß-catenin, BAX, and BCL2 mRNA were detected by qRT-PCR. RESULTS: The miR106a mimic (MM) group's tumor volume and weight were significantly bigger than those of the model group. miR106a mRNA content was higher than the blank control group, and ß-catenin and Ki67 protein were strongly positive. ß-catenin, BCL2, and ABCG2 mRNA content was were increased. P53, BAX, and RUNX3 mRNA content was decreased. The number of positive cells on TUNEL staining was significantly lower in the miR106a inhibitor (MI) group. After cisplatin treatment, inhibition of tumor growth was most obvious in the MI+DDP (cisplatin) group. Compared with the MM group, tumor growth in the MM+FH535 (Wnt-pathway inhibitor) group was significantly lower, and Wnt-pathway activity was decreased. CONCLUSION: Overexpression of miR106a can promote the growth of transplanted breast cancer and decrease the sensitivity of transplanted tumors to cisplatin. The mechanism may be related to abnormal activation of the Wnt-signaling pathway.

7.
DNA Cell Biol ; 38(2): 198-207, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30570350

RESUMEN

To explore the effect of miR-106a in breast cancer cell behavior and sensitivity to chemotherapeutic agents. Tumor tissue and adjacent normal tissue were derived from 40 breast cancer patients, and miR-106a expression was measured by reverse transcription-qPCR. Breast cancer cells, MDA-MB-231 and MCF-7, were treated with miRNA-106a mimic (MM) or miRNA-106a inhibitor (MI) and negative controls. Cell proliferation was measured by MTT assay. Clonogenicity was measured by colony-forming assay. Cell migration and invasion ability were measured by scratch test and transwell assay, respectively. Apoptosis was determined by flow cytometry, and chemosensitivity to cisplatin was measured by MTT assay. Finally, protein expression of p53, Bax, Bcl-2, RUNX3, and ABCG2 was quantified by western blot. miR-106a expression was significantly upregulated in human breast cancer tissue relative to adjacent normal tissue. Upregulation of miR-106a enhanced breast cancer cell proliferation, colony-forming capacity, migration, and invasion of cultured breast cancer cells. Additionally, miR-106a overexpression significantly decreased breast cancer cell apoptosis and sensitivity to cisplatin. Finally, we showed miR-106a overexpression upregulated the levels of Bcl-2 and ABCG2, and downregulated the expression of P53, Bax, and RUNX3. miR-106a promotes breast cancer cell proliferation and invasion through upregulation of Bcl-2, ABCG2, and P53, and downregulation of Bax and RUNX3.


Asunto(s)
Apoptosis/genética , Neoplasias de la Mama/genética , Movimiento Celular/genética , Proliferación Celular/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cisplatino/farmacología , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células MCF-7 , Invasividad Neoplásica/patología , Regulación hacia Arriba/genética
8.
Pak J Pharm Sci ; 31(6(Special)): 2869-2872, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30630801

RESUMEN

To investigate the efficaciousness of breast-conserving therapy in connection with neoadjuvant chemotherapy on breast cancer. 68 patients, who were confirmed going down with breast cancer and hospitalized from June 2015 and June 2017, were sampled and divided into two groups using the random digit table, i.e. the observation group (n=34) and the control group (n=34). Patients in the observation group experienced breast-conserving therapy integrated with neoadjuvant chemotherapy, but those in the control group received the radical resection of breast cancer. Patients' condition in surgery, incidence of post-surgery complications as well as patient survivals were compared and coded. In the observation group, surgical duration, intraoperative bleeding amount, length of stay in hospital and incidence rate of post-surgery complications were all lower than the patients with the similar conditions in the control group with evident distinctions in statistics (p<0.05). In the observation group, survival ratios of one-to-five-year living patients were evidently higher than those in the control group. The distinctions owned evident significance in calculations (p<0.05). In comparison of the recurrence ratio of disease and the rate of distant metastasis between the observation group (5.88% and 8.82%) and the control group (11.76% and 8.82%), differences had no statistical significance (p>0.05). Before treatment, compared with the score of life quality in the two groups, no evident distinction in statistical exists (p>0.05), however, after that, the life quality in the observation group evidently outweighs the quality in the control group, which shows the distinctions in statistics (p<0.05). Breast-conserving therapy in combination with neoadjuvant chemotherapy shows promising clinical value in ameliorating the life quality, decreasing the mortality rate and the incidence of adverse reaction, which is expected to be applied in clinical practices as a kind of safe and effective method.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/cirugía , Docetaxel/uso terapéutico , Epirrubicina/uso terapéutico , Mastectomía Segmentaria/métodos , Terapia Neoadyuvante/métodos , Adulto , Anciano , Neoplasias de la Mama/secundario , China/epidemiología , Femenino , Humanos , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Calidad de Vida , Recurrencia , Análisis de Supervivencia , Resultado del Tratamiento
9.
Int Immunopharmacol ; 52: 163-167, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28918288

RESUMEN

Breast cancer is a major cause of cancer-related death in women. Antitumor T cell responses play critical therapeutic roles, including direct cytotoxicity mediated by CD8+ T cells and immunomodulatory roles mediated by CD4+ T cells. The IL-9-expressing Th9 cells are recently found to present antitumor immunity in melanoma and lung adenocarcinoma. In this study, we found that IL-9 expression in the serum and in circulating CD4+ T cells were significantly upregulated in breast cancer patients compared to healthy controls. The IL-9-expressing Th9 cells were enriched in the CCR4-CCR6-CXCR3- subset. Upon TCR stimulation, this subset also presented potent IL-10 and IL-21 expression in addition to IL-9 expression. CCR4-CCR6-CXCR3- CD4+ T cells also assisted in the killing of autologous tumor cells by CD8+ T cells, but did not initiate cytotoxicity by themselves. This enhancement in CD8+ T cell-mediated cytotoxicity was dependent on IL-9 as well as on IL-21. Interestingly, the tumor-infiltrating Th9 cells presented comparable IL-9, reduced IL-10, and elevated IL-21 expression compared with their counterparts in the peripheral blood. Together, these results demonstrated that IL-9-expressing Th9 cells were upregulated in breast cancer patients and potentially possessed antitumor roles by enhancing CD8+ T cell-mediated cytotoxicity.


Asunto(s)
Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-9/metabolismo , Interleucinas/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Adulto , Anciano , Antígenos CD4/metabolismo , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular , Citotoxicidad Inmunológica , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-10/metabolismo , Interleucina-9/genética , Interleucinas/genética , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/metabolismo
10.
Biomed Pharmacother ; 88: 443-448, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28122310

RESUMEN

Emerging evidences indicate that dysregulated microRNAs are implicated in the process of tumorigenesis and progression. The miRNA-125b (miR-125b) is downregulated and identified as tumor supressor in various cancers including thyroid cancer. However, the role and mechanism of miR-125b in anaplastic thyroid cancer (ATC) migration and invasion remain unknown. In the present study, the expression levels of miR-125b were downregulated and the expression levels of phosphoinositide 3-kinase catalytic subunit delta (PIK3CD) were upregulated in ATC tissues and cell lines. Moreover, miR-125b expression was negatively related to PIK3CD expression in ATC tissues. A computational search and luciferase assay identified PIK3CD as a direct target of miR-125b in ATC and PIK3CD expression was downregulated by miR-125b in ATC cells. In terms of function, miR-125b repressed migration and invasion of ATC cells, whereas PIK3CD overexpression reversed this effect. Furthermore, we showed that exogenous miR-125b decreased the PI3K, phospho-Akt, and phospho-mTOR expression in ATC cells. In conclusion, these results indicated that miR-125b suppressed ATC cell migration and invasion by targeting PIK3CD expression, and suggested novel potential therapeutic targets for the treatment of ATC.


Asunto(s)
Movimiento Celular/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , MicroARNs/metabolismo , Carcinoma Anaplásico de Tiroides/genética , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Invasividad Neoplásica , Transducción de Señal/genética
11.
Int J Clin Exp Pathol ; 8(1): 368-73, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25755724

RESUMEN

OBJECTIVES: To investigate the crucial role of miR-26a in breast cancer and to validate whether miR-26a could regulate proliferation of breast cancer cells by targeting high mobility group AT-hook 1 (HMGA1). METHODS: Reverse transcription-polymerase chain reaction (RT-PCR) was used to quantify the expression levels of miR-26a in breast cancer and adjacent non-cancerous breast tissues. MTT, cell migration and invasion assay were carried out to characterize the miR-26a function. Finally, to validate the target gene of miR-26a, luciferase reporter assay was employed, followed by RT-PCR and Western blot confirmation. RESULTS: Compared with normal tissues, a significant down-regulation of miR-26a expression was observed in breast cancer tissues (P=0.002). miR-26a suppresses MDA-MB-231 and Mcf-7 breast cancer cell lines proliferation and motility. The luciferase activity was significantly decreased after co-transfection with psiCHECK-2/HMGA1 3'-UTR and miR-26a mimics in comparison with control cells, and qRT-PCR and Western blotting analysis found that HMGA1 expression at the mRNA and protein levels decreased in the miR-26a mimic-treatment group relative to NC. MTT assay showed that down regulation of HMGA1 by siRNA could significantly enhance the tumor-suppressive effect of miR-26a (P < 0.05). CONCLUSIONS: The results of the present study indicate that miR-26a may be associated with human breast carcinogenesis, which inhibits tumor cell proliferation by targeting HMGA1.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/genética , Proteínas HMGA/biosíntesis , MicroARNs/genética , Western Blotting , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Exp Ther Med ; 2(5): 951-955, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22977604

RESUMEN

microRNAs (miRNAs) have been proven to play key regulatory roles in hepatocarcinogenesis. In the present study, the possible role of microRNA-450a (miR-450a) in hepatocarcinogenesis was investigated. Our study revealed that miR-450a was significantly down-regulated in hepatocellular carcinoma (HCC) tissues compared with that in normal liver (NL) and para-tumorous (PT) tissues, and miR-450a expression in HepG2 cells was significantly lower than that in L02 cells. Both the mRNA and protein levels of the miR-450a potential target gene, DNA methyltransferase 3a (DNMT3a), were obviously higher in HCC compared with levels in the NL and PT tissues. We further identified DNMT3a as the direct target gene for miR-450a, and ectopic miR-450a expression in HepG2 cells caused the down-regulation of DNMT3a and an inhibition of cell proliferation. Taken together, these findings suggest that miR-450a plays an important regulatory role in hepatocarcinogenesis through inhibition of DNMT3a expression, and miR-450a may be a potential target for the treatment of HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...