Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Acta Biochim Biophys Sin (Shanghai) ; 55(4): 633-648, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36916297

RESUMO

Ginsenoside Rh2, which is extracted from ginseng, exerts antitumor activity. Recent studies suggest that Rh2 may suppress the growth of colon cancer (CC) in vitro. However, the underlying mechanism remains unclear. In this study, we identified the relative levels of miR-150-3p in CC tissues and cells by a comprehensive strategy of data mining, computational biology, and real-time reverse transcription PCR (qRT-PCR) experiments. The regulatory effects of miR-150-3p/SRCIN1 on the proliferative and invasive abilities of CC cells are evaluated by CCK-8, EdU, wound healing, and transwell assays. Cell cycle- and apoptosis-related protein levels are assessed by western blot analysis. An in vivo tumor formation assay was conducted to explore the effects of miR-150-3p on tumor growth. Furthermore, bioinformatics and dual luciferase reporter assays are applied to determine the functional binding of miRNA to mRNA of the target gene. Finally, the relationship between Rh2 and miR-150-3p was further verified in SW620 and HCT-116 cells. miR-150-3p is downregulated in CC tissues and cell lines. Functional assays indicate that the upregulation of miR-150-3p inhibits tumor growth both in vivo and in vitro. In addition, SRCIN1 is upregulated in CC and predicts a poor prognosis, and it is the direct target for miR-150-3p. Moreover, the miR-150-3p mimic decreases Topflash/Fopflash-dependent luciferase activity, resulting in the inhibition of Wnt pathway activity. Rh2 can suppress the growth of CC by increasing miR-150-3p expression. Rh2 alleviates the accelerating effect on Wnt pathway activity, cell proliferation/migration, and colony formation caused by miR-150-3p inhibition. Rh2 inhibits the miR-150-3p/SRCIN1/Wnt axis to suppress colon cancer growth.


Assuntos
Neoplasias do Colo , Ginsenosídeos , MicroRNAs , Humanos , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Ginsenosídeos/farmacologia , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular
2.
Cell Mol Life Sci ; 78(4): 1355-1367, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33079227

RESUMO

The p140Cap adaptor protein is a scaffold molecule encoded by the SRCIN1 gene, which is physiologically expressed in several epithelial tissues and in the neurons. However, p140Cap is also strongly expressed in a significant subset of cancers including breast cancer and neuroblastoma. Notably, cancer patients with high p140Cap expression in their primary tumors have a lower probability of developing a distant event and ERBB2-positive breast cancer sufferers show better survival. In neuroblastoma patients, SRCIN1 mRNA levels represent an independent risk factor, which is inversely correlated to disease aggressiveness. Consistent with clinical data, SRCIN1 gain or loss of function mouse models demonstrated that p140Cap may affect tumor growth and metastasis formation by controlling the signaling pathways involved in tumorigenesis and metastatic features. This study reviews data showing the relevance of SRCIN1/p140Cap in cancer patients, the impact of SRCIN1 status on p140Cap expression, the specific mechanisms through which p140Cap can limit cancer progression, the molecular functions regulated by p140Cap, along with the p140Cap interactome, to unveil its key role for patient stratification in clinics.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Neoplasias da Mama/genética , Carcinogênese/genética , Neuroblastoma/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Neoplasias da Mama/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Metástase Neoplásica , Neuroblastoma/patologia , Receptor ErbB-2/genética , Transdução de Sinais/genética
3.
J Neurosci ; 39(29): 5634-5646, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31092585

RESUMO

Addictive behaviors, including relapse, are thought to depend in part on long-lasting drug-induced adaptations in dendritic spine signaling and morphology in the nucleus accumbens (NAc). While the influence of activity-dependent actin remodeling in these phenomena has been studied extensively, the role of microtubules and associated proteins remains poorly understood. We report that pharmacological inhibition of microtubule polymerization in the NAc inhibited locomotor sensitization to cocaine and contextual reward learning. We then investigated the roles of microtubule end-binding protein 3 (EB3) and SRC kinase in the neuronal and behavioral responses to volitionally administered cocaine. In synaptoneurosomal fractions from the NAc of self-administering male rats, the phosphorylation of SRC at an activating site was induced after 1 d of withdrawal, while EB3 levels were increased only after 30 d of withdrawal. Blocking SRC phosphorylation during early withdrawal by virally overexpressing SRCIN1, a negative regulator of SRC activity known to interact with EB3, abolished the incubation of cocaine craving in both male and female rats. Conversely, mimicking the EB3 increase observed after prolonged withdrawal increased the motivation to consume cocaine in male rats. In mice, the overexpression of either EB3 or SRCIN1 increased dendritic spine density and altered the spine morphology of NAc medium spiny neurons. Finally, a cocaine challenge after prolonged withdrawal recapitulated most of the synaptic protein expression profiles observed at early withdrawal. These findings suggest that microtubule-associated signaling proteins such as EB3 cooperate with actin remodeling pathways, notably SRC kinase activity, to establish and maintain long-lasting cellular and behavioral alterations following cocaine self-administration.SIGNIFICANCE STATEMENT Drug-induced morphological restructuring of dendritic spines of nucleus accumbens neurons is thought to be one of the cellular substrates of long-lasting drug-associated memories. The molecular basis of these persistent changes has remained incompletely understood. Here we implicate for the first time microtubule function in this process, together with key players such as microtubule-bound protein EB3 and synaptic SRC phosphorylation. We propose that microtubule and actin remodeling cooperate during withdrawal to maintain the plastic structural changes initially established by cocaine self-administration. This work opens new translational avenues for further characterization of microtubule-associated regulatory molecules as putative drug targets to tackle relapse to drug taking.


Assuntos
Cocaína/administração & dosagem , Locomoção/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Sinapses/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/patologia , Feminino , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ratos , Autoadministração , Síndrome de Abstinência a Substâncias/patologia , Sinapses/efeitos dos fármacos , Sinapses/patologia
4.
J Cell Biochem ; 120(4): 5495-5504, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30302813

RESUMO

MicroRNA-17-5p (miR-17-5p) and epithelial-mesenchymal transition (EMT) have been reported to participate in the development and progression of multiple cancers. However, the relationship between the miR-17-5p and EMT in osteosarcoma (OS) is still poorly understood. This study was to investigate the effects of the miR-17-5p and its potential mechanism in regulating proliferation, apoptosis, and EMT of human OS. Quantitative real-time PCR was used to detect the miR-17-5p and SRC kinase signaling inhibitor 1 (SRCIN1) messenger RNA expression in OS specimens and cell lines. After transfection with miR-17-5p inhibitors, proliferation, apoptosis, migration, and invasion of OS cells were assessed by using the Cell Counting Kit-8, the annexin V-FITC apoptosis, wound-healing, and transwell assays. The SRCIN1 was validated as a target of the miR-17-5p through bioinformatics algorithms and luciferase reporter assay. Moreover, the expression of EMT markers, E-cadherin, N-cadherin, and Snail was identified by the Western blot analysis. MiR-17-5p was significantly upregulated in OS tumor samples and cell lines. It inhibited proliferation and EMT, and promoted apoptosis in OS. The SRCIN1 was identified as a direct target of the miR-17-5p. Silenced miR-17-5p could change the expression of EMT markers, such as upregulating the expression of E-cadherin, and downregulating the expression of N-cadherin and Snail through targeting the antioncogenic SRCIN1. These findings suggest that the miR-17-5p promotes cell proliferation, and EMT in human OS by directly targeting the SRCIN1, and reveal a branch of the miR-17-5p/SRCIN1/EMT signaling pathway involved in the progression of OS.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Neoplasias Ósseas/metabolismo , Proliferação de Células , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , Osteossarcoma/metabolismo , RNA Neoplásico/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Feminino , Humanos , Masculino , MicroRNAs/genética , Proteínas de Neoplasias/genética , Osteossarcoma/genética , Osteossarcoma/patologia , RNA Neoplásico/genética
5.
Exp Cell Res ; 370(1): 174-188, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29928866

RESUMO

Hepatocellular carcinoma (HCC) is a male-dominant cancer. Several factors may contribute to the gender difference. Recent investigations have reported that miRNAs are involved in sex-linked signaling pathways and play a critical role in the molecular pathogenesis of hepatitis B virus (HBV)-related HCC. Therefore, we speculated that some of these miRNAs might contribute to the gender differences observed in HBV-related HCC. Our results showed that miR-371a-5p was significantly upregulated in tumor tissue and serum from HCC patients and that the expression level of miR-371a-5p was related to HBV infection, sexuality, TNM stage, adjacent organ invasion and microvascular invasion. Moreover, a high level of miR-371a-5p expression predicted poor overall survival of HCC patients, and in vitro and in vivo studies revealed that the overexpression of miR-371a-5p promoted proliferation and metastasis. Mechanistic investigations suggested that miR-371a-5p was upregulated by HBV and testosterone through LEF-1. SRCIN1 was a direct target of miR-371a-5p and reversed the effects of miR-371a-5p on HCC tumorigenesis. Our results also revealed that SRCIN1 negatively regulated the expression of PTN by inhibiting the activity of NF-κB. As a hepatocyte growth factor, PTN promoted EMT-induced metastasis in vitro and in vivo through the AKT/Slug pathway. These data strongly suggested that the upregulation of miR-371a-5p played an important role in HBV-related HCC. Through the LEF-1/miR-371a-5p/SRCIN1/PTN/Slug pathway, HBV and testosterone promote the proliferation and metastasis of hepatoma cells, especially in male patients with HBV-related HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Proliferação de Células/fisiologia , Vírus da Hepatite B/patogenicidade , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Metástase Neoplásica/patologia , Transdução de Sinais/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Carcinoma Hepatocelular/genética , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Citocinas/genética , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Hepatite B/genética , Hepatite B/patologia , Hepatite B/virologia , Humanos , Neoplasias Hepáticas/genética , Fator 1 de Ligação ao Facilitador Linfoide/genética , Masculino , MicroRNAs/genética , Fatores de Transcrição da Família Snail/genética , Regulação para Cima/genética
6.
Tumour Biol ; 37(2): 2765-71, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26408183

RESUMO

Cutaneous squamous cell carcinoma (cSCC) is an epidermal keratinocyte-derived skin tumor, which is the second most common skin cancer in the general population. Recently, studies showed that microRNAs (miRNAs) played an important role in the development of cancer. In our study, we showed that the expression of SRCIN1 was lower in cSCC tissues than in the matched normal tissues. Moreover, there was significant inversed correlation between miR-346 and SRCIN1 in cSCC tissues. The luciferase reporter assay data showed that miR-346 can target the SRCIN1 message via the 3'-untranslated region (UTR) of SRCIN1. Overexpression of miR-346 inhibited the messenger RNA (mRNA) and protein expression of SRCIN1 in the A431 cells. In addition, ectopic expression of miR-346 promoted the A431 cell proliferation and migration. Meanwhile, SRCIN1 overexpression inhibited the A431 cell proliferation and migration. Rescue experiment has showed that SRCIN1 overexpression reduced the miR-346-induced A431 cell proliferation and migration. Herein, this study may provide miR-346 as a new therapeutic target for cSCC.


Assuntos
Carcinoma de Células Escamosas/genética , MicroRNAs/genética , Oncogenes/genética , Neoplasias Cutâneas/genética , Regiões 3' não Traduzidas/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Queratinócitos/patologia , RNA Mensageiro/genética , Neoplasias Cutâneas/patologia
7.
Tumour Biol ; 37(1): 1151-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26277787

RESUMO

MicroRNAs (miRNAs) are a class of small non-coding RNAs that, when dysregulated, are involved in the initiation and progression of various cancers, including lung cancer, in humans. In the current study, qRT-PCR was performed to measure miR-211 expression in human non-small cell lung cancer (NSCLC) cell lines and tissues. Cell proliferation, cell cycle, colony formation, and invasion were performed to detect the functional role of miR-211 in human NSCLC cell line. We used luciferase reporter assay to find the potential target of miR-211. We found that miR-211 expression was upregulated in human non-small cell lung cancer (NSCLC) cell lines and tissues. The overexpression of miR-211 enhanced NSCLC cell proliferation, colony formation, and invasion. SRC kinase signaling inhibitor 1 (SRCIN1) was identified as a direct target of miR-211. SRCIN1 silencing promoted cell proliferation, and SRCIN1 expression was downregulated in human NSCLC cell lines. Thus, miR-211 may function as an oncogenic miRNA in NSCLC, partly by regulating SRCIN1, and the modulation of miR-211 expression represents a potential strategy for the treatment of NSCLC patients.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Células A549 , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Genes Reporter , Humanos , Invasividade Neoplásica
8.
Clinics (Sao Paulo) ; 79: 100403, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38878321

RESUMO

OBJECTIVES: This study aims to elucidate the role of circUSP9X (Circular RNA Ubiquitin Specific Peptidase 9 X-Linked) in the development of venous thrombosis in the lower extremities. METHODS: An animal model of Deep Vein Thrombosis (DVT) and a hypoxic model of Human Umbilical Vein Endothelial Cells (HUVECs) treated with Cobalt (II) Chloride (CoCl2) were developed. The expression levels of circUSP9X, microRNA-148b-3p (miR-148b-3p), and SRC Kinase Signaling Inhibitor 1 (SRCIN1) were quantified using quantitative reverse transcription Polymerase Chain Reaction and Western blot analysis. Cell cytotoxicity, viability, apoptosis, and inflammation in HUVECs were assessed via Lactate Dehydrogenase (LDH) assay, MTT assay, flow cytometry, Enzyme-Linked Immunosorbent Assay, and Western blot, respectively. Hematoxylin and Eosin staining were employed for histopathological examination of the venous tissues in the animal model. The interaction between circUSP9X, miR-148b-3p, and SRCIN1 was further explored through dual-luciferase reporter assays and RNA Immunoprecipitation experiments. RESULTS: The present findings reveal a significant upregulation of circUSP9X and SRCIN1 and a concurrent downregulation of miR-148b-3p in DVT cases. Knockdown of circUSP9X or overexpression of miR-148b-3p ameliorated CoCl2-induced apoptosis in HUVECs, reduced LDH release, enhanced cellular viability, and mitigated inflammation. Conversely, overexpression of circUSP9X intensified CoCl2's cytotoxic effects. The effects of manipulating circUSP9X expression were counteracted by the corresponding modulation of miR-148b-3p and SRCIN1 levels. Additionally, circUSP9X knockdown effectively inhibited the formation of DVT in the mouse model. A competitive binding mechanism of circUSP9X for miR-148b-3p, modulating SRCIN1 expression, was identified. CONCLUSION: circUSP9X promotes the formation of DVT through the regulation of the miR-148b-3p/SRCIN1 axis.


Assuntos
Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , MicroRNAs , Regulação para Cima , Trombose Venosa , Animais , Humanos , Masculino , Camundongos , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Apoptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , MicroRNAs/metabolismo , RNA Circular/genética , Regulação para Cima/efeitos dos fármacos
9.
Biomolecules ; 14(5)2024 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-38785978

RESUMO

Breast cancer is a leading cause of cancer mortality in women worldwide. Using the Infinium MethylationEPIC BeadChip, we analyzed plasma sample methylation to identify the SRCIN1 gene in breast cancer patients. We assessed SRCIN1-related roles and pathways for their biomarker potential. To verify the methylation status, quantitative methylation-specific PCR (qMSP) was performed on genomic DNA and circulating cell-free DNA samples, and mRNA expression analysis was performed using RT‒qPCR. The results were validated in a Western population; for this analysis, the samples included plasma samples from breast cancer patients from the USA and from The Cancer Genome Atlas (TCGA) cohort. To study the SRCIN1 pathway, we conducted cell viability assays, gene manipulation and RNA sequencing. SRCIN1 hypermethylation was identified in 61.8% of breast cancer tissues from Taiwanese patients, exhibiting specificity to this malignancy. Furthermore, its presence correlated significantly with unfavorable 5-year overall survival outcomes. The levels of methylated SRCIN1 in the blood of patients from Taiwan and the USA correlated with the stage of breast cancer. The proportion of patients with high methylation levels increased from 0% in healthy individuals to 63.6% in Stage 0, 80% in Stage I and 82.6% in Stage II, with a sensitivity of 78.5%, an accuracy of 90.3% and a specificity of 100%. SRCIN1 hypermethylation was significantly correlated with increased SRCIN1 mRNA expression (p < 0.001). Knockdown of SRCIN1 decreased the viability of breast cancer cells. SRCIN1 silencing resulted in the downregulation of ESR1, BCL2 and various cyclin protein expressions. SRCIN1 hypermethylation in the blood may serve as a noninvasive biomarker, facilitating early detection and prognosis evaluation, and SRCIN1-targeted therapies could be used in combination regimens for breast cancer patients.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama , Proliferação de Células , Metilação de DNA , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Neoplasias da Mama/diagnóstico , Metilação de DNA/genética , Feminino , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/sangue , Proliferação de Células/genética , Prognóstico , Pessoa de Meia-Idade , Regulação Neoplásica da Expressão Gênica , Detecção Precoce de Câncer , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/sangue , Linhagem Celular Tumoral , Adulto
10.
Andrology ; 9(6): 1923-1933, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34185441

RESUMO

BACKGROUND: The tricho-rhino-phalangeal syndrome-1 gene (Trps1) is an atypical GATA family member. Although current studies of Trps1 mainly focus on tumors, whether Trps1 plays a role in the male reproductive system remains unknown. OBJECTIVES: The purpose of this study was to elucidate the function of Trps1 in Leydig cells, indicating its regulatory mechanism on the cell cycle. METHODS: Gene-silencing technology, RNA-seq, RT-qPCR, and western blotting were used to evaluate the function of Trps1 in mouse primary Leydig cells and MLTC-1 cells. In addition, ChIP-base sets and ChIP-qPCR were employed to further assess the regulatory mechanism of Trps1 in MLTC-1 cells. RESULTS: Knockdown of Trps1 in Leydig cells significantly suppressed phosphorylation of Src and Akt and expression of Ccnd1, which was accompanied by impairment of cell proliferative ability. Trps1 may affect the cell cycle through the Src/Akt/Ccnd1 signaling pathway. In addition, Trps1 may bind to the promoter of Srcin1 to regulate its transcription, thus influencing Src phosphorylation levels and the proliferation of Leydig cells. DISCUSSION AND CONCLUSION: Src increases in Leydig cells during pubertal development, suggesting its functional involvement in differentiated adult Leydig cells. Inhibition of the Src/Akt pathway would reduce Ccnd1 expression. In the present study, we found that Trps1 may regulate the phosphorylation level of Src and Akt through Srcin1, targeting Ccnd1 to influence mouse Leydig cell proliferation. These findings shed light on the regulation of Trps1 on cell proliferation and differentiation of mouse Leydig cells.


Assuntos
Proliferação de Células/genética , Ciclina D1/fisiologia , Células Intersticiais do Testículo/metabolismo , Proteínas Repressoras/fisiologia , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Masculino , Camundongos , Transdução de Sinais/genética
11.
Cancer Biomark ; 29(3): 417-427, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32924985

RESUMO

BACKGROUND: Long non-coding RNA (lncNRA) forkhead box D3 antisense RNA 1 (FOXD3-AS1) has been proved to promote or suppress the occurrence and development of multiple types of human tumors. However, the function and mechanism of FOXD3-AS1 in non-small cell lung cancer (NSCLC) are scarcely understood. METHODS: qRT-PCR was used for detecting FOXD3-AS1, miR-150 and SRC kinase signaling inhibitor 1 (SRCIN1) mRNA expression in NSCLC tissues, and the relationship between pathological characteristics of NSCLC patients and FOXD3-AS1 expression level was analyzed. With human NSCLC cell lines H1299 and A549 as cell models, CCK-8 and BrdU assays were employed for detecting cancer cell proliferation, and Transwell assay was employed for detecting cell invasion ability. Dual luciferase reporter gene assay and RNA immunoprecipitation (RIP) assay were used for the verification of the targeting relationshipe between FOXD3-AS1 and miR-150, and Western blot was employed for detecting SRCIN1 protein expression. RESULTS: FOXD3-AS1 expression was significantly reduced in NSCLC tissues and cell lines, and low expression of FOXD3-AS1 was closely related to positive lymph node metastasis and relatively high tumor grade. FOXD3-AS1 over-expression inhibited the proliferation and invasion of H1299 cell lines, while its knockdown promoted the proliferation and invasion of A549 cells. Additionally, it was confirmed that FOXD3-AS1 suppressed the expression of miR-150 by targeting it, and up-regulated the expression of SRCIN1. CONCLUSIONS: FOXD3-AS1 indirectly enhances the expression of SRCIN1 by targeting miR-150, thereby inhibiting NSCLC progression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Células A549 , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão/patologia , Pulmão/cirurgia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Estadiamento de Neoplasias , Pneumonectomia , Regulação para Cima
12.
Front Oncol ; 10: 79, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117734

RESUMO

Background: Lung cancer is the most common malignant tumor worldwide. Accumulating results have shown that long non-coding RNAs (lncRNAs) play a key role in tumorigenesis. Patients and Methods: A total of 163 tumor tissues were collected from non-small cell lung cancer (NSCLC) patients from West China Hospital of Sichuan University. LincRNA00494 is a novel lncRNA, and its expression and biological effect in NSCLC were reported in this study. NSCLC cell lines were used in this study. Results: LincRNA00494 is mainly distributed in the cytoplasm. LincRNA00494 was downregulated in the tumor tissues compared with the adjacent non-tumor tissues. LincRNA00494 expression was positively correlated with SRCIN1 expression (R = 0.57, P < 0.05). Silencing of LincRNA00494 in the cell lines substantially decreased SRCIN1 expression at the mRNA and protein levels, whereas overexpression of LincRNA00494 enhanced the SRCIN1 levels. miR-150-3p significantly decreased the luciferase signals of LincRNA00494 and SRCIN1 reporters. After transfection with miR-150-3p mimics and miR-150-3p inhibitor, overexpression of LincRNA00494 decreased the proliferation of the H358 (36%) and H1299 (29%) cell lines compared with that of the control cells, as shown by CCK-8 assays, whereas silencing LincRNA00494 promoted the proliferation of the H358 (47%) and H1299 (35%) cells. Tumor growth from LincRNA00494-overexpressing xenografts was significantly decreased; additionally, LincRNA00494 silencing substantially increased tumor growth compared with that of the control cells. Conclusions: Functional experiments revealed that LincRNA00494 inhibited NSCLC cell proliferation, which might be related to the suppression of SRCIN1, a tumor suppressor gene, by acting as a decoy for miR-150-3p. The data showed that LincRNA00494 might have antineoplastic effects during NSCLC tumorigenesis through its role as a ceRNA.

13.
Onco Targets Ther ; 12: 4927-4936, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31417287

RESUMO

INTRODUCTION: Previous studies have shown that miR-373 functions as either a tumor suppressor or an oncogene depending on which type of cancer it's operating in. However, the functional role of miR-373 in neuroblastoma (NB) remains largely unclear. METHODS: Expression of miR-373 and SRC kinase signaling inhibitor 1 (SRCIN1) in 20 metastatic and 20 primary NB tissues was detected by quantitative real-time PCR (qRT-PCR) and Western blotting. MTT assay, flow cytometry analysis and transwell migration and invasion assays were performed to evaluate the influence of miR-373 inhibition on the growth, migration and invasion of NB cells, respectively. In vivo experiment was applied to determine the effect of miR-373 inhibition on tumor growth. Dual-luciferase reporter assay was used to confirm the interaction between miR-373 and SRCIN1. RESULTS: We observed a significant increase in the expression of miR-373 in metastatic NB samples compared with primary NB samples, and this was inversely correlated with SRCIN1 expression. Functional studies revealed that depletion of miR-373 inhibited in vitro NB cell growth, migration and invasion, and also suppressed tumor growth in an in vivo mouse model. Moreover, we identified that SRCIN1 was a direct and functional target gene of miR-373. Silencing of SRCIN1 partially rescued the antimiR-373-mediated inhibition of cell growth, migration and invasion. CONCLUSION: The data from our study verified a potential oncogenic role of miR-373 in NB cells that occurs through direct targeting SRCIN1. The newly identified miR-373/SRCIN1 axis represents a new potential candidate for therapeutic intervention of malignant NB.

14.
Cancer Med ; 8(12): 5687-5701, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31385464

RESUMO

Bone metastasis of breast cancer makes patients suffer from pain, fractures, spinal cord compression, and hypercalcemia, and is almost incurable. Although the mechanisms of bone metastasis in breast cancers have been studied intensively, novel specific target will be helpful to the development of new therapeutic strategy of breast cancer. Herein, we focused on the microRNA of tumor cell-derived exosomes to investigate the communication between the bone microenvironment and tumor cells. The expression of miR-20a-5p in the primary murine bone marrow macrophages (BMMs), MCF-10A, MCF-7, and MDA-MB-231 cell lines, as well as the cell-derived exosomes were assessed by qRT-PCR. Transwell assays were used to evaluate the effects of miR-20a-5p on tumor cell migration and invasion. The expression of exosomes marker including CD63and TSG101 was detected by Western Blot. Cell cycle distribution of BMMs was analyzed by flow cytometry. 3-UTR luciferase reporter assays were used to validate the putative binding between miR-20a-5p and SRCIN1. MiR-20a-5p was highly expressed in breast tumor tissues and the exosomes of MDA-MB-231 cells. MiR-20a-5p promoted migration and invasion in MDA-MB-231 cells, and the proliferation and differentiation of osteoclasts. MDA-MB-231 cell-derived exosomes transferred miR-20a-5p to BMMs and facilitated the osteoclastogenesis via targeting SRCIN1. The present work provides evidence that miR-20a-5p transferred from breast cancer cell-derived exosomes promotes the proliferation and differentiation of osteoclasts by targeting SRCIN1, providing scientific foundations for the development of exosome or miR-20a-5p targeted therapeutic intervention in breast cancer progression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Neoplasias da Mama/patologia , Exossomos/genética , MicroRNAs/genética , Regulação para Cima , Regiões 3' não Traduzidas , Animais , Neoplasias da Mama/genética , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Tetraspanina 30/metabolismo , Fatores de Transcrição/metabolismo , Microambiente Tumoral
15.
Pathol Res Pract ; 215(6): 152382, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30890278

RESUMO

MicroRNAs (miRNAs) play a vital role in the progress of cancer. Whereas the expression and function of miR-374a in pancreatic cancer remain largely unknown. In this study, pancreatic cancer samples and its adjacent normal tissues were obtained from 30 clinical patients with pancreatic cancer. Quantitative real-time PCR (qRT-PCR) was used to measure miR-374a and SRC Kinase Signaling Inhibitor 1 (SRCIN1) expression. Western blotting assay was performed to measure the levels of SRCIN1, E-cadherin, N-cadherin, Vimentin, Zonula occludens-1 (ZO-1) and ß-catenin in PANC-1 cells. Luciferase reporter assay was conducted to confirm the direct targeting of SRCIN1 by miR-374a. Cell proliferation and migration assays were utilized to analyze the role of miR-374a in PANC-1 cells. We found that miR-374a expression was upregulated in pancreatic cancer tissues and cell lines. Over-expression of miR-374a promoted cell proliferation, migration and epithelial-mesenchymal transition (EMT) in pancreatic cancer. While, SRCIN1 expression was downregulated in pancreatic cancer tissues and cells. SRCIN1 was found to be a potential targets of miR-374a by dual-luciferase reporter assay. And SRCIN1 was down-regulated after miR-374a transfection. More than that, over-expression of SRCIN1 inhibited cell proliferation, migration and EMT in pancreatic cancer cell. Therefore, this study revealed that miR-374a promoted cell proliferation, migration and EMT via targeting SRCIN1 in pancreatic cancer.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Adenocarcinoma/patologia , Transição Epitelial-Mesenquimal/genética , MicroRNAs/metabolismo , Neoplasias Pancreáticas/patologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
16.
Pathol Res Pract ; 215(4): 738-747, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30679084

RESUMO

Cervical carcinoma is one of the most universal cancers among women. Recent researches have reported that microRNA-150-5p (miR-150-5p) is up-regulated in diverse carcinomas containing cervical carcinoma. The purpose of this study was to further investigate the potential role of miR-150-5p in the progress of cervical carcinoma cells including proliferation and epithelial-mesenchymal transition (EMT).The ability of miR-150-5p to promote carcinogenesis was analyzed using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot assays, respectively. Bioinformatics analyses predicted and identified whether SRC kinase signaling inhibitor 1 (SRCIN1) was served as a potential target of miR-150-5p. C-33A and HeLa cells were utilized to determine the function of miR-150-5p through targeting SRCIN1. Among the aberrantly expressed miRNAs, miR-150-5p was significantly revealed differential expression in cervical carcinoma cell lines and was closely relevant to cell growth regulation. Furthermore, we found that SRCIN1 overexpression could obviously inhibit the proliferation and EMT of cervical cancer cells triggered by miR-150-5p mimics as well as accelerated the apoptosis of cervical carcinoma cells. In conclusion, our data demonstrated that miR-150-5p could promote the proliferation and EMT of cervical carcinoma cells via targeting SRCIN1. Thus, miR-150-5p may hold a promise as a prognostic biomarker and potential therapeutic target for cervical carcinoma.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , MicroRNAs/metabolismo , Neoplasias do Colo do Útero/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , MicroRNAs/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia
17.
Front Cell Dev Biol ; 7: 222, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31681758

RESUMO

The p140Cap adaptor protein is a scaffold molecule physiologically expressed in few epithelial tissues, such as the mammary gland, and in differentiated neurons. While the role of p140Cap in mammary gland epithelia is not still understood, we already know that a significant subset of breast cancers express p140Cap. In the subgroup of ERBB2-amplified breast cancers, a high p140Cap status predicts a significantly lower probability of developing a distant event and a clear difference in survival. p140Cap is causal in dampening ERBB2-positive tumor cell progression, impairing tumor onset and growth, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. Since only a few p140Cap interacting proteins have been identified in breast cancer and the molecular complexes and pathways underlying the cancer function of p140Cap are largely unknown, we generated a p140Cap interactome from ERBB2-positive breast cancer cells, identifying cancer specific components and those shared with the synaptic interactome. We identified 373 interacting proteins in cancer cells, including those with functions relevant to cell adhesion, protein homeostasis, regulation of cell cycle and apoptosis, which are frequently deregulated in cancer. Within the interactome, we identified 15 communities (clusters) with topology-functional relationships. In neurons, where p140Cap is key in regulating synaptogenesis, synaptic transmission and synaptic plasticity, it establishes an extensive interactome with proteins that cluster to sub complexes located in the postsynaptic density. p140Cap interactors converge on key synaptic processes, including synaptic transmission, actin cytoskeleton remodeling and cell-cell junction organization. Comparing the breast cancer to the synaptic interactome, we found 39 overlapping proteins, a relatively small overlap. However, cell adhesion and remodeling of actin cytoskeleton clearly emerge as common terms in the shared subset. Thus, the functional signature of the two interactomes is primarily determined by organ/tissue and functional specificity, while the overlap provides a list of shared functional terms, which might be linked to both cancer and neurological functions.

18.
Cancer Biother Radiopharm ; 33(4): 131-138, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29694242

RESUMO

Pathological angiogenesis can be a significant barrier to effective cancer therapy. Recent evidence suggests that Endostar may induce vascular normalization, thereby improving tumor perfusion and systemic chemotherapy. However, the molecular mechanism by which Endostar makes chemotherapy more effective remains to be fully elucidated. In this study, established 4T1 breast tumor-bearing animals treated with Endostar were evaluated at serial time points for treatment-associated changes in vascular architecture. As a result, Endostar induced a morphologically and functionally normalized vascular network. Combined Endostar and doxorubicin exhibited significant antitumor (34% of control size) and antimetastatic effects (29% of control metastatic nodules) in vivo. Finally, a two-dimensional gel electrophoresis and MALDIQ-TOF MS/MS-based proteomics approach was used to identify differentially expressed proteins involved in vascular normalization during Endostar administration. SRCIN1 was detected as one of the most significantly increased proteins. SRCIN1 is a novel Src-binding protein that regulates Src activation through C-terminal Src kinase, and attenuated Src activation during Endostar treatment was further confirmed by immunoblotting. Collectively, these data provided a molecular basis for vascular normalization, which were associated with the observed synergistic effect in vivo.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endostatinas/uso terapêutico , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Endostatinas/farmacologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos BALB C , Transdução de Sinais
19.
Gene ; 600: 21-28, 2017 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-27913185

RESUMO

MicroRNAs (miRNAs) are a class of highly conserved small noncoding RNAs that play pivotal roles at the post-transcriptional level in the biological function of various cancers, including breast cancer. In our study, miR-346 mimic, inhibitor, negative control or si-SRCIN1 were transfected into MCF-7 and MCF-7/Doc cells, respectively. Quantitative real time PCR (qRT-PCR) was used to measure miR-346 and SRCIN1 mRNA expressions and western blot was used to detect the expression of SRCIN1 in protein level. CCK-8 and colony formation were employed to verify cell viability and proliferation. Flow cytometry showed the apoptosis. Transwell was performed to detect migration and invasion. The luciferase reporter assay data showed the target correlation of miR-346 and SRCIN1. Firstly, we found that the expression of miR-346 was higher in breast cancer tissues than in their paired corresponding non-cancerous tissues and there was significant inversed correlation between miR-346 and SRCIN1. Overexpression of miR-346 promoted cell proliferation, colony formation, migration and invasion, and reduced apoptosis, sensitivity to Docetaxel (Doc). SRCIN1 was identified as a direct target of miR-346, whose silencing promoted cell proliferation and the IC50 of Doc. Moreover, SRCIN1 silencing reduced the effect of miR-346 down-expression. Taken together, miR-346 may function as an oncogenic miRNA and mediate chemosensitivity to docetaxel through targeting SRCIN1 in breast cancer, targeted modulation of miR-346 expression may became a potential strategy for the treatment.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/genética , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , MicroRNAs/genética , Taxoides/farmacologia , Apoptose/genética , Neoplasias da Mama/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Docetaxel , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Invasividade Neoplásica/genética , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
20.
Am J Transl Res ; 7(11): 2519-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26807196

RESUMO

microRNAs (miRNAs) are endogenously expressed, conserved and small noncoding RNA that regulate gene expression by the post-transcriptional level. In this study, we aim to examine the role of miR-873 in lung adenocarcinoma. We found that the expression of miR-873 was upregulated in four lung adenocarcinoma cell lines and tissues. In addition, the expression levels of SRCIN1 were inversely correlated with the expression levels of miR-873 in lung adenocarcinoma tissues. Furthermore, SRCIN1 was confirmed asthe direct target of miR-873 by luciferase reporter assay and Western blotting. Overexpression of miR-873 promoted the proliferation and migration of lung adenocarcinoma cells, while SRCIN1 upregulation inhibited their proliferation and migration. Restoration of SRCIN1 could significantly reverse the proliferation and migration promotion imposed by miR-873. In summary, this study reveals for the first time that miR-873 increase the lung adenocarcinoma cell proliferation and migration through directly inhibiting SRCIN1 expression.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA