Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Urol ; 23(1): 155, 2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37789353

RESUMO

Bladder cancer stem cells (BCSCs) are considered as the root cause of BC initiation and recurrence, and exosomes derived from BCSCs (CSCs-exo) are the vital tool for establishing a stable tumor microenvironment. miR-105-5p has been revealed to promote tumor growth in a variety of cancers, but the effects on BC are still not included.Characteristics of CSCs-exo were examined by transmission electron microscope and nanoparticle tracking analysis. PKH67 dye was used to observe the cellular uptake of exosomes. Cell viability, migration and invasion were detected by CCK-8, wound healing and transwell invasion assays, respectively. The interaction between miR-105-5p and GPR12 was verified by luciferase activity assay. Xenografts were induced in the nude mice, and H&E staining method was applied to analyze the histological changes of xenografts. CSCs-exo efficiently promoted BC cell viability, migration and invasion. miR-105-5p was highly expressed in CSCs and CSCs-exo treatment significantly upregulated the expression of miR-105-5p in BC cells.GPR12 was subsequently verified to be the target gene of miR-105-5p, and overexpression of GPR12 abrogated the effects of miR-105-5p on BC cell growth and metastasis. Reversely, the anti-tumor function of miR-105-5p antagomir was observed in the xenograft mice.CSCs aggravated the malignancy of BC partly through transmitting exosomal miR-105-5p to BC cells to inhibit the expression of GPR12, which developed a novel aspect for CSC-targeted therapies.


Assuntos
MicroRNAs , Neoplasias da Bexiga Urinária , Humanos , Animais , Camundongos , Bexiga Urinária/patologia , Camundongos Nus , Movimento Celular , MicroRNAs/genética , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Microambiente Tumoral , Receptores Acoplados a Proteínas G/genética
2.
BMC Cancer ; 21(1): 1069, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34592939

RESUMO

BACKGROUND: Colorectal cancer (CRC) represents one of the major malignant cancers in the world. It has been demonstrated that long non-coding RNAs (lncRNAs) can cause great influences on various human cancers. Though MCF.2 cell line derived transforming sequence like antisense RNA 1 (MCF2L-AS1) and its carcinogenic effect in CRC has been elucidated by several previous researches, the underlying mechanism remains unknown. AIM: We aimed at exploring the function and regulatory mechanism of MCF2L-AS1 in CRC. METHODS: MCF2L-AS1 expression in CRC cells was tested via RT-qPCR assay. The effects of MCF2L-AS1 on the biological properties of CRC cells were testified through functional experiments. The molecular mechanism of MCF2L-AS1 was verified through mechanism experiments. RESULTS: MCF2L-AS1 was highly expressed in CRC cells, and it could enhance the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) process of CRC cells. MiR-105-5p was sponged by MCF2L-AS1 in CRC cells and Ras-related protein Rab-22A (RAB22A) was verified to be the downstream target of miR-105-5p. It was verified through rescue assays that RAB22A overexpression or miR-105-5p silencing could reverse the repressive impact of MCF2L-AS1 silencing on CRC progression. CONCLUSION: MCF2L-AS1 accelerated the malignant development of CRC cells by targeting the miR-105-5p/RAB22A axis.


Assuntos
Neoplasias Colorretais/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/patologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Inativação Gênica , Humanos , MicroRNAs/genética , Invasividade Neoplásica
3.
Aging (Albany NY) ; 16(15): 11568-11576, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39103205

RESUMO

Osteosarcoma is a highly metastatic, aggressive bone cancer that occurs in children and young adults worldwide. Circular RNAs (circRNAs) are crucial molecules for osteosarcoma progression. In this study, we aimed to investigate the impact of circMRPS35 overexpression and its interaction with FOXO1 via evaluating apoptosis, cell cycle, and bioinformatic analyses on the malignant development of osteosarcoma in MG63 and MNNG/HOS cells. We found that circMRPS35 overexpression reduced osteosarcoma cell viability and inhibited tumor growth in vivo. It increased the apoptosis rate and induced cell cycle arrest in osteosarcoma cells. We identified a potential interaction between circMRPS35 and FOXO1 with miR-105-5p using bioinformatics analysis. Overexpression of circMRPS35 decreased miR-105-5p expression, whereas miR-105-5p mimic treatment increased its expression. This mimic also suppressed the luciferase activity of circMRPS35 and FOXO1 and reduced FOXO1 expression. Overexpression of circMRPS35 elevated FOXO1 protein levels, but this effect was reversed by co-treatment with the miR-105-5p mimic. We demonstrated that inhibiting miR-105-5p decreased viability and induced apoptosis. Overexpression of FOXO1 or treatment with a miR-105-5p inhibitor could counteract the effects of circMRPS35 on viability and apoptosis in osteosarcoma cells. Therefore, we concluded that circMRPS35 suppressed the malignant progression of osteosarcoma via targeting the miR-105-5p/FOXO1 axis.


Assuntos
Apoptose , Neoplasias Ósseas , Proteína Forkhead Box O1 , Regulação Neoplásica da Expressão Gênica , MicroRNAs , Osteossarcoma , RNA Circular , Animais , Humanos , Camundongos , Apoptose/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Progressão da Doença , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , RNA Circular/genética , RNA Circular/metabolismo
4.
Am J Med Sci ; 2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39313115

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer (BC). Some potential molecular targets have been identified, and miR-105-5p was found to be abnormally expressed in TNBC tissues. OBJECTIVE: The objective of this study was to probe the effect of miR-105-5p on TNBC via FOXG1/HDAC2-mediated acetylation. METHODS: An animal model of TNBC was established by injecting BC cells into the axillary area of nude mice. The levels of miR-105-5p, FOXG1, HDAC2, Bcl-2, Bax, and Ki67 were detected via RT‒qPCR, Western blotting and immunohistochemistry. Flow cytometry, CCK-8, Transwell and colony formation assays were used to measure apoptosis, proliferation and migration, respectively. Total histone acetylation levels were measured by ELISA. The binding of FOXG1 to HDAC2 was detected by co-immunoprecipitation. The binding relationship between miR-105-5p and FOXG1 was verified using a dual-luciferase reporter gene assay. RESULTS: In this study, miR-105-5p and HDAC2 were highly expressed in the MDA-MB-231 and BT-549 BC cell lines, whereas FOXG1 was expressed at low levels. The inhibition of miR-105-5p inhibited the proliferation and migration of MDA-MB-231 and BT-549 cells and promoted their apoptosis. Bioinformatics analysis revealed that miR-105-5p and FOXG1 had a negative targeting regulatory relationship. FOXG1 overexpression had a similar effect on cancer cells as the inhibition of miR-105-5p. Moreover, experiments revealed that FOXG1 and HDAC2 could bind to each other and that HDAC2 overexpression or treatment with the histone acetyltransferase inhibitor Garcinol weakened the effect of FOXG1 overexpression. In addition, FOXG1 knockdown inhibited the effect of the miR-105-5p inhibitor, while Garcinol treatment further enhanced the effect of FOXG1 knockdown, inhibited histone acetylation, promoted the proliferation and migration of cancer cells, and inhibited apoptosis. Moreover, the in vivo results confirmed the in vitro results. CONCLUSION: miR-105-5p promotes HDAC2 expression by reducing FOXG1, inhibits histone acetylation, and aggravates the malignant biological behavior of TNBC cells.

5.
Cell Cycle ; 22(7): 808-817, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36529892

RESUMO

As a type of lung cancer, non-small cell lung cancer (NSCLC) has the characteristics of high mortality and high recurrence rate, which poses a great threat to human life and health. Due to the high risk of surgical treatment and the slow recovery of wounds, non-coding RNAs, especially lncRNAs are used as new potential clinical prognostic markers to prevent and treat cancer in advance. This study aims to explore the role of FAM138B in NSCLC and its possibility as a prognostic biomarker. Real-timequantitative polymerase chain reaction (RT-qPCR) was used to detect the expression and overexpression level of lncRNA FAM138B (FAM138B) in cells and tissues. The CCK-8, Transwell migration and invasion methods were performed to observe the cell transfection.The interaction between FAM138B and miR-105-5p was predicted by the bioinformatics tool starBase v2.0, and verified by the luciferase reporter gene experiment. Kaplan-Meier and Cox regression analyses were used to determine the prognostic significance of FAM138B in NSCLC. The expression of FAM138B is down-regulated in NSCLC cells and tissues. Overexpression of FAM138B can inhibit the expression level of miR-105-5p in NSCLC cells, and the ability of NSCLC cells to proliferate, migrate and invade is downregulated. FAM138B targets miR-105-5p, and there is a negative correlation between FAM138B and miR-105-5p. It is confirmed that FAM138B inhibits the progression of NSCLC by targeting miR-105-5p and can be a potential prognostic biomarker for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , MicroRNAs/metabolismo , Proliferação de Células/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
6.
PeerJ ; 10: e12588, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35702258

RESUMO

Background: Previous studies have been reported the immune dysfunction of various live tissues. However, the potential molecular mechanism of post-transcriptional regulation of immune related genes in hepatocellular carcinoma (HCC) is still not clear. We tried to identify crucial immune related biomarkers associated with HCC patients' outcomes and to reveal the transcriptional regulation. Method: The fractions of 22 immune cells in tumor and adjacent tissues were estimated by CIBERSORT. Kruskal-Wallis test and differentially expressed analyzes were used for comparative studies. Cox proportional hazard regression model, Kaplan-Meier estimates and Log-rank test were used for survival analyses. Results: From The Cancer Genome Atlas (TCGA), the gene, lncRNA and miRNA expression profiles of 379 HCC samples with clinical information were used for comparative studies. Eleven adaptive and innate immune cell types were significantly altered in HCC samples, including B cell memory, regulatory T cells and follicular helper T cells. Differentially expressed competing endogenous RNA (ceRNA) network associated with patients' overall survival was identified. Then, the novel pathway, including LINC00261, MiR105-5p and selectin L(SELL) was found and may be potential novel biomarkers for patients' outcomes and immunotherapy. Furthermore, SELL was significantly positively correlated (correlation coefficients: 0.47-0.69) with 12 known gene signatures of immunotherapy except for programmed cell death 1 (PDCD1). Conclusions: Our findings could provide insights into the selection of novel LINC00261/MiR105-5p/SELL pathway which is associated with overall survival and may impact on efficacy of immunotherapy in HCC.


Assuntos
Carcinoma Hepatocelular , Selectina L , Neoplasias Hepáticas , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/genética , Modelos de Riscos Proporcionais , RNA Longo não Codificante/genética , Selectina L/genética , Linfócitos B
7.
Front Genet ; 13: 819699, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35309127

RESUMO

Objective: Esophageal squamous cell carcinoma (ESCC) presents high morbidity and mortality. It was demonstrated that blood-derived vesicles can facilitate ESCC development and transmit regulating signals. However, the molecular mechanism of vesicle miRNA secreted by tumor cells affecting ESCC progression has not been explored. Methods: The mRNA-related signaling pathways and differentially expressed genes were screened out in TCGA dataset. The levels of miRNA-105-5p and SPARCL1 were determined by qRT-PCR. Protein level determination was processed using Western blot. The interaction between the two genes was verified with the dual-luciferase method. A transmission electron microscope was utilized to further identify extracellular vesicles (EVs), and co-culture assay was performed to validate the intake of EVs. In vitro experiments were conducted to evaluate cell function changes in ESCC. A mice tumor formation experiment was carried out to observe tumor growth in vivo. Results: MiRNA-105-5p expression was increased in ESCC, while SPARCL1 was less expressed. MiRNA-105-5p facilitated cell behaviors in ESCC through targeting SPARCL1 and regulating the focal adhesion kinase (FAK)/Akt signaling pathway. Blood-derived external vesicles containing miRNA-105-5p and EVs could be internalized by ESCC cells. Then, miRNA-105-5p could be transferred to ESCC cells to foster tumorigenesis as well as cell behaviors. Conclusion: EV-carried miRNA-105-5p entered ESCC cells and promoted tumor-relevant functions by mediating SPARCL1 and the FAK/Akt signaling pathway, which indicated that the treatment of ESCC via serum EVs might be a novel therapy and that miRNA-105-5p can be a molecular target for ESCC therapy.

8.
Immunobiology ; 227(1): 152163, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34896914

RESUMO

BACKGROUND: Docetaxel (DTX) resistance is a common factor in metastatic prostate cancer (PC) chemotherapy that leads to treatment failure. Because lncRNA is involved in a variety of regulatory processes in tumor progression, this study aimed to explore the function and mechanism of LINC00184 in docetaxel resistance of PC. METHODS: Two PC cell lines and their docetaxel resistant cell lines (DU145/DTX and PC3/DTX) were used. The expression of LINC00184 in both cell lines and PC patient samples were evaluated. SiRNA knocking down was used to test the function of LINC00184 in proliferation and colony formation. Interaction between LINC00184 and its target miR-105-5p, as well as miR-105-5p and PD-L1 was checked by luciferase reporter assay and RNA pull-down assay. PC cell line and CD8 + T cell co-culture system was established, miR-105-5p inhibitor was co-transfected with LINC00184 siRNA to investigate the underline mechanism. RESULTS: LINC00184 was found to be associated with docetaxel resistance and adverse prognosis of prostate cancer. It regulated docetaxel resistance and T-cell-mediated immune response in prostate cancer cells. LINC00184 was induced by adsorption of miR-105-5p and negatively regulated it, subsequently inhibited the expression level of PD-L1. CONCLUSIONS: LINC00184 promoted docetaxel resistance and immune escape in prostate cancer cells by adsorption of miR-105-5p, resulted in upregulation of the expression of PD-L1. LINC00184 could possibly be considered as a potential target for treatment in prostate cancer patients with docetaxel-resistance.


Assuntos
MicroRNAs , Neoplasias da Próstata , RNA Longo não Codificante , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Docetaxel/farmacologia , Docetaxel/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Longo não Codificante/genética , RNA Interferente Pequeno
9.
Stem Cell Res Ther ; 12(1): 286, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33985571

RESUMO

BACKGROUND: Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) have emerged as a promising new therapeutic strategy for intervertebral disc degeneration (IVDD). However, the drawbacks of MSCs, including their invasive access, the donor age, and their limited proliferative capacity, hinder the quantity and quality of MSC-sEVs. Induced pluripotent stem cell-derived MSCs (iMSCs) provide an indefinite source of MSCs with well-defined phenotype and function. This study aimed to investigate the therapeutic effect of sEVs derived from iMSC (iMSC-sEVs) on IVDD and explore the underlying molecular mechanisms. METHODS: IVDD models were established by puncturing discs from the tails of rats. Then, iMSC-sEVs were injected into the punctured discs. The degeneration of punctured discs was assessed using MRI and HE and immunofluorescence staining. The age-related phenotypes were used to determine the effects of iMSC-sEVs on senescent nucleus pulposus cells (NPCs) in vitro. Western blotting was used to detect the expression of Sirt6. miRNA sequencing analysis was used to find miRNAs that potentially mediate the activation of Sirt6. RESULTS: After intradiscally injecting iMSC-sEVs, NPC senescence and IVDD were significantly improved. iMSC-sEVs could rejuvenate senescent NPCs and restore the age-related function by activating the Sirt6 pathway in vitro. Further, microRNA sequence analysis showed that iMSC-sEVs were highly enriched in miR-105-5p, which played a pivotal role in the iMSC-sEV-mediated therapeutic effect by downregulating the level of the cAMP-specific hydrolase PDE4D and could lead to Sirt6 activation. CONCLUSION: iMSC-sEVs could rejuvenate the senescence of NPCs and attenuate the development of IVDD. iMSC-sEVs exerted their anti-ageing effects by delivering miR-105-5p to senescent NPCs and activating the Sirt6 pathway. Our findings indicate that iMSCs are a promising MSC candidate for obtaining sEVs on a large scale, while avoiding several defects related to the present applications of MSCs, and that iMSC-sEVs could be a novel cell-free therapeutic tool for the treatment of IVDD.


Assuntos
Vesículas Extracelulares , Células-Tronco Pluripotentes Induzidas , Degeneração do Disco Intervertebral , Disco Intervertebral , Células-Tronco Mesenquimais , MicroRNAs , Núcleo Pulposo , Animais , Degeneração do Disco Intervertebral/terapia , MicroRNAs/genética , Ratos
10.
J Mol Histol ; 52(5): 991-1006, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34387789

RESUMO

Emerging evidence reveals that long noncoding RNAs (lncRNAs) contribute to human tumorigenesis. Nevertheless, the function of HOXC cluster antisense RNA 3 (HOXC-AS3) in human cervical cancer (CC) remains largely unknown. The levels of HOXC-AS3, miR-105-5p and SOS1 in CC tissues and cells were monitored by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Gain- and loss-of-function experiments were conducted to verify the function of HOXC-AS3 and miR-105-5p in CC cells. Meanwhile, cell proliferation, apoptosis, migration and invasion were examined by the cell counting kit-8 (CCK8) experiment, colony formation assay, flow cytometry and Transwell assay. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were carried out to test the regulatory interaction of HOXC-AS3, miR-105-5p and SOS1. In addition, in vivo experiment was performed to certain the role of HOXC-AS3 in tumorigenesis of CC. HOXC-AS3 was overexpressed in CC tissues (vs. adjacent normal tissues) and CC cells. Besides, the higher HOXC-AS3 profile was associated with the poorer clinical prognosis of CC patients. Overexpression of HOXC-AS3 promoted cell growth, migration and invasion, hampered apoptosis, whereas knocking down HOXC-AS3 exhibited the reverse effects. MiR-105-5p was a downstream target of HOXC-AS3, and it mediated the HOXC-AS3-induced oncogenic effects. Mechanistically, the bioinformatic analysis illustrated that SOS1 was targeted by miR-105-5p. Up-regulating SOS1 heightened the growth, migration and invasion of CC cells by enhancing the ErbB signaling pathway, which was reversed by miR-105-5p. Up-regulated HOXC-AS3 aggravates CC by promoting SOS1 expression via targeting miR-105-5p.


Assuntos
Progressão da Doença , Receptores ErbB/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Animais , Sequência de Bases , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Metástase Neoplásica , RNA Longo não Codificante/genética , Proteína SOS1/metabolismo
11.
Thorac Cancer ; 12(4): 539-548, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33372399

RESUMO

BACKGROUND: Accumulating evidence shows that microRNAs are aberrantly expressed and exert essential roles in the tumorigenesis and tumor progression of non-small cell lung cancer (NSCLC). METHODS: The plasma miRNAs from five healthy donors and four NSCLC patients were profiled by miRNA microarray. The differentially expressed miRNAs from 154 primary NSCLC patients and 146 healthy donors were subjected to RNA isolation and verified by quantitative PCR (qPCR). RESULTS: The miRNA microarray analysis revealed that 40 differential miRNAs between NSCLC patients and healthy donors were selected. We found that the plasma miR-1247-5p, miR-301b-3p and miR-105-5p levels of patients were significantly higher than those of healthy controls. The receiver operating characteristic curve (ROC) analyses revealed higher area under the ROC curve (AUC) values and higher sensitivity/specificity of carcinoembryonic antigen (CEA) in combination with miR-1247-5p, miR-301b-3p, or miR-105-5p were superior to that of CEA alone. CONCLUSIONS: High miR-1247-5p, miR-301b-3p and miR-105-5p expression have been demonstrated to accelerate tumorigenesis, and these three miRNAs in plasma act as novel biomarkers for the early diagnosis of NSCLC patients. KEY POINTS: Plasma miR-1247-5p, miR-301b-3p and miR-105-5p act as novel biomarkers for early NSCLC and NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , MicroRNAs/sangue , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/patologia , Estudos de Casos e Controles , Detecção Precoce de Câncer , Feminino , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade
12.
FEBS Lett ; 593(14): 1777-1786, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31127852

RESUMO

Pescadillo (PES1) is a key molecule for ribosome formation in mammalian cells. In this study, human hepatoma C3A cells were reprogrammed by four transcription factors, Oct4, Sox2, Klf4 and c-Myc, into induced cancer stem cells, termed C3A-induced cancer stem cells (C3A-iCSCs). We found that PES1 was up-regulated in C3A-iCSCs and promoted cell proliferation. Moreover, the cancer stem cell marker CD44, which is located in the cytomembrane, translocated to the nucleus and was up-regulated in C3A-iCSCs. Our results suggest that CD44 has a negative effect on miR-105-5p. We found that PES1 is a direct target of, and was negatively regulated by, miR-105-5p. In summary, CD44 regulates PES1 in liver cancer stem cells via miR-105-5p to promote cell growth.


Assuntos
Receptores de Hialuronatos/metabolismo , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas/genética , Proliferação de Células , Células Hep G2 , Humanos , Fator 4 Semelhante a Kruppel , Proteínas de Ligação a RNA/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA