Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 238
Filtrar
1.
Mol Biol Rep ; 48(8): 6093-6101, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34374893

RESUMO

BACKGROUND: Strong evidence indicated that high expression of HBXIP (also known as LAMTOR5) promotes cancer cells proliferation and helps cancer progression. Long non-coding RNAs (lncRNA) have also a crucial role in developing cancer. In this study, we aimed to determine the expression of LAMTOR5 and its nearby lncRNA, LAMTOR5-AS1 and investigate their potential as a biomarker in colorectal cancer (CRC) patients. METHODS: 75 tissues of colorectal tumors and non-tumor adjacent normal sampled in this study. After RNA procedure then RT-qPCR was applied for expression analysis. Moreover, in silico investigation also enrolled for predicting sponging effect of lncRNA with miRNAs. RESULTS: LAMTOR5 transcription level significantly overexpressed (p value < 0.001) and has shown a diagnostic potential (AUC = 0.8) in CRC. LAMTOR5-AS1 did not indicate any remarkable expression change overall, but showed a significant overexpressed in elderly patients (> 60) with CRC (p value < 0.0097). Moreover, the correlation analysis between LAMTOR5 and LAMTOR5-AS1 revealed a significant association in CRC (p value = 0.0074) which can be partly explained by its predicting act as a mediator with sponging effect on hsa-miR-let-7b-3p and hsa-miR-20a-5p. CONCLUSION: LAMTOR5 gene can be considered as prognostic biomarker for CRC. LAMTOR5-AS5 which is a nearby lncRNA of this gene could play a regulatory impact through its sponging effect on hsa-miR-let-7b-3p and hsa-miR-20a-5p which both have shown a significant impact on overall survival rate in CRC patients in high expression levels.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais/genética , RNA Longo não Codificante/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , MicroRNAs/genética , Oligorribonucleotídeos Antissenso/genética , Transcriptoma/genética
2.
Cells ; 9(11)2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33138194

RESUMO

Antisense DNA oligonucleotides, short interfering RNAs (siRNAs), and CRISPR/Cas9 genetic tools are the most useful therapeutic nucleic acids regulating gene expression based on the antisense specificity towards messenger RNA. Here, we present an effective novel strategy for inhibiting translation based on the antisense-controlled formation of an RNA quadruplex-duplex hybrid (QDH) between a G-rich RNA antisense oligoribonucleotide (Q-ASO) and specific mRNA, comprising two distant G-tracts. We selected epidermal growth factor receptor (EGFR) as a well-established target protein in anticancer therapy. The chemically modified, bi-functional anti-EGFR Q-ASO and a 56-nt long EGFR mRNA fragment, in the presence of potassium ions, were shown to form in vitro very stable parallel G-quadruplex containing a 28-nt long external loop folding to two duplex-stem structure. Besides, the Q-ASOs effectively reduced EGFR mRNA levels compared to the non-modified RNA and DNA antisense oligonucleotides (rASO, dASO). In addition, the hybridization specificity of Q-ASO comprising a covalently attached fluorescent tag was confirmed in living cells by visualization of the G4 green fluorescent species in the presence of other antisense inhibitors under competitive conditions. The results presented here offer novel insights into the potential application of Q-ASOs for the detection and/or alteration of (patho)biological processes through RNA:RNA quadruplex-duplex formation in cellular systems.


Assuntos
Receptores ErbB/metabolismo , Quadruplex G , Oligorribonucleotídeos Antissenso/metabolismo , RNA Mensageiro/genética , Sobrevivência Celular , Fluorescência , Inativação Gênica , Células HeLa , Humanos , Mitocôndrias/metabolismo , Conformação de Ácido Nucleico , Hibridização de Ácido Nucleico , Oligorribonucleotídeos Antissenso/química , Espectroscopia de Prótons por Ressonância Magnética , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Temperatura
3.
Cell Biol Int ; 44(11): 2284-2292, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32749739

RESUMO

Radiotherapy is an important adjuvant treatment for large intestine cancer even though it does not cause any response in many patients. The present study aimed to investigate the effects of the TTN antisense RNA 1 (TTN-AS1) long noncoding RNA (lncRNA) on radiotherapy dynamics of large intestine cancer cells and to explore the underlying molecular mechanisms. TTN-AS1 expression was evaluated by reverse-transcription quantitative polymerase chain reaction, western blot, and cellular immunofluorescence, and flow cytometry analysis was used to measure apoptosis. Radiotherapy was simulated in vitro by exposing cancer cells to X-ray. TTN-AS1 was highly expressed in large intestine cancer cells after an X-ray exposition for 24 hr. TTN-AS1 knockdown improved the radiosensitivity of large intestine cancer cells and promoted apoptosis by increasing Bax/Bcl2 protein expression and the active-caspase 3/caspase 3 ratios following X-ray treatment. In addition, TTN-AS1 negatively regulated miR-134-5p expression, and miR-134-5p-mimic transfection decreased PAK3 protein expression in large intestine cancer cells. Importantly, TTN-AS1 promoted PAK3 and P21 protein expression in HT29 cells after X-ray treatment. Moreover, the knockdown of P21 protein expression improved radiosensitivity and promoted X-ray-induced apoptosis of HT29 cells. Finally, PAK3 knockdown expression decreased the p-AKT/AKT and p-GSK-3ß/GSK-3ß ratios and promoted the ß-catenin transfer from the nucleus to the cytoplasm. These data suggest that the TTN-AS1 lncRNA promoted resistance to radiotherapy of large intestine cancer cells by increasing PAK3 expression via miR-134-5p inhibition, and this may be related to the P21 and AKT/GSK-3ß/ß-catenin pathway.


Assuntos
Neoplasias Intestinais/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Conectina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Neoplasias Intestinais/metabolismo , Intestino Grosso/metabolismo , Intestino Grosso/patologia , MicroRNAs/metabolismo , Oligorribonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Tolerância a Radiação/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , beta Catenina/metabolismo , Quinases Ativadas por p21/genética
4.
Sci Rep ; 10(1): 343, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31941923

RESUMO

During intercellular communication, cells release extracellular vesicles such as exosomes, which contain proteins, ncRNAs and mRNAs that can influence proliferation and/or trigger apoptosis in recipient cells, and have been proposed to play an essential role in promoting invasion of tumor cells and in the preparation of metastatic niches. Our group proposed the antisense non-coding mitochondrial RNA (ASncmtRNA) as a new target for cancer therapy. ASncmtRNA knockdown using an antisense oligonucleotide (ASO-1537S) causes massive death of tumor cells but not normal cells and strongly reduces metastasis in mice. In this work, we report that exosomes derived from ASO-1537S-treated MDA-MB-231 breast cancer cells (Exo-1537S) inhibits tumorigenesis of recipient cells, in contrast to exosomes derived from control-ASO-treated cells (Exo-C) which, in contrast, enhance these properties. Furthermore, an in vivo murine peritoneal carcinomatosis model showed that Exo-1537S injection reduced tumorigenicity compared to controls. Proteomic analysis revealed the presence of Lactadherin and VE-Cadherin in exosomes derived from untreated cells (Exo-WT) and Exo-C but not in Exo-1537S, and the latter displayed enrichment of proteasomal subunits. These results suggest a role for these proteins in modulation of tumorigenic properties of exosome-recipient cells. Our results shed light on the mechanisms through which ASncmtRNA knockdown affects the preparation of breast cancer metastatic niches in a peritoneal carcinomatosis model.


Assuntos
Exossomos/metabolismo , Mitocôndrias/genética , RNA não Traduzido/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Superfície/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Proteínas do Leite/metabolismo , Oligorribonucleotídeos Antissenso/metabolismo , Oligorribonucleotídeos Antissenso/farmacologia , RNA não Traduzido/antagonistas & inibidores , RNA não Traduzido/genética , Transplante Heterólogo
5.
PLoS One ; 14(11): e0225468, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31756204

RESUMO

RATIONALE: Endogenous tissue mediators inducing lung inflammation in the context of ventilator-induced lung injury (VILI) and acute respiratory distress syndrome (ARDS) are ill-defined. OBJECTIVES: To test whether mitochondrial alarmins are released during VILI, and are associated with lung inflammation. METHODS: Release of mitochondrial DNA, adenosine triphosphate (ATP), and formyl-Met-Leu-Phe (fMLP) peptide-dependent neutrophil chemotaxis were measured in conditioned supernatants from human alveolar type II-like (A549) epithelial cells submitted to cyclic stretch in vitro. Similar measurements were performed in bronchoalveolar lavage fluids from rabbits submitted to an injurious ventilatory regimen, and from patients with ARDS. MEASUREMENTS AND MAIN RESULTS: Mitochondrial DNA was released by A549 cells during cell stretching, and was found elevated in BAL fluids from rabbits during VILI, and from ARDS patients. Cyclic stretch-induced interleukin-8 (IL-8) of A549 cells could be inhibited by Toll-like receptor 9 (TLR9) blockade. ATP concentrations were increased in conditioned supernatants from A549 cells, and in rabbit BAL fluids during VILI. Neutrophil chemotaxis induced by A549 cells conditioned supernatants was essentially dependent on fMLP rather than IL-8. A synergy between cyclic stretch-induced alarmins and lipopolysaccharide (LPS) was found in monocyte-derived macrophages in the production of IL-1ß. CONCLUSIONS: Mitochondrial alarmins are released during cyclic stretch of human epithelial cells, as well as in BAL fluids from rabbits ventilated with an injurious ventilatory regimen, and found in BAL fluids from ARDS patients, particularly in those with high alveolar inflammation. These alarmins are likely to represent the proximal endogenous mediators of VILI and ARDS, released by injured pulmonary cells.


Assuntos
Alarminas/metabolismo , Mitocôndrias/metabolismo , Síndrome do Desconforto Respiratório/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Células A549 , Animais , Líquido da Lavagem Broncoalveolar/química , Meios de Cultivo Condicionados/farmacologia , DNA Mitocondrial/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Infiltração de Neutrófilos/efeitos dos fármacos , Oligorribonucleotídeos Antissenso/metabolismo , Coelhos , Síndrome do Desconforto Respiratório/metabolismo , Estresse Fisiológico , Receptor Toll-Like 9/antagonistas & inibidores , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo
6.
J Orthop Surg Res ; 14(1): 305, 2019 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-31492154

RESUMO

BACKGROUND: Staphylococcus aureus (S. aureus) has the potential to opportunistically cause infectious diseases, including osteomyelitis, skin infections, pneumonia, and diarrhea. We previously reported that ASyycG RNA reduced the transcripts of virulent genes, and biofilm formation of S. aureus. Currently, graphene oxide (GO) nanosheets are used to efficiently deliver nucleic acids with favorable biocompatibility. METHODS: In the current study, a GO-based recombinant pDL278 ASyycG vector transformation strategy was developed. The particle size distributions and zeta-potential of the GO-PEI-based ASyycG were evaluated. The ASyycG plasmids were labeled with gene-encoding enhanced green fluorescent protein (ASyycG-eGFP). Quantitative real-time PCR assays were performed to investigate the expression of yycF/G/H and icaADB genes. Biofilm biomass and bacterial viability of S. aureus were evaluated by scanning electron microscopy and confocal laser scanning microscopy. We found that the expression of the yycG gene was inversely correlated with levels of the ASyycG transcripts and that the GO-PEI-ASyycG strain had the lowest expression of biofilm organization-associated genes. RESULTS: The results showed that the GO-based strategy significantly increased ASyycG transformation as a delivery system compared to the conventional competence-stimulating peptide strategy. Furthermore, GO-PEI-ASyycG suppressed bacterial biofilm aggregation and improved bactericidal effects on S. aureus after 24 h biofilm establishment. CONCLUSIONS: Our findings demonstrated that nano-GO with antisense yycG RNA is a more effective and relatively stable strategy for the management of S. aureus infections.


Assuntos
Antibacterianos/farmacologia , Grafite/farmacologia , RNA Antissenso/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Humanos , Testes de Sensibilidade Microbiana/métodos , Nanoestruturas , Oligorribonucleotídeos Antissenso/farmacologia , Tamanho da Partícula , Staphylococcus aureus/genética , Staphylococcus aureus/fisiologia
7.
Epigenetics Chromatin ; 12(1): 35, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-31196171

RESUMO

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) is among the most common malignant tumors with poor prognosis. Accumulating evidences have identified the important roles of long noncoding RNAs (lncRNAs) in the initiation and progression of various cancer types; however, the global lncRNAs expression profile for metastatic LSCC is limited. RESULTS: In the present study, we screen expression profiles of lncRNAs in advanced LSCC patients with paired tumor tissues and corresponding normal tissues by microarrays. We identify numerous differentially expressed transcripts, and after the necessary verification of the transcripts expression in expanded samples, we experimentally validate the expression patterns of the remarkable low expressed gene, SSTR5, and its antisense lncRNA, SSTR5-AS1. Downregulation of SSTR5 is detected in LSCC tissues and laryngeal carcinoma cells. Aberrant DNA hypermethylation of the CpG sites clustered in the exon 1 and accumulation of inactive histone modifications at SSTR5 promoter region may be epigenetic mechanisms for its inactivation in LSCC. SSTR5-AS1 may play antitumor role in LSCC and may be regulated by the hypermethylation of the same CpG sites with SSTR5. SSTR5-AS1 inhibits laryngeal carcinoma cells proliferation, migration, and invasion. SSTR5-AS1 increases the enrichment of MLL3 and H3K4me3 at the promoter region of SSTR5 by interacting with MLL3 and further induces the transcription of SSTR5. Furthermore, SSTR5-AS1 interacts with and recruits TET1 to its target gene E-cadherin to activate its expression. CONCLUSION: These findings suggest that the identified lncRNAs and mRNAs may be potential biomarkers in metastatic LSCC, and SSTR5-AS1 may act as a tumor suppressor as well as a potential biomarker for antitumor therapy.


Assuntos
Metilação de DNA , Neoplasias Laríngeas/genética , Oligorribonucleotídeos Antissenso/genética , RNA Longo não Codificante/genética , Receptores de Somatostatina/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Idoso , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Regulação para Baixo , Feminino , Humanos , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Masculino , Pessoa de Meia-Idade , Oxigenases de Função Mista/metabolismo , Metástase Neoplásica , Oligorribonucleotídeos Antissenso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , RNA Longo não Codificante/metabolismo , Receptores de Somatostatina/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
8.
Neuron ; 102(2): 294-320, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30998900

RESUMO

RNA binding proteins are critical to the maintenance of the transcriptome via controlled regulation of RNA processing and transport. Alterations of these proteins impact multiple steps of the RNA life cycle resulting in various molecular phenotypes such as aberrant RNA splicing, transport, and stability. Disruption of RNA binding proteins and widespread RNA processing defects are increasingly recognized as critical determinants of neurological diseases. Here, we describe distinct mechanisms by which the homeostasis of RNA binding proteins is compromised in neurological disorders through their reduced expression level, increased propensity to aggregate or sequestration by abnormal RNAs. These mechanisms all converge toward altered neuronal function highlighting the susceptibility of neurons to deleterious changes in RNA expression and the central role of RNA binding proteins in preserving neuronal integrity. Emerging therapeutic approaches to mitigate or reverse alterations of RNA binding proteins in neurological diseases are discussed.


Assuntos
Doenças do Sistema Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Animais , Autofagia , Sistemas CRISPR-Cas , Terapia Genética , Vetores Genéticos , Homeostase , Humanos , Terapia de Alvo Molecular , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/terapia , Oligorribonucleotídeos Antissenso/uso terapêutico , Síndromes Paraneoplásicas do Sistema Nervoso/genética , Síndromes Paraneoplásicas do Sistema Nervoso/metabolismo , Síndromes Paraneoplásicas do Sistema Nervoso/terapia , Processamento Pós-Transcricional do RNA , Splicing de RNA , Estabilidade de RNA , Transporte de RNA
9.
Cancer Lett ; 450: 63-75, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30771425

RESUMO

LncRNAs have been proven to play crucial roles in various processes of breast cancer. LncRNA FGF13-AS1 has been identified as one of the 25 downregulated lncRNAs in breast cancer through analyzing data from two cohorts and TCGA by another group of our lab. In this study, we report that FGF13-AS1 expression is decreased in breast cancer tissue compared with corresponding normal tissue, and the downregulation of FGF13-AS1 is associated with poor prognosis. Functional studies show that FGF13-AS1 inhibits breast cancer cells proliferation, migration, and invasion by impairing glycolysis and stemness properties. Mechanistically, FGF13-AS1 reduces the half-life of c-Myc (Myc) mRNA by binding RNA-binding proteins, insulin-like growth factor 2 mRNA binding proteins (IGF2BPs) and disrupting the interaction between IGF2BPs and Myc mRNA. Furthermore, Myc transcriptionally inhibits FGF13-AS1, forming a feedback loop in this signaling pathway. These results reveal for the first time that FGF13-AS1 functions as a tumor suppressor by inhibiting glycolysis and stemness properties of breast cancer cells, and the FGF13-AS1/IGF2BPs/Myc feedback loop could be a novel therapeutic target for breast cancer patients.


Assuntos
Neoplasias da Mama/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Retroalimentação Fisiológica , Feminino , Fatores de Crescimento de Fibroblastos , Glicólise , Xenoenxertos , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/patologia , Oligorribonucleotídeos Antissenso , Proteínas Proto-Oncogênicas c-myc/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais
10.
Methods ; 156: 16-24, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30502398

RESUMO

Adenosine Deaminases that Act on RNA (ADARs) are a group of enzymes that catalyze the conversion of adenosines (A's) to inosines (I's) in a process known as RNA editing. Though ADARs can act on different types of RNA, editing events in coding regions of mRNA are of particular interest as I's base pair like guanosines (G's). Thus, every A-to-I change catalyzed by ADAR is read as an A-to-G change during translation, potentially altering protein sequence and function. This ability to re-code makes ADAR an attractive therapeutic tool to correct genetic mutations within mRNA. The main challenge in doing so is to re-direct ADAR's catalytic activity towards A's that are not naturally edited, a process termed Site-Directed RNA Editing (SDRE). Recently, a handful of labs have taken up this challenge and two basic strategies have emerged. The first involves redirecting endogenous ADAR to new sites by making editable structures using antisense RNA oligonucleotides. The second also utilizes antisense RNA oligonucleotides, but it uses them as guides to deliver the catalytic domain of engineered ADARs to new sites, much as CRISPR guides deliver Cas nucleases. In fact, despite the intense current focus on CRISPR-Cas9 genome editing, SDRE offers a number of distinct advantages. In the present review we will discuss these strategies in greater detail, focusing on the concepts on which they are based, how they were developed and tested, and their respective advantages and disadvantages. Though the precise and efficient re-direction of ADAR activity still remains a challenge, the systems that are being developed lay the foundation for SDRE as a powerful tool for transient genome editing.


Assuntos
Adenosina Desaminase/genética , Sistemas CRISPR-Cas , Mutagênese Sítio-Dirigida/métodos , Edição de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Adenosina/metabolismo , Adenosina Desaminase/metabolismo , Animais , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Genoma Humano , Humanos , Inosina/metabolismo , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , Domínios Proteicos , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo
11.
Biochem Biophys Res Commun ; 509(2): 441-447, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30594388

RESUMO

Long non-coding RNAs (lncRNAs) are extensively involved in multiple malignancies including colorectal cancer (CRC). In the present study, we found a novel lncRNA, long intergenic non-protein coding RNA 483 (LINC00483), which was upregulated in CRC. We also illustrated that upregulated LINC00483 was correlated with poor clinicopathological features of patients with CRC. Functionally, we displayed that a knockdown of LINC00483 suppressed LOVO and HT29 cells proliferation and metastatic ability. We further illustrated that miR-204-3p was involved in LINC00483 induced proliferation and metastasis. An overexpression of miR-204-3p could attenuate the facilitative effect which LINC00483 presented. Through a luciferase assay, we showed the direct binding effect between LINC00483 and miR-204-3p. Even further, we revealed that LINC00483 and formin like 2 (FMNL2) shared a similar miR-204-3p response elements (MREs-204-3p). FMNL2 was a direct target of miR-204-3p. FMNL2 was a downstream gene of LINC00483 and participated in LINC00483 mediated proliferation and metastasis. Lastly, we proved that LINC00483 promoted proliferation and metastasis via modulating of FMNL2 in LOVO and HT29 cells. In summary, the outcomes of this study illustrated that LINC00483 promoted CRC cells proliferation and metastasis via modulating of FMNL2 by acting as a ceRNA of miR-204-3p. LINC00483/miR-204-3p/FMNL2 axial might be a novel target in molecular treatment of CRC.


Assuntos
Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , MicroRNAs/genética , Proteínas/genética , RNA Longo não Codificante/genética , Adulto , Idoso , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Feminino , Forminas , Genes Reporter , Células HT29 , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Luciferases/genética , Luciferases/metabolismo , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , Prognóstico , Proteínas/metabolismo , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Análise de Sobrevida
12.
Biochem Biophys Res Commun ; 503(4): 2603-2610, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30139516

RESUMO

Circular RNAs (circRNAs) are important non-coding RNAs that are reportedly involved in the progression of diverse human cancers through their action as a microRNA (miRNA) sponge. However, the exact roles of circRNAs in breast cancer (BC) remain largely unknown. The present data demonstrate the significantly upregulated expression of hsa_circ_0007534 circRNA in BC tissues and cell lines. In contrast, miR-593 expression was significantly downregulated. Knockdown of hsa_circ_0007534 inhibited BC cell proliferation, colony formation, and invasion, and promoted apoptosis in BC cells. Moreover, hsa_circ_0007534 was demonstrated to be a sponge of miR-593, and expression of miR-593 in BC cells was negatively correlated with hsa_circ_0007534. MUC19 expression was markedly increased in BC tissues and cell lines, and the 3'-UTR of MUC19 was targeted by miR-593. The expression of MUC19 was negatively regulated by miR-593 in BC cells. Our findings suggest an oncogenic role for hsa_circ_0007534 in BC by acting as a miR-593 sponge to promote MUC19 expression.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Mucinas/genética , RNA Neoplásico/genética , RNA/genética , Regiões 3' não Traduzidas , Apoptose , Sequência de Bases , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , MicroRNAs/metabolismo , Mucinas/metabolismo , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , RNA/metabolismo , RNA Circular , RNA Neoplásico/metabolismo , Transdução de Sinais , Análise de Sobrevida
13.
Biochem Biophys Res Commun ; 503(4): 2659-2665, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30086881

RESUMO

Emerging evidence indicated that abnormally expressed circular RNAs (circRNAs) are critically involved in tumorigenesis and development of several cancers. However, the study relevant to the relationship between circRNAs and hepatocellular carcinoma (HCC) is rare. In this study, the expression of circ_001569 in HCC tissue specimens and cells were determined by qRT-PCR. The clinical relevance of circ_001569 was also analyzed. In addition, loss-of-function and gain-of-function assays were conducted to explore the biological functions of circ_001569. Furthermore, tumor formation and lung metastasis studies were induced to confirm the in vitro data. Mechanistically, bioinformatics analysis and luciferase reporter assays were conducted to illustrate the underlying mechanism of circ_001569. The results indicated that circ_001569 was overexpressed in HCC tissue samples and cells. This overexpression is correlated with larger tumor size, advanced TNM stages and unfavorable prognosis in the patients with HCC. Additionally, circ_001569 significantly facilitated HCC cell growth, migration and invasion. The animal studies further confirmed thein vitroresults. More importantly, circ_001569 could directly sponge miR-411-5p and miR-432-5p. The oncogenic functions of circ_001569 is partially dependent on its regulation on miR-411-5p and miR-432-5p. In summary, circ_001569/miR-411-5p/miR-432-5p regulatory signaling might be a rational HCC-related therapeutic target.


Assuntos
Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , RNA/genética , Idoso , Animais , Sequência de Bases , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/secundário , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/secundário , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , Prognóstico , RNA/antagonistas & inibidores , RNA/metabolismo , RNA Circular , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Commun ; 9(1): 2713, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30006596

RESUMO

Liposomes have been well established as an effective drug delivery system, due to simplicity of their preparation and unique characteristics. However conventional liposomes are unsuitable for the on-demand content release, which limits their therapeutic utility. Here we report X-ray-triggerable liposomes incorporating gold nanoparticles and photosensitizer verteporfin. The 6 MeV X-ray radiation induces verteporfin to produce singlet oxygen, which destabilises the liposomal membrane and causes the release of cargos from the liposomal cavity. This triggering strategy is demonstrated by the efficiency of gene silencing in vitro and increased effectiveness of chemotherapy in vivo. Our work indicates the feasibility of a combinatorial treatment and possible synergistic effects in the course of standard radiotherapy combined with chemotherapy delivered via X-ray-triggered liposomes. Importantly, our X-ray-mediated liposome release strategy offers prospects for deep tissue photodynamic therapy, by removing its depth limitation.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Terapia Combinada/métodos , Preparações de Ação Retardada , Lipossomos , Nanopartículas Metálicas/efeitos da radiação , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Etoposídeo/farmacocinética , Etoposídeo/farmacologia , Fluoresceínas/química , Fluoresceínas/metabolismo , Ácido Fólico/química , Inativação Gênica , Ouro/química , Células HCT116 , Humanos , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , Células PC12 , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/antagonistas & inibidores , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Oxigênio Singlete/metabolismo , Verteporfina/farmacologia , Raios X , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biochem Biophys Res Commun ; 499(1): 66-70, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29555473

RESUMO

Non-alcoholic fatty liver disease and steatohepatitis (NAFLD and NASH) account for the majority of liver disease in industrialized countries. However, the pathogenesis still unclear. Long non-coding RNAs (lncRNAs) has been reported to be involved in various pathophysiological processes. Here, we reported a novel role of lncARSR in hepatic lipogenesis in NAFLD. The expression of lncARSR was induced both in NAFLD patients and mouse model, as well as in hepatocytes treated with fatty acid. Moreover, overexpression of lncARSR enhanced while knockdown of lncARSR ameliorated hepatic lipid accumulation in vivo and in vitro. Furthermore, the expression of genes related to fatty acid synthesis and oxidation increased with lncARSR overexpression in vivo. Mechanistically, we identified that lncARSR regulated hepatic lipogenesis via upregulating SREBP-1c, the key regulatory molecule involved in lipogenesis. Knockdown of SREBP-1c by shRNA blocked the effect of lncARSR on lipogenesis. Furthermore, we demonstrated that lncARSR regulated SREBP-1c expression by PI3K/Akt pathway. In conclusion, our data indicated that lncARSR potentially contributes to the hepatic steatosis in NAFLD, which may be a new therapeutic target against NAFLD.


Assuntos
Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Lipogênese/genética , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
16.
Neuropharmacology ; 135: 11-21, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29510185

RESUMO

It is well known that Wnt5a activation plays a pivotal role in brain injury and ß-arrestin2 induces c-Jun N-terminal kinase (JNK3) activation is involved in neuronal cell death. Nonetheless, the relationship between Wnt5a and JNK3 remains unexplored during cerebral ischemia/reperfusion (I/R). In the present study, we tested the hypothesis that Wnt5a-mediated JNK3 activation via the Wnt5a-Dvl-1-ß-arrestin2-JNK3 signaling pathway was correlated with I/R brain injury. We found that cerebral I/R could enhance the assembly of the Dvl-1-ß-arrestin2-JNK3 signaling module, Dvl-1 phosphorylation and JNK3 activation. Activated JNK3 could phosphorylate the transcription factor c-Jun, prompt caspase-3 activation and ultimately lead to neuronal cell death. To further explore specifically Wnt5a mediated JNK3 pathway activation in neuronal injury, we used Foxy-5 (a peptide that mimics the effects of Wnt5a) and Box5 (a Wnt5a antagonist) both in vitro and in vivo. AS-ß-arrestin2 (an antisense oligonucleotide against ß-arrestin2) and RRSLHL (a small peptide that competes with ß-arrestin2 for binding to JNK3) were applied to confirm the positive signal transduction effect of the Dvl-1-ß-arrestin2-JNK3 signaling module during cerebral I/R. Furthermore, Box5 and the RRSLHL peptide were found to play protective roles in neuronal death both in vivo global and focal cerebral I/R rat models and in vitro oxygen glucose deprivation (OGD) neural cells. In summary, our results indicate that Wnt5a-mediated JNK3 activation participates in I/R brain injury by targeting the Dvl-1-ß-arrestin2/JNK3 interaction. Our results also point to the possibility that disrupting Wnt5a-JNK3 signaling pathway may provide a new approach for stroke therapy.


Assuntos
Região CA1 Hipocampal/metabolismo , Proteínas Desgrenhadas/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Neuroproteção , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Wnt-5a/metabolismo , beta-Arrestina 2/metabolismo , Animais , Região CA1 Hipocampal/citologia , Morte Celular/efeitos dos fármacos , Masculino , Neuroproteção/efeitos dos fármacos , Oligopeptídeos/farmacologia , Oligorribonucleotídeos Antissenso/farmacologia , Peptídeos/farmacologia , Fosforilação , Ratos , Traumatismo por Reperfusão/patologia , Proteína Wnt-5a/agonistas , beta-Arrestina 2/antagonistas & inibidores
17.
Gene ; 652: 48-58, 2018 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-29427737

RESUMO

Nasopharyngeal carcinoma (NPC) is a head and neck cancer associated with poor prognosis. Many studies have shown that the epithelial-to-mesenchymal transition (EMT) is important in cancer progression, metastasis, and chemotherapy resistance and that microRNAs (miRNAs) play a key role in chemotherapy resistance associated with EMT. The miRNA miR-139-5p is downregulated in many human cancers and is closely related to tumor progression. The aim of this study was to investigate the ability of miR-139-5p to influence the cisplatin resistance, apoptosis, invasion and migration in NPC cells through the regulation of the EMT. We investigated these processes in parental HNE1 and cisplatin-resistant HNE1/DDP cells transfected with miR-139-5p inhibitors and mimics, respectively. Our results suggest that the upregulation of miR-139-5p expression inhibits proliferation, invasion, migration and EMT in human NPC cells. In addition, we found that miR-139-5p expression levels and DDP-induced apoptosis positively correlate in NPC cells. In conclusion, our results demonstrate that miR-139-5p can regulate the migration, invasion and DDP resistance in human NPC by modulating the EMT. The regulation of miR-139-5p levels might be a new approach to reverse EMT and DDP resistance and counteract metastasis and chemotherapy resistance in human NPC.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Linhagem Celular Tumoral , Movimento Celular , Resistencia a Medicamentos Antineoplásicos/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Mimetismo Molecular , Nasofaringe/efeitos dos fármacos , Nasofaringe/metabolismo , Nasofaringe/patologia , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/metabolismo
20.
Horm Cancer ; 8(4): 219-229, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28577281

RESUMO

While selective estrogen receptor modulators, such as tamoxifen, have contributed to increased survival in patients with hormone receptor-positive breast cancer, the development of resistance to these therapies has led to the need to investigate other targetable pathways involved in oncogenic signaling. Approval of the mTOR inhibitor everolimus in the therapy of secondary endocrine resistance demonstrates the validity of this approach. Importantly, mTOR activation regulates eukaryotic messenger RNA translation. Eukaryotic translation initiation factor 4E (eIF4E), a component of the cap-dependent translation complex eIF4F, confers resistance to drug-induced apoptosis when overexpressed in multiple cell types. The eIF4F complex is downstream of multiple oncogenic pathways, including mTOR, making it an appealing drug target. Here, we show that the eIF4F translation pathway was hyperactive in tamoxifen-resistant (TamR) MCF-7L breast cancer cells. While overexpression of eIF4E was not sufficient to confer resistance to tamoxifen in MCF-7L cells, its function was necessary to maintain resistance in TamR cells. Targeting the eIF4E subunit of the eIF4F complex through its degradation using an antisense oligonucleotide (ASO) or via sequestration using a mutant 4E-BP1 inhibited the proliferation and colony formation of TamR cells and partially restored sensitivity to tamoxifen. Further, the use of these agents also resulted in cell cycle arrest and induction of apoptosis in TamR cells. Finally, the use of a pharmacologic agent which inhibited the eIF4E-eIF4G interaction also decreased the proliferation and anchorage-dependent colony formation in TamR cells. These results highlight the eIF4F complex as a promising target for patients with acquired resistance to tamoxifen and, potentially, other endocrine therapies.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Fator de Iniciação 4F em Eucariotos/metabolismo , Biossíntese de Proteínas , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Fator de Iniciação 4F em Eucariotos/genética , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Células MCF-7 , Oligorribonucleotídeos Antissenso/genética , Fosforilação , Polirribossomos , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA